1
|
Prieto-Dominguez N, Goel P, Ojo OA, Moretto K, Holtzhausen A, Humphryes A, Zhou X, Kuznetsova V, Dempsey F, Pittman K, Lu R, Green TJ, Shi LZ, Welner RS, Earp HS, Ubil E. PTP Inhibition Improves the Macrophage Antitumor Immune Response and the Efficacy of Chemo- and Radiotherapy. Cancer Immunol Res 2025; 13:749-766. [PMID: 39937118 PMCID: PMC12048275 DOI: 10.1158/2326-6066.cir-24-0335] [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: 04/12/2024] [Revised: 10/15/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Traditional anticancer therapies induce tumor cell death and subsequent release of damage-associated molecular patterns (DAMPs) that activate the innate inflammatory response. Paradoxically, after treatment, macrophages often adopt a pro-wound healing, rather than proinflammatory, phenotype and contribute to cancer progression. We found that in areas proximal to DAMP release, tumor cells upregulate the expression of Pros1. Tumor-secreted Pros1 binds to the macrophage Mer receptor, consequently limiting responsiveness to DAMPs by preventing Toll-like receptor signal transduction. Pharmacological inhibition of PTP1b signaling downstream of Mer rescued the proinflammatory response, even in the presence of Pros1. Combining protein tyrosine phosphatase (PTP) inhibition with traditional therapeutics, such as chemo- or radiotherapy, rescued the innate immune response to DAMPs, increased immune infiltration, and resulted in a 40% to 90% reduction in tumor growth in multiple treatment-refractory preclinical models. Our findings suggest using PTP1b inhibitors may be a tumor agnostic means of improving the efficacy of some of the most widely used anticancer therapeutic agents.
Collapse
Affiliation(s)
| | - Paran Goel
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Oluwagbemiga A. Ojo
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Katarina Moretto
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alisha Holtzhausen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Departments of Medicine and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Angel Humphryes
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xinyue Zhou
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Valeriya Kuznetsova
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Francesca Dempsey
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kelly Pittman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rui Lu
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Todd J. Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lewis Z. Shi
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Robert S. Welner
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Departments of Medicine and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eric Ubil
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
2
|
Zhao X, Chen C, Qiu H, Liu J, Shao N, Guo M, Jiang Y, Zhao J, Xu L. The landscape of ATF3 in tumors: Metabolism, expression regulation, therapy approach, and open concerns. Pharmacol Res 2025; 214:107666. [PMID: 39978658 DOI: 10.1016/j.phrs.2025.107666] [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: 09/30/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Cellular stress response is a pivotal process in tumor development and therapy. Activating transcription factor 3 (ATF3), a representative stress-responsive protein, plays pleiotropic roles in various biological processes. Over the past decade, studies have described not only the general role of ATF3 in tumor metabolism but also the complexity of ATF3 expression regulation and its associated modifications, including phosphorylation, ubiquitination, SUMOylation, and NEDDylation. Interestingly, beyond being a transcription factor, ATF3 can act as a modifier to control the ubiquitination of target molecules, such as p53, to exert its function in tumors. These advances in uncovering ATF3 biological function have yielded new insights into the cellular stress response during tumor development and will be instrumental in developing novel interventions. In this review, we update the role of ATF3 as a nexus in amino acid metabolism, lipid metabolism, glycometabolism, and other metabolic pathways in tumors; delineate the underlying mechanisms involving DNA level regulation, epigenetic regulation, and post-translational modifications of ATF3; and summarize the progression of tumor mono/combination therapies related to ATF3. In particular, we discuss the challenges that need to be addressed to provide a new conceptual framework for further understanding the potential therapeutic value of ATF3 in ongoing clinical trials.
Collapse
Affiliation(s)
- Xu Zhao
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Chao Chen
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Hui Qiu
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Jing Liu
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Nan Shao
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Mengmeng Guo
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China
| | - Yuanye Jiang
- Department of Gastroenterology, Putuo hospital, Shanghai University of Tradtional Chinese Medicine, Shanghai 200062, China.
| | - Juanjuan Zhao
- Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China.
| | - Lin Xu
- Medical College, Guizhou University, Guiyang, Guizhou Province 550025, China; Key Laboratory for Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Guizhou 563000, China; Innovation Center for Tissue Damage Repair, Ministry of Education, Zunyi, Guizhou 563000, China.
| |
Collapse
|
3
|
Li X, Zhang X, Liu T, Zhang G, Chen D, Lin S. Identification of immune characteristic biomarkers and therapeutic targets in cuproptosis for rheumatoid arthritis by integrated bioinformatics analysis and single-cell RNA sequencing analysis. Front Med (Lausanne) 2025; 12:1520400. [PMID: 40166070 PMCID: PMC11955502 DOI: 10.3389/fmed.2025.1520400] [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: 11/01/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic autoimmune disorder intricately liked with inflammation. Cuproptosis, an emerging type of cell death, has been implicated in the initiation and development of RA. However, the exact alterations in the expression and biological function of cuproptosis-related genes (CRGs) in RA remain poorly understood. Therefore, our study aims to elucidate the potential association between CRGs and RA, with the goal of identifying novel biomarkers for the treatment and prognosis of RA. Methods In this study, we identified ten differentially expressed cuproptosis-related genes (DE-CRGs) between patients with RA and controls. Through comprehensive functional enrichment and protein-protein interaction (PPI) network analysis, we explored the functional roles of the DE-CRGs. Additionally, we investigated the correlation between DE-CRGs and immune infiltration, immune factors, diagnostic efficacy, and potential therapeutic drugs. Results Leveraging single-cell RNA sequencing data, we conducted a detailed analysis to elucidate alterations in various cell clusters associated with RA. Our study unveiled a significant association between DE-CRGs and diverse biological functions, as well as potential drug candidates. Discussion These findings provide crucial insights into the involvement of DE-CRGs in the pathogenesis of RA and shed light on potential therapeutic strategies.
Collapse
Affiliation(s)
- Xianbin Li
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou, China
- Jiujiang Key Laboratory of Digital Technology, Jiujiang, China
| | - Xueli Zhang
- Department of Medical Technology, Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Tao Liu
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
| | - Guodao Zhang
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou, China
| | - Dan Chen
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Suxian Lin
- Department of Rheumatology, Wenzhou People’s Hospital, Wenzhou, China
| |
Collapse
|
4
|
Li R, Ma Y, He A, Pu Y, Wan X, Sun H, Wang N, Luo M, Wang G, Xia Y. Fasting enhances the efficacy of Sorafenib in breast cancer via mitophagy mediated ROS-driven p53 pathway. Free Radic Biol Med 2025; 229:350-363. [PMID: 39864757 DOI: 10.1016/j.freeradbiomed.2025.01.047] [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: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
The multi-kinase inhibitor sorafenib has shown potential to inhibit tumor cell growth and intra-tumoral angiogenesis by targeting several kinases, including VEGFR2 and RAF. Abnormal activation of the Ras/Raf/MAPK/ERK kinase cascade and the VEGF pathway is a common feature in breast cancer. However, the efficacy of sorafenib in breast cancer treatment remains limited. Recently, fasting has emerged as a promising non-pharmacological approach to modulate cancer metabolism and enhance the effectiveness of cancer therapies. In this study, we found that fasting significantly enhances the anti-cancer effects of sorafenib monotherapy and its combination with immunotherapy in breast cancer models without causing obvious side effects. This combined treatment effectively inhibits tumor cell proliferation and intra-tumoral angiogenesis. The fasting-induced reduction in peripheral blood glucose levels strongly correlated with enhanced sensitivity to sorafenib. Mechanistically, the combined treatment induced mitophagy, characterized by mitochondrial dysfunction and activation of the PINK1-Parkin pathway. Consequently, increased mitochondrial ROS levels promoted p53 expression, amplifying cell cycle arrest and apoptosis in breast cancer cells. Furthermore, fasting reduced lactate levels within the tumor, and the consequent glucose limitation synergized with sorafenib to activate AMPK, which in turn elevated PD-L1 expression in tumor cells, potentially enhancing their sensitivity to immunotherapy. In summary, our findings demonstrate that fasting and sorafenib, as a rational combination therapy, induce mitophagy, thereby enhancing sorafenib's efficacy in treating breast cancer through the ROS-driven p53 pathway. This study underscores the potential of fasting in breast cancer therapy and provides a foundation for optimizing the clinical application of sorafenib.
Collapse
Affiliation(s)
- Ru Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China
| | - Yimei Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Anqi He
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yamin Pu
- Department of Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuanting Wan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Hongbao Sun
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Min Luo
- Department of Biotherapy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China.
| | - Yong Xia
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Rehabilitation Medicine Center, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu, 610041, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Li T, Chen G, Cao L, Rong W, Zhao H, Xiong Z, Liu Q, Song J, Wang W, Liu Y, Wang X, Liu S. Effects of combined exposure to 17α-methyltestosterone and polystyrene microplastics on lipid metabolism and the nervous system in Danio rerio. J Steroid Biochem Mol Biol 2025; 247:106665. [PMID: 39694074 DOI: 10.1016/j.jsbmb.2024.106665] [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: 09/18/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/20/2024]
Abstract
Polystyrene (PS) microplastics are pervasive environmental pollutants that are harmful to aquatic organisms upon degradation. The synthetic androgen 17α-methyltestosterone (MT) is an environmental endocrine-disrupting chemical. This study aimed to systematically evaluate the combined histological and molecular effects of MT and PS exposure on the liver and brain tissues of Danio rerio with focus on lipid metabolism and neural function disruption. Female D. rerio were exposed to 50 ng/L MT and 0.5 mg/L PS (5 μm in diameter) for 21 d. Histological observations, real-time quantitative PCR (qPCR), and RNA-sequencing (RNA-seq) analysis were employed to assess the effects of PS and MT. These results indicated that MT and PS co-exposure caused fatty degeneration of liver cells and a significant upregulation of lipid synthesis-related genes (ACSS1, CEL, FASN, and GK5). In brain tissue, the observed effects included reduced marginal layer neuron counts, cytoplasmic loosening of central layer neurons, disordered gray matter layer cells, and vascular congestion. RNA-seq analysis further revealed significant enrichment of differentially expressed genes in the "glycine, serine, and threonine metabolism" and "neuroactive ligand-receptor interaction" signaling pathways. Thus, MT and PS co-exposure induced lipid metabolism disorders in D. rerio and influence neural signaling by altering the "neuroactive ligand-receptor interaction" pathway. These findings highlight the complex risks posed by environmental pollutants to aquatic life and provide critical insights for environmental protection and aquatic health research.
Collapse
Affiliation(s)
- Tongyao Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Gen Chen
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Lu Cao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Weiya Rong
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Haiyan Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Zijun Xiong
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Qing Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Jing Song
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Weiwei Wang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Yu Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xianzong Wang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China.
| | - Shaozhen Liu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China.
| |
Collapse
|
6
|
Liu N, Yan M, Lu C, Tao Q, Wu J, Zhou Z, Chen J, Chen X, Peng C. Eravacycline improves the efficacy of anti-PD1 immunotherapy via AP1/CCL5 mediated M1 macrophage polarization in melanoma. Biomaterials 2025; 314:122815. [PMID: 39288620 DOI: 10.1016/j.biomaterials.2024.122815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024]
Abstract
Screening approved library is a promising and safe strategy to overcome the limitation of low response rate and drug resistance in immunotherapy. Accumulating evidence showed that the application of antibiotics has been considered to reduce the effectiveness of anti-PD1 immunotherapy in tumor treatment, however, in this study, an antibiotic drug (Eravacycline, ERV) was identified to improve the efficacy of anti-PD1 immunotherapy in melanoma through screening approved library. Administration of ERV significantly attenuated melanoma cells growth as well as directly or indirectly benefited M1 macrophage polarization. Meanwhile, ERV treatment significantly induced cellular autophagy via damage of mitochondria, leading to up-regulation of ROS production, subsequently, raised CCL5 secretion through elevation AP1 binding to CCL5 promoter via p38 or JNK1/2 activation. Knockdown of Ccl5 expression attenuated ERV triggered M1 macrophage polarization in melanoma cells. Clinical analysis revealed a positive association between high expression of CCL5 and improved prognosis as well as a favorable anti-PD1 therapy in melanoma patients. As expected, application of ERV improved the efficacy of anti-PD1. Overall, our results approved that ERV enhances the efficacy of anti-PD1 immunotherapy in melanoma by promoting the polarization of M1 macrophages, which provided novel therapeutic strategy for improving the effectiveness of melanoma anti-PD1 immunotherapy.
Collapse
Affiliation(s)
- Nian Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Mingjie Yan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Can Lu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Qian Tao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Jie Wu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Henan, 450052, China
| | - Jing Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China.
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China; Furong Laboratory, Central South University, Changsha, Hunan, 410000, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China.
| |
Collapse
|
7
|
Kiełbowski K, Bakinowska E, Becht R, Pawlik A. Metabolism of Tryptophan, Glutamine, and Asparagine in Cancer Immunotherapy-Synergism or Mechanism of Resistance? Metabolites 2025; 15:144. [PMID: 40137109 PMCID: PMC11944271 DOI: 10.3390/metabo15030144] [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: 01/19/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Amino acids are crucial components of proteins, key molecules in cellular physiology and homeostasis. However, they are also involved in a variety of other mechanisms, such as energy homeostasis, nitrogen exchange, further synthesis of bioactive compounds, production of nucleotides, or activation of signaling pathways. Moreover, amino acids and their metabolites have immunoregulatory properties, significantly affecting the behavior of immune cells. Immunotherapy is one of the oncological treatment methods that improves cytotoxic properties of one's own immune system. Thus, enzymes catalyzing amino acid metabolism, together with metabolites themselves, can affect immune antitumor properties and responses to immunotherapy. In this review, we will discuss the involvement of tryptophan, glutamine, and asparagine metabolism in the behavior of immune cells targeted by immunotherapy and summarize results of the most recent investigations on the impact of amino acid metabolites on immunotherapy.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Rafał Becht
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| |
Collapse
|
8
|
Zhang M, Song X, Qin Y, Peng Y, Zhang S, Feng W, Huang H, Chen Y, Zhou J. Single-atom-doped piezocatalyst induces copper-free cuproptosis in tumor therapy. SCIENCE ADVANCES 2025; 11:eadt8451. [PMID: 39951535 PMCID: PMC11827870 DOI: 10.1126/sciadv.adt8451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/14/2025] [Indexed: 02/16/2025]
Abstract
Cuproptosis, a distinct cell death pathway, has been integrated into nanomedicine for disease theranostics. However, current nanosystems inducing cuproptosis rely on exogenous toxic copper ions, limiting the scope of biomaterials. Developing nanoplatforms that induce cuproptosis without exogenous copper holds substantial promise. Here, we engineered a two-dimensional iron (Fe) single-atom-doped molybdenum disulfide (MoS2) piezocatalyst (Fe-MoS2) for tumor therapy. Incorporating single Fe atoms enhances MoS2 piezoelectric polarization via charge redistribution and modulates Fe and Mo oxidation states, enabling multifaceted enzymatic activities, including peroxidase-, glutathione oxidase-, oxidase-, and catalase-like activities. Upon ultrasound stimulation, the Fe-MoS2 nanocatalyst generates reactive oxygen species and depletes glutathione via synergistic piezocatalytic and enzyocatalytic effects, disrupting copper ion homeostasis and inducing cuproptosis, concurrently triggering ferroptosis and ferritinophagy, which collectively enhances tumor suppression. This study represents the first paradigm to introduce a copper-free piezocatalyst for initiating cuproptosis, substantially advancing the applications of cuproptosis in tumor therapy.
Collapse
Affiliation(s)
- Meixiang Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Xinran Song
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Qin
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yuanyuan Peng
- Anhui University of Science and Technology. School of Medicine, Huainan, Anhui 232000, P.R. China
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Wei Feng
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hui Huang
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- Materdicine Laboratory, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| | - Jianqiao Zhou
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| |
Collapse
|
9
|
Wu J, Liu N, Chen J, Tao Q, Lu C, Li Q, Chen X, Peng C. Clofarabine induces tumor cell apoptosis, GSDME-related pyroptosis, and CD8 + T-cell antitumor activity via the non-canonical P53/STING pathway. J Immunother Cancer 2025; 13:e010252. [PMID: 39915005 PMCID: PMC11804206 DOI: 10.1136/jitc-2024-010252] [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: 07/31/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Clofarabine (Clo) is a Food and Drug Administration (FDA)-approved drug for the treatment of acute lymphoblastic leukemia; however, its effects on solid tumors remain largely unknown. METHODS In vitro and in vivo experiments have demonstrated the cytotoxic effects of Clo on melanoma and lung cancer. The molecular mechanisms of Clo-induced tumor cell death were analyzed using western blotting, ELISA, reverse transcription-PCR, immunofluorescence, co-immunoprecipitation (CO-IP), short hairpin RNA, co-culture, chromatin immunoprecipitation, and flow cytometry. Clinical data sets were used to analyze the correlation between stimulator of interferon genes (STING)-NFκB signaling and immune infiltration. RESULTS In this study, Clo significantly reduced the growth of melanoma and lung cancer cells. Furthermore, Clo treatment induced GSDME-mediated pyroptosis. Most importantly, Clo administration dramatically increased the cytotoxic activity of CD8+ T cells in vitro and in vivo. Mechanistically, the administration of Clo induced the interaction of P53 with STING, which activated the non-canonical STING-NFκB pathway; consequently, NF-κB directly bound to the promoter regions of its target genes, including CCL5, CXCL10, HLAs and BAX. This resulted in apoptosis, pyroptosis, and immunogenic cell death in tumor cells by Clo. Furthermore, Clo-induced GSDME-mediated pyroptosis partly assists in activating T cell immunity via CCL5 and CXCL10. The non-canonical STING-NF-κB pathway is the crucial signaling pathway that initiates and links apoptosis, pyroptosis, and immunogenic cell death. CONCLUSIONS Our study is the first to show that Clo, an FDA-approved drug, induces tumor cell apoptosis, GSDME-related pyroptosis, and CD8+ T-cell antitumor activity via the non-canonical P53-STING-NF-κB signaling pathway, providing a novel strategy for the clinical therapy of melanoma and lung cancer.
Collapse
Affiliation(s)
- Jie Wu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nian Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Tao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Can Lu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiuqiu Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Human Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Furong Laboratory, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Niu L, Liu S, Shen J, Chang J, Li X, Zhang L. ATF3 regulates CDC42 transcription and influences cytoskeleton remodeling, thus inhibiting the proliferation, migration and invasion of malignant skin melanoma cells. Melanoma Res 2025; 35:37-49. [PMID: 39591541 DOI: 10.1097/cmr.0000000000001011] [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: 11/28/2024]
Abstract
Cutaneous malignant melanoma (CMM) is one of the most aggressive and lethal types of skin cancer. Cytoskeletal remodeling is a key factor in the progression of CMM. Previous research has shown that activating transcription factor 3 (ATF3) inhibits metastasis in bladder cancer by regulating actin cytoskeleton remodeling through gelsolin. However, whether ATF3 plays a similar role in cytoskeletal remodeling in CMM cells remains unknown. Various gene and protein expression analyses were performed using techniques such as reverse transcription quantitative PCR, western blot, immunofluorescent staining, and immunohistochemical staining. CMM viability, migration, and invasion were examined through cell counting kit-8 and transwell assays. The interactions between cell division cycle 42 (CDC42) and ATF3 were investigated using chromatin immunoprecipitation and dual-luciferase reporter assays. CDC42 was upregulated in CMM tissues and cells. Cytoskeletal remodeling of CMM cells, as well as CMM cell proliferation, migration, and invasion, were inhibited by CDC42 or ATF3. ATF3 targeted the CDC42 promoter region to regulate its transcriptional activity. ATF3 suppresses cytoskeletal remodeling in CMM cells, thereby inhibiting CMM progression and metastasis through CDC42. This research may provide a foundation for using ATF3 as a therapeutic target for CMM.
Collapse
Affiliation(s)
- Liang Niu
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University
| | - Shuo Liu
- Department of Stomatology, Handan First Hospital
| | - Jiuxiao Shen
- Medical Cosmetic Center, Affiliated Hospital of Hebei Engineering University, Handan City, Hebei Province, China
| | - Jin Chang
- Medical Cosmetic Center, Affiliated Hospital of Hebei Engineering University, Handan City, Hebei Province, China
| | - Xiaojing Li
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University
| | - Ling Zhang
- Department of Dermatology, Affiliated Hospital of Hebei Engineering University
| |
Collapse
|
11
|
Wang H, Zhang Y, Jiang Y, Xiang R, Gong H, Gong Y, Xu H, Ma Z, Xie Y, Zhu Y, Hu B, He X, Liu J, Zhang J, Xiao X. The function and mechanism of clinical trial agent CPI-613 in multiple myeloma. Biochem Pharmacol 2025; 232:116717. [PMID: 39675585 DOI: 10.1016/j.bcp.2024.116717] [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: 07/20/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Multiple myeloma (MM) is an incurable malignant hematological neoplasm characterized by clonal proliferation of plasma cells accumulating in the bone marrow. Currently, the treatment of MM is usually based on a multi-drug combination strategy, and the remission rates of MM patients have been greatly improved. However, MM is still not immune to drug resistance and recurrence and is an incurable tumor. In this study, a comprehensive screen of the TCA cycle identified oxoglutarate dehydrogenase (OGDH) and pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1) as the most clinically relevant genes in MM, highlighting their potential as therapeutic targets. CPI-613, a novel non-redox-active lipoic acid analog that causes mitochondrial metabolism dysfunction by targeting OGDH and PDHA1, is currently in clinical trials in a variety of malignancies. In our study, CPI-613 was found to inhibit the proliferation of MM cells, and its combination with bortezomib (BTZ) produced a significant inhibitory effect at lower doses. In addition, CPI-613 can disrupt various mitochondrial functions, such as disrupting mitochondrial morphology, reducing oxidative phosphorylation, decreasing 5'- adenylate triphosphate production, and increasing reactive oxygen species, which ultimately leads to cell death mediated by the intrinsic apoptotic pathway in vitro. Furthermore, we found CPI-613 significantly inhibited tumor growth and induced intrinsic apoptosis in the MM mouse xenograft model. This study reveals the mechanism and effect of CPI-613 in MM, which suggests that CPI-613 may be a new drug option for the clinical treatment of MM, but further clinical trials are needed for evaluation.
Collapse
Affiliation(s)
- Haiqin Wang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Yibin Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yu Jiang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Ruohong Xiang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Han Gong
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Yanfei Gong
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Hao Xu
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Zekang Ma
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Yifang Xie
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Yu Zhu
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Bin Hu
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Xiao He
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China.
| | - Ji Zhang
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, University of South China, Hengyang 421001, China.
| | - Xiaojuan Xiao
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
12
|
Bai J, Wan Z, Zhou W, Wang L, Lou W, Zhang Y, Jin H. Global trends and emerging insights in BRAF and MEK inhibitor resistance in melanoma: a bibliometric analysis. Front Mol Biosci 2025; 12:1538743. [PMID: 39897423 PMCID: PMC11782018 DOI: 10.3389/fmolb.2025.1538743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Objective This study aims to perform a comprehensive bibliometric analysis of global research on BRAF and MEK inhibitor resistance in melanoma, identifying key research trends, influential contributors, and emerging themes from 2003 to 2024. Methods A systematic search was conducted in the Web of Science Core Collection (WoSCC) database to retrieve publications related to BRAF and MEK inhibitor resistance from 1 January 2003, to 1 September 2024. Bibliometric analyses, including publication trends, citation networks, and keyword co-occurrence patterns, were performed using VOSviewer and CiteSpace. Collaborative networks, co-cited references, and keyword burst analyses were mapped to uncover shifts in research focus and global cooperation. Results A total of 3,503 documents, including 2,781 research articles and 722 review papers, were analyzed, highlighting significant growth in this field. The United States, China, and Italy led in publication volume and citation impact, with Harvard University and the University of California System among the top contributing institutions. Research output showed three phases of growth, peaking in 2020. Keyword and co-citation analyses revealed a transition from early focus on BRAF mutations and MAPK pathway activation to recent emphasis on immunotherapy, combination therapies, and non-apoptotic cell death mechanisms like ferroptosis and pyroptosis. These trends reflect the evolving priorities and innovative approaches shaping the field of resistance to BRAF and MEK inhibitors in melanoma. Conclusion Research on BRAF and MEK inhibitor resistance has evolved significantly. This analysis provides a strategic framework for future investigations, guiding the development of innovative, multi-modal approaches to improve treatment outcomes for melanoma patients.
Collapse
Affiliation(s)
- Jianhao Bai
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongqi Wan
- Department of Ophthalmology, Shanghai Tenth People’s Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai, China
| | - Wanru Zhou
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lijun Wang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Lou
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiying Jin
- Department of Ophthalmology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Huang A, Xue H, Xie T, Xiang L, Chen Z, Ma A, Yan H, Yuan J. A review of the pathogenesis of mitochondria in breast cancer and progress of targeting mitochondria for breast cancer treatment. J Transl Med 2025; 23:70. [PMID: 39815317 PMCID: PMC11734335 DOI: 10.1186/s12967-025-06077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025] Open
Abstract
With breast cancer being the most common tumor among women in the world today, it is also the leading cause of cancer-related deaths. Standard treatments include chemotherapy, surgery, endocrine therapy, and targeted therapy. However, the heterogeneity, drug resistance, and poor prognosis of breast cancer highlight an urgent need for further exploration of its underlying mechanisms. Mitochondria, highly dynamic intracellular organelles, play a pivotal role in maintaining cellular energy metabolism. Altered mitochondrial function plays a critical role in various diseases, and recent studies have elucidated its pathophysiological mechanisms in breast carcinogenesis. This review explores the role of mitochondrial dysfunction in breast cancer pathogenesis and assesses potential mitochondria-targeted therapies.
Collapse
Affiliation(s)
- Aoling Huang
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Haochen Xue
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Ting Xie
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Lingyan Xiang
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Zhengzhuo Chen
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Aolong Ma
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Jiefang-Road, Wuchang District, Wuhan, 430060, P. R. China.
| |
Collapse
|
14
|
Feng R, Guo Y, Chen M, Tian Z, Liu Y, Jiang S, Zhou J, Liu Q, Li X, Xiong W, Shi L, Fan S, Li G, Zhang W. PLUNC downregulates the expression of PD-L1 by inhibiting the interaction of DDX17/β-catenin in nasopharyngeal carcinoma. J Pathol Transl Med 2025; 59:68-83. [PMID: 39815745 PMCID: PMC11736280 DOI: 10.4132/jptm.2024.11.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is characterized by high programmed death-ligand 1 (PD-L1) expression and abundant infiltration of non-malignant lymphocytes, which renders patients potentially suitable candidates for immune checkpoint blockade therapies. Palate, lung, and nasal epithelium clone (PLUNC) inhibit the growth of NPC cells and enhance cellular apoptosis and differentiation. Currently, the relationship between PLUNC (as a tumor-suppressor) and PD-L1 in NPC is unclear. METHODS We collected clinical samples of NPC to verify the relationship between PLUNC and PD-L1. PLUNC plasmid was transfected into NPC cells, and the variation of PD-L1 was verified by western blot and immunofluorescence. In NPC cells, we verified the relationship of PD-L1, activating transcription factor 3 (ATF3), and β-catenin by western blot and immunofluorescence. Later, we further verified that PLUNC regulates PD-L1 through β-catenin. Finally, the effect of PLUNC on β-catenin was verified by co-immunoprecipitation (Co-IP). RESULTS We found that PLUNC expression was lower in NPC tissues than in paracancer tissues. PD-L1 expression was opposite to that of PLUNC. Western blot and immunofluorescence showed that β-catenin could upregulate ATF3 and PD-L1, while PLUNC could downregulate ATF3/PD-L1 by inhibiting the expression of β-catenin. PLUNC inhibits the entry of β-catenin into the nucleus. Co-IP experiments demonstrated that PLUNC inhibited the interaction of DEAD-box helicase 17 (DDX17) and β-catenin. CONCLUSIONS PLUNC downregulates the expression of PD-L1 by inhibiting the interaction of DDX17/β-catenin in NPC.
Collapse
Affiliation(s)
- Ranran Feng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Laboratory Medicine, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yilin Guo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Blood Transfusion, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Meilin Chen
- The First Affiliated Hospital of Sun Yatsen University, Guangzhou, China
| | - Ziying Tian
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Yijun Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Su Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Jieyu Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Qingluan Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Lei Shi
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Songqing Fan
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Wenling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Laboratory Science , Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
15
|
Cordani M, Michetti F, Zarrabi A, Zarepour A, Rumio C, Strippoli R, Marcucci F. The role of glycolysis in tumorigenesis: From biological aspects to therapeutic opportunities. Neoplasia 2024; 58:101076. [PMID: 39476482 PMCID: PMC11555605 DOI: 10.1016/j.neo.2024.101076] [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: 08/02/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 11/11/2024]
Abstract
Glycolytic metabolism generates energy and intermediates for biomass production. Tumor-associated glycolysis is upregulated compared to normal tissues in response to tumor cell-autonomous or non-autonomous stimuli. The consequences of this upregulation are twofold. First, the metabolic effects of glycolysis become predominant over those mediated by oxidative metabolism. Second, overexpressed components of the glycolytic pathway (i.e. enzymes or metabolites) acquire new functions unrelated to their metabolic effects and which are referred to as "moonlighting" functions. These functions include induction of mutations and other tumor-initiating events, effects on cancer stem cells, induction of increased expression and/or activity of oncoproteins, epigenetic and transcriptional modifications, bypassing of senescence and induction of proliferation, promotion of DNA damage repair and prevention of DNA damage, antiapoptotic effects, inhibition of drug influx or increase of drug efflux. Upregulated metabolic functions and acquisition of new, non-metabolic functions lead to biological effects that support tumorigenesis: promotion of tumor initiation, stimulation of tumor cell proliferation and primary tumor growth, induction of epithelial-mesenchymal transition, autophagy and metastasis, immunosuppressive effects, induction of drug resistance and effects on tumor accessory cells. These effects have negative consequences on the prognosis of tumor patients. On these grounds, it does not come to surprise that tumor-associated glycolysis has become a target of interest in antitumor drug discovery. So far, however, clinical results with glycolysis inhibitors have fallen short of expectations. In this review we propose approaches that may allow to bypass some of the difficulties that have been encountered so far with the therapeutic use of glycolysis inhibitors.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Federica Michetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, Rome 00161, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, Rome 00149, Italy.
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, Milan 20134, Italy.
| |
Collapse
|
16
|
Pan Y, Zhang Y, Mao D, Fang Z, Ma Y, Jin D, Li S. Multi-omics Insights into PDHA1 as a Predictive Biomarker for Prognosis, Immunotherapy Efficacy, and Drug Sensitivity in Hepatocellular Carcinoma. ACS OMEGA 2024; 9:46492-46504. [PMID: 39583658 PMCID: PMC11579764 DOI: 10.1021/acsomega.4c08010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
PDHA1 was associated with metabolic reprogramming in tumor progression. However, the clinical value of PDHA1, especially for prediction of drug sensitivity in hepatocellular carcinoma (HCC), has not been fully investigated. In this study, we found that PDHA1 expression was higher in HCC tissues compared to normal tissues and was correlated with poor prognosis in HCC patients. PDHA1 expression was mainly positively associated with immune cell infiltration using the TIMER, XCell, MCPCOUNTER, CIBERSORT, EPIC, and QUANTISEQ algorithms, which was validated by single-cell RNA-sequencing analysis. We also discovered that PDHA1 expression was correlated with six immune checkpoint-related genes. Univariate and multivariate Cox regression analyses revealed that PDHA1 expression was an independent prognostic indicator for HCC patients, and the nomogram incorporating PDHA1 expression exhibited excellent predictive capacity. Furthermore, PDHA1 expression was positively linked to the sensitivity of 5-fluorouracil, gemcitabine, paclitaxel, and sorafenib, and the molecular docking analysis demonstrated their excellent binding affinity.
Collapse
Affiliation(s)
- Yong Pan
- Department
of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316021, China
- State
Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Yiru Zhang
- State
Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital of Zhejiang University, Hangzhou 310003, China
| | - Daiwen Mao
- Department
of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316021, China
| | - Zhou Fang
- Department
of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316021, China
| | - Yingqiu Ma
- Department
of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316021, China
| | - Danwen Jin
- Pathological
Diagnosis Center, Zhoushan Hospital, Wenzhou
Medical University, Zhoushan 316021, China
| | - Shibo Li
- Department
of Infectious Diseases, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316021, China
| |
Collapse
|
17
|
Phomvisith O, Muroya S, Otomaru K, Oshima K, Oshima I, Nishino D, Haginouchi T, Gotoh T. Maternal Undernutrition Affects Fetal Thymus DNA Methylation, Gene Expression, and, Thereby, Metabolism and Immunopoiesis in Wagyu (Japanese Black) Cattle. Int J Mol Sci 2024; 25:9242. [PMID: 39273192 PMCID: PMC11395129 DOI: 10.3390/ijms25179242] [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: 07/30/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
We aimed to determine the effects of maternal nutrient restriction (MNR) on the DNA methylation and gene expression patterns associated with metabolism and immunopoiesis in the thymuses of fetal Wagyu cattle. Pregnant cows were allocated to two groups: a low-nutrition (LN; 60% nutritional requirement; n = 5) and a high-nutrition (HN; 120% nutritional requirement, n = 6) group, until 8.5 months of gestation. Whole-genome bisulfite sequencing (WGBS) and RNA sequencing were used to analyze DNA methylation and gene expression, while capillary electrophoresis-Fourier transform mass spectrometry assessed the metabolome. WGBS identified 4566 hypomethylated and 4303 hypermethylated genes in the LN group, with the intergenic regions most frequently being methylated. Pathway analysis linked hypoDMGs to Ras signaling, while hyperDMGs were associated with Hippo signaling. RNA sequencing found 94 differentially expressed genes (66 upregulated, 28 downregulated) in the LN group. The upregulated genes were tied to metabolic pathways and oxidative phosphorylation; the downregulated genes were linked to natural killer cell cytotoxicity. Key overlapping genes (GRIA1, CACNA1D, SCL25A4) were involved in cAMP signaling. The metabolomic analysis indicated an altered amino acid metabolism in the MNR fetuses. These findings suggest that MNR affects DNA methylation, gene expression, and the amino acid metabolism, impacting immune system regulation during fetal thymus development in Wagyu cattle.
Collapse
Affiliation(s)
- Ouanh Phomvisith
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| | - Susumu Muroya
- Department of Animal Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Konosuke Otomaru
- Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Kazunaga Oshima
- Division of Year-Round Grazing Research, NARO Western Region Agricultural Research Center, 60 Yoshinaga, Ohda 694-0013, Shimane, Japan
| | - Ichiro Oshima
- Department of Animal Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-8580, Kagoshima, Japan
| | - Daichi Nishino
- Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Fukuoka, Japan
| | - Taketo Haginouchi
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| | - Takafumi Gotoh
- Field Science Center for Northern Biosphere, Hokkaido University, N11W10, Kita, Sapporo 060-0811, Hokkaido, Japan
| |
Collapse
|
18
|
Shan Z, Liu F. Advances in immunotherapy for mucosal melanoma: harnessing immune checkpoint inhibitors for improved treatment outcomes. Front Immunol 2024; 15:1441410. [PMID: 39234260 PMCID: PMC11373357 DOI: 10.3389/fimmu.2024.1441410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 09/06/2024] Open
Abstract
Mucosal melanoma (MM) poses a significant clinical challenge due to its aggressive nature and limited treatment options. In recent years, immunotherapy has emerged as a promising strategy for MM, with a particular focus on immune checkpoint inhibitors such as PD-1 and CTLA-4 inhibitors. These inhibitors have demonstrated substantial efficacy by harnessing the body's immune response against tumors. Moreover, adoptive cell transfer (ACT), anti-angiogenic therapy, and combination therapies have garnered attention for their potential in MM treatment. ACT involves modifying T cells to target melanoma cells, showing promising antitumor activity. Anti-angiogenic therapy aims to impede tumor growth by inhibiting angiogenesis, while combination therapies, including immune checkpoint inhibitors and targeted therapies, offer a multifaceted approach to overcome treatment resistance. This comprehensive review explores the advancements in immunotherapy for MM, highlighting the role of diverse therapeutic modalities in enhancing treatment outcomes and addressing the challenges posed by this aggressive malignancy.
Collapse
Affiliation(s)
- Zexing Shan
- Department of Gastric Surgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Fei Liu
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
19
|
Di SC, Chen WJ, Yang W, Zhang XM, Dong KQ, Tian YJ, Sun Y, Qian C, Chen JX, Liu ZC, Gong ZX, Chu J, Zhou W, Pan XW, Cui XG. DEPDC1 as a metabolic target regulates glycolysis in renal cell carcinoma through AKT/mTOR/HIF1α pathway. Cell Death Dis 2024; 15:533. [PMID: 39068164 PMCID: PMC11283501 DOI: 10.1038/s41419-024-06913-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Renal cell carcinoma (RCC) is considered a "metabolic disease" characterized by elevated glycolysis in patients with advanced RCC. Tyrosine kinase inhibitor (TKI) therapy is currently an important treatment option for advanced RCC, but drug resistance may develop in some patients. Combining TKI with targeted metabolic therapy may provide a more effective approach for patients with advanced RCC. An analysis of 14 RCC patients (including three needle biopsy samples with TKI resistance) revealed by sing-cell RNA sequencing (scRNA-seq) that glycolysis played a crucial role in poor prognosis and drug resistance in RCC. TCGA-KIRC and glycolysis gene set analysis identified DEPDC1 as a target associated with malignant progression and drug resistance in KIRC. Subsequent experiments demonstrated that DEPDC1 promoted malignant progression and glycolysis of RCC, and knockdown DEPDC1 could reverse TKI resistance in RCC cell lines. Bulk RNA sequencing (RNA-seq) and non-targeted metabolomics sequencing suggested that DEPDC1 may regulate RCC glycolysis via AKT/mTOR/HIF1α pathway, a finding supported by protein-level analysis. Clinical tissue samples from 98 RCC patients demonstrated that DEPDC1 was associated with poor prognosis and predicted RCC metastasis. In conclusion, this multi-omics analysis suggests that DEPDC1 could serve as a novel target for TKI combined with targeted metabolic therapy in advanced RCC patients with TKI resistance.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/drug therapy
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Regulation, Neoplastic
- Glycolysis/drug effects
- GTPase-Activating Proteins/metabolism
- GTPase-Activating Proteins/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/genetics
- Mice, Nude
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- TOR Serine-Threonine Kinases/metabolism
Collapse
Affiliation(s)
- Si-Chen Di
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wen-Jin Chen
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, China
| | - Wei Yang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiang-Min Zhang
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Ke-Qin Dong
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Department of Urology, Chinese PLA General Hospital of Central Theater Command, Wuhan, China
| | - Yi-Jun Tian
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ye Sun
- Department of Urology, Taian 88 Hospital, Taian, Shandong, China
| | - Cheng Qian
- Department of Urology, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Jia-Xin Chen
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Chang Liu
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Xuan Gong
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jian Chu
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, China.
| | - Wang Zhou
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Xiu-Wu Pan
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Xin-Gang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
20
|
Agarwala Y, Brauns TA, Sluder AE, Poznansky MC, Gemechu Y. Targeting metabolic pathways to counter cancer immunotherapy resistance. Trends Immunol 2024; 45:486-494. [PMID: 38876831 DOI: 10.1016/j.it.2024.05.006] [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: 04/24/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/16/2024]
Abstract
Immunotherapies have revolutionized the treatment of certain cancers, but challenges remain in overcoming immunotherapy resistance. Research shows that metabolic modulation of the tumor microenvironment can enhance antitumor immunity. Here, we discuss recent preclinical and clinical evidence for the efficacy of combining metabolic modifiers with immunotherapies. While this combination holds great promise, a few key areas must be addressed, which include identifying the effects of metabolic modifiers on immune cell metabolism, the putative biomarkers of therapeutic efficacy, the efficacy of modifiers on tumors harboring metabolic heterogeneity, and the potential development of resistance due to tumor reliance on alternative metabolic pathways. We propose solutions to these problems and posit that assessing these parameters is crucial for considering the potential of metabolic modifiers in sensitizing tumors to immunotherapies.
Collapse
Affiliation(s)
- Yuki Agarwala
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Imperial College School of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Timothy A Brauns
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ann E Sluder
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yohannes Gemechu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Zhang Y, Yan H, Wei Y, Wei X. Decoding mitochondria's role in immunity and cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189107. [PMID: 38734035 DOI: 10.1016/j.bbcan.2024.189107] [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: 02/08/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The functions of mitochondria, including energy production and biomolecule synthesis, have been known for a long time. Given the rising incidence of cancer, the role of mitochondria in cancer has become increasingly popular. Activated by components released by mitochondria, various pathways interact with each other to induce immune responses to protect organisms from attack. However, mitochondria play dual roles in the progression of cancer. Abnormalities in proteins, which are the elementary structures of mitochondria, are closely linked with oncogenesis. Both the aberrant accumulation of intermediates and mutations in enzymes result in the generation and progression of cancer. Therefore, targeting mitochondria to treat cancer may be a new strategy. Several drugs aimed at inhibiting mutated enzymes and accumulated intermediates have been tested clinically. Here, we discuss the current understanding of mitochondria in cancer and the interactions between mitochondrial functions, immune responses, and oncogenesis. Furthermore, we discuss mitochondria as hopeful targets for cancer therapy, providing insights into the progression of future therapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Hong Yan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| |
Collapse
|
22
|
Wu J, Liu N, Chen J, Tao Q, Li Q, Li J, Chen X, Peng C. The Tricarboxylic Acid Cycle Metabolites for Cancer: Friend or Enemy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0351. [PMID: 38867720 PMCID: PMC11168306 DOI: 10.34133/research.0351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/18/2024] [Indexed: 06/14/2024]
Abstract
The tricarboxylic acid (TCA) cycle is capable of providing sufficient energy for the physiological activities under aerobic conditions. Although tumor metabolic reprogramming places aerobic glycolysis in a dominant position, the TCA cycle remains indispensable for tumor cells as a hub for the metabolic linkage and interconversion of glucose, lipids, and certain amino acids. TCA intermediates such as citrate, α-ketoglutarate, succinate, and fumarate are altered in tumors, and they regulate the tumor metabolism, signal transduction, and immune environment to affect tumorigenesis and tumor progression. This article provides a comprehensive review of the modifications occurring in tumor cells in relation to the intermediates of the TCA cycle, which affects tumor pathogenesis and current therapeutic strategy for therapy through targeting TCA cycle in cancer cells.
Collapse
Affiliation(s)
- Jie Wu
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Nian Liu
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jing Chen
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Qian Tao
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Qiuqiu Li
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jie Li
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Cong Peng
- The Department of Dermatology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Furong Labratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
Chen D, Liu P, Lu X, Li J, Qi D, Zang L, Lin J, Liu Y, Zhai S, Fu D, Weng Y, Li H, Shen B. Pan-cancer analysis implicates novel insights of lactate metabolism into immunotherapy response prediction and survival prognostication. J Exp Clin Cancer Res 2024; 43:125. [PMID: 38664705 PMCID: PMC11044366 DOI: 10.1186/s13046-024-03042-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Immunotherapy has emerged as a potent clinical approach for cancer treatment, but only subsets of cancer patients can benefit from it. Targeting lactate metabolism (LM) in tumor cells as a method to potentiate anti-tumor immune responses represents a promising therapeutic strategy. METHODS Public single-cell RNA-Seq (scRNA-seq) cohorts collected from patients who received immunotherapy were systematically gathered and scrutinized to delineate the association between LM and the immunotherapy response. A novel LM-related signature (LM.SIG) was formulated through an extensive examination of 40 pan-cancer scRNA-seq cohorts. Then, multiple machine learning (ML) algorithms were employed to validate the capacity of LM.SIG for immunotherapy response prediction and survival prognostication based on 8 immunotherapy transcriptomic cohorts and 30 The Cancer Genome Atlas (TCGA) pan-cancer datasets. Moreover, potential targets for immunotherapy were identified based on 17 CRISPR datasets and validated via in vivo and in vitro experiments. RESULTS The assessment of LM was confirmed to possess a substantial relationship with immunotherapy resistance in 2 immunotherapy scRNA-seq cohorts. Based on large-scale pan-cancer data, there exists a notably adverse correlation between LM.SIG and anti-tumor immunity as well as imbalance infiltration of immune cells, whereas a positive association was observed between LM.SIG and pro-tumorigenic signaling. Utilizing this signature, the ML model predicted immunotherapy response and prognosis with an AUC of 0.73/0.80 in validation sets and 0.70/0.87 in testing sets respectively. Notably, LM.SIG exhibited superior predictive performance across various cancers compared to published signatures. Subsequently, CRISPR screening identified LDHA as a pan-cancer biomarker for estimating immunotherapy response and survival probability which was further validated using immunohistochemistry (IHC) and spatial transcriptomics (ST) datasets. Furthermore, experiments demonstrated that LDHA deficiency in pancreatic cancer elevated the CD8+ T cell antitumor immunity and improved macrophage antitumoral polarization, which in turn enhanced the efficacy of immunotherapy. CONCLUSIONS We unveiled the tight correlation between LM and resistance to immunotherapy and further established the pan-cancer LM.SIG, holds the potential to emerge as a competitive instrument for the selection of patients suitable for immunotherapy.
Collapse
Affiliation(s)
- Dongjie Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Pengyi Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Xiongxiong Lu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jingfeng Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Debin Qi
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Longjun Zang
- Department of General Surgery, Taiyuan Central Hospital, Taiyuan, Shanxi, 030009, China
| | - Jiayu Lin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yihao Liu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Shuyu Zhai
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Da Fu
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Yuanchi Weng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Hongzhe Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Research Institute of Pancreatic Diseases, Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|