1
|
Xie Y, Wang R, Xu M, Chen J, Tan W, Chen Y, Bai Y, Wu N, Wu F, Xu X, Ma X, Liu Y. Potential of CLSPN as a therapeutic target in melanoma: a key player in melanoma progression and tumor microenvironment. J Transl Med 2025; 23:470. [PMID: 40275302 PMCID: PMC12020306 DOI: 10.1186/s12967-025-06455-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Melanoma is a highly aggressive form of skin cancer. Despite significant advances in targeted therapies and immunotherapeutic approaches, some patients still have poor response rates, making a deeper understanding of melanoma pathogenesis essential. METHODS The expression of Claspin (CLSPN), prognosis and immune infiltration in skin cutaneous melanoma patients were analyzed by public databases. Immunohistochemistry was used to validate. Moreover, quantitative real-time polymerase chain reaction analysis, western blot, cell counting kit-8 assay, colony formation assay, flow cytometry, animal experiments, and RNA-seq were applied to explore its biological functions and potential molecular mechanisms of CLSPN in melanoma. RESULTS Our results demonstrated that abnormal CLSPN expression was correlated with poor prognosis in melanoma. Meanwhile, CLSPN may promote melanoma growth and progression in vivo and in vitro through IFI44L/JAK/STAT1 signaling. Additionally, CLSPN was associated with negative immune microenvironment in melanoma and may be related to polarization of tumor associated macrophages towards M2-type. CONCLUSIONS These findings suggest that CLSPN may be a promising new target for melanoma and accelerate personalized therapeutic strategies.
Collapse
Affiliation(s)
- Yongyi Xie
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Ruoqi Wang
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Skin Disease Hospital, Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai, 200443, China
| | - Mingyuan Xu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Jiashe Chen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Wei Tan
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Yanbin Chen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Yun Bai
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Nanhui Wu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Fei Wu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxiang Xu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China
| | - Xin Ma
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China.
- , Baode Road 1278 street, Shanghai, 200433, China.
| | - Yeqiang Liu
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Skin Disease Hospital, Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai, 200443, China.
- , Baode Road 1278 street, Shanghai, 200433, China.
| |
Collapse
|
2
|
Bonofiglio D. Effects of Mediterranean Diet on Chronic Degenerative Diseases and Human Healthy Lifestyle. Nutrients 2025; 17:1231. [PMID: 40218989 PMCID: PMC11990738 DOI: 10.3390/nu17071231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Noncommunicable diseases (NCDs), known as chronic diseases, including diabetes mellitus, obesity, cardiovascular diseases, chronic kidney disease, neurodegenerative diseases, and cancers, represent an important public concern, leading to prolonged disability and death [...].
Collapse
Affiliation(s)
- Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Cosenza, Italy; ; Tel.: +39-0984-496208
- Centro Sanitario, University of Calabria, 87036 Rende, Cosenza, Italy
| |
Collapse
|
3
|
Yan D, Hou Y, Lei X, Xiao H, Zeng Z, Xiong W, Fan C. The Impact of Polyunsaturated Fatty Acids in Cancer and Therapeutic Strategies. Curr Nutr Rep 2025; 14:46. [PMID: 40085324 DOI: 10.1007/s13668-025-00639-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE OF REVIEW Cancer is a disease influenced by both genetic and environmental factors, with dietary lipids being a significant contributing factor. This review summarizes the role of polyunsaturated fatty acids (PUFAs) in the mechanism of tumor occurrence and development, and elucidate the role of PUFAs in tumor treatment. RECENT FINDINGS PUFAs exert their impact on cancer through altering lipid composition in cell membranes, interacting with cell membrane lipid receptors, directly modulating gene expression in the cell nucleus, and participating in the metabolism of lipid mediators. Most omega-3 PUFAs are believed to inhibit cell proliferation, promote cancer cell death, suppress cancer metastasis, alter energy metabolism, inhibit tumor microenvironment inflammation, and regulate immune responses involving macrophages, T cells, NK cells, and others. However, certain omega-6 PUFAs exhibit weaker anti-tumor effects and may even promote tumor development, such as by fostering inflammatory tumor microenvironment and enhancing tumor cell proliferation. PUFAs play important roles in hallmarks of cancer including tumor cell proliferation, cell death, migration and invasion, energy metabolism remodeling, epigenetics, and immunity. These findings provide insights into the mechanisms of cancer development and offers options for dietary management of cancer.
Collapse
Affiliation(s)
- Dong Yan
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Yingshan Hou
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Xinyi Lei
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Hao Xiao
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Zhaoyang Zeng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
- Department of Histology and Embryology, School of Basic Medicine Sciences, Central South University, Changsha, 410013, Hunan Province, China.
| |
Collapse
|
4
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
van der Meij B, Parsons S, Mazurak V. The impact of n-3 polyunsaturated fatty acids in patients with cancer: emerging themes. Curr Opin Clin Nutr Metab Care 2025; 28:75-85. [PMID: 39750501 DOI: 10.1097/mco.0000000000001102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
PURPOSE OF REVIEW This review summarizes recent literature falling broadly under the topic of n-3 polyunsaturated fatty acids (PUFAs) in the oncology setting, highlighting emerging themes and emphasizing novel explorations. RECENT FINDINGS Meta-analyses continue to confirm safety and efficacy of n-3 PUFA supplementation on reducing inflammation and improving survival in people with cancer. Common themes in recent studies emphasize improving tumor-directed efficacy and reducing toxicities of common cancer therapies. New areas of interest include the impact of n-3 PUFA when combined with immunotherapies and applications in pediatric acute lymphoid leukemia. Novel assessments include specialized pro-resolving lipid mediators, the intestinal microbiome and psychological well being. A variety of clinically relevant outcomes including nutritional status, toxicities and survival are being explored in ongoing clinical studies. SUMMARY Evidence confirms the safety of n-3 PUFA for patients with cancers, as well as benefits in some, but not all areas of exploration. Larger, well designed trials with biological assessment of compliance compared to the prescribed n-3 PUFA dose would strengthen the evidence needed to integrate n-3 PUFA recommendations into clinical practice for patients with cancer.
Collapse
Affiliation(s)
- Barbara van der Meij
- Department of Human Nutrition and Health, Wageningen University & Research
- Nutrition, Dietetics and Lifestyle, HAN University of Applied Sciences, Nijmegen, the Netherlands
| | - Sarah Parsons
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science. University of Alberta, Edmonton, Alberta, Canada
| | - Vera Mazurak
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science. University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Marchio V, Augimeri G, Morelli C, Vivacqua A, Giordano C, Catalano S, Sisci D, Barone I, Bonofiglio D. Omega-3 fatty acids: molecular weapons against chemoresistance in breast cancer. Cell Mol Biol Lett 2025; 30:11. [PMID: 39863855 PMCID: PMC11762563 DOI: 10.1186/s11658-025-00694-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Breast cancer is the most commonly diagnosed type of cancer and the leading cause of cancer-related death in women worldwide. Highly targeted therapies have been developed for different subtypes of breast cancer, including hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, triple-negative breast cancer (TNBC) and metastatic breast cancer disease are primarily treated with chemotherapy, which improves disease-free and overall survival, but does not offer a curative solution for these aggressive forms of breast cancer. Moreover, the development of chemoresistance is a major cause of therapeutic failure in this neoplasia, leading to disease relapse and patient death. In addition, chemotherapy's adverse side effects may substantially worsen health-related quality of life. Therefore, to improve the outcome of patients with breast cancer who are undergoing chemotherapy, several therapeutic options are under investigation, including the combination of chemotherapeutic drugs with natural compounds. Omega-3 (ω-3) polyunsaturated fatty acids (PUFAs), including docosahexaenoic and eicosapentaenoic acids, have drawn attention for their antitumoral properties and their preventive activities against chemotherapy-induced toxicities in breast cancer. A literature review was conducted on PubMed using keywords related to breast cancer, omega-3, chemoresistance, and chemotherapy. This review aims to provide an overview of the molecular mechanisms driving breast cancer chemoresistance, focusing on the role of ω-3 PUFAs in these recognized cellular paths and presenting current findings on the effects of ω-3 PUFAs combined with chemotherapeutic drugs in breast cancer management.
Collapse
Affiliation(s)
- Vittoria Marchio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Catia Morelli
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Diego Sisci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy.
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
- Centro Sanitario, University of Calabria, Via P. Bucci, Arcavacata Di Rende (CS), 87036, Rende, Cosenza, Italy
| |
Collapse
|
7
|
Rahman MK, Umashankar B, Choucair H, Bourget K, Rawling T, Murray M. The inositol-requiring enzyme 1 (IRE1) endoplasmic reticulum stress pathway promotes MDA-MB-231 cell survival and renewal in response to the aryl-ureido fatty acid CTU. Int J Biochem Cell Biol 2024; 171:106571. [PMID: 38608921 DOI: 10.1016/j.biocel.2024.106571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
Current treatment options for triple-negative breast cancer (TNBC) are limited to toxic drug combinations of low efficacy. We recently identified an aryl-substituted fatty acid analogue, termed CTU, that effectively killed TNBC cells in vitro and in mouse xenograft models in vivo without producing toxicity. However, there was a residual cell population that survived treatment. The present study evaluated the mechanisms that underlie survival and renewal in CTU-treated MDA-MB-231 TNBC cells. RNA-seq profiling identified several pro-inflammatory signaling pathways that were activated in treated cells. Increased expression of cyclooxygenase-2 and the cytokines IL-6, IL-8 and GM-CSF was confirmed by real-time RT-PCR, ELISA and Western blot analysis. Increased self-renewal was confirmed using the non-adherent, in vitro colony-forming mammosphere assay. Neutralizing antibodies to IL-6, IL-8 and GM-CSF, as well as cyclooxygenase-2 inhibition suppressed the self-renewal of MDA-MB-231 cells post-CTU treatment. IPA network analysis identified major NF-κB and XBP1 gene networks that were activated by CTU; chemical inhibitors of these pathways and esiRNA knock-down decreased the production of pro-inflammatory mediators. NF-κB and XBP1 signaling was in turn activated by the endoplasmic reticulum (ER)-stress sensor inositol-requiring enzyme 1 (IRE1), which mediates the unfolded protein response. Co-treatment with an inhibitor of IRE1 kinase and RNase activities, decreased phospho-NF-κB and XBP1s expression and the production of pro-inflammatory mediators. Further, IRE1 inhibition also enhanced apoptotic cell death and prevented the activation of self-renewal by CTU. Taken together, the present findings indicate that the IRE1 ER-stress pathway is activated by the anti-cancer lipid analogue CTU, which then activates secondary self-renewal in TNBC cells.
Collapse
Affiliation(s)
- Md Khalilur Rahman
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, and School of Pharmacy, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Balasubrahmanyam Umashankar
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, and School of Pharmacy, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Hassan Choucair
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, and School of Pharmacy, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Kirsi Bourget
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, and School of Pharmacy, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Tristan Rawling
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Michael Murray
- Pharmacogenomics and Drug Development Group, Discipline of Pharmacology, School of Medical Sciences, and School of Pharmacy, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
8
|
Jiang S, Xu L, Chen Y, Shu Z, Lv L, Zhao Y, Bi K, Yang S, Wang Q, Li L. Longitudinal gut fungal alterations and potential fungal biomarkers for the progression of primary liver disease. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1183-1198. [PMID: 38413553 DOI: 10.1007/s11427-023-2458-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/25/2023] [Indexed: 02/29/2024]
Abstract
Liver disease, a major health concern worldwide, is a serious and progressive disorder. Herein, we not only established a mouse model of DEN+CCl4-induced primary liver disease but also collected clinical human samples to investigate longitudinal alterations in the gut mycobiome. As liver disease advanced, gut integrity was disrupted, and the mycobiota was disturbed in the mouse models. The metabolites associated with hepatocellular carcinoma (HCC) differed from those associated with the cirrhotic phase as follows: levels of stercobilin and aflatoxin B1 dialcohol were reduced, while levels of triterpenoids, bafilomycin A1, and DHEA were increased in the HCC group. The abundance of the phylum Chytridiomycota increased as the chronic liver disease progressed and was then replaced by the phylum Ascomycota in HCC. Based on the results from clinical human samples, the genus Candida (Ascomycota) (in humans) and the genus Kazachstania (Ascomycota) (in mice) occupied a dominant position in the HCC group, while other fungi were depleted. The increased abundance of C. albicans and depletion of S. cerevisiae may be hallmarks of the progression of liver cirrhosis to early HCC. Moreover, the administration of C. albicans and S. cerevisiae in the LC-HCC progression could accelerate or retard the progression of HCC. Therefore, gut fungi have the potential to serve as a noninvasive clinical biomarker and even a treatment method.
Collapse
Affiliation(s)
- Shiman Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lvwan Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zheyue Shu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuxi Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Kefan Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Sisi Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
9
|
Chang J, Xin C, Wang Y, Wang Y. Dihydroartemisinin inhibits liver cancer cell migration and invasion by reducing ATP synthase production through CaMKK2/NCLX. Oncol Lett 2023; 26:540. [PMID: 38020296 PMCID: PMC10660190 DOI: 10.3892/ol.2023.14127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/08/2023] [Indexed: 12/01/2023] Open
Abstract
Calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) and mitochondrial sodium/calcium exchanger protein (NCLX) are key regulatory factors in calcium homeostasis. Finding natural drugs that target regulators of calcium homeostasis is critical. Dihydroartemisinin (DHA) is considered to have anticancer effects. The present study aimed to investigate the mechanism of DHA in regulating liver cancer migration and invasion. The present study used HepG2 and HuH-7 cells and overexpressed CaMKK2 and knocked down CaMKK2 and NCLX. The antiproliferative activity of DHA on liver cancer cells was assessed through colony formation and EdU assays. Cell apoptosis was detected through YO-PRO-1/PI staining. The levels of reactive oxygen species (ROS) were measured using a ROS detection kit (DCFH-DA fluorescent probe). Cell migratory and invasive abilities were examined using wound healing and Transwell assays. The ATP production of liver cancer cells was detected using ATP fluorescent probes. Cell microfilaments were monitored for changes using Actin-Tracker Green-488. The effects of DHA on the expression of CaMKK2, NCLX, sodium/potassium-transporting ATPase subunit α-1 (ATP1A1) and ATP synthase subunit d, mitochondrial (ATP5H) were determined by western blotting and reverse transcription-quantitative PCR. The results revealed that DHA significantly inhibited proliferation, reduced ROS levels and promoted apoptosis in liver cancer cells. CaMKK2 overexpression significantly enhanced the invasive and migratory ability of liver cancer cells, whereas DHA inhibited the pro-migratory effects of CaMKK2 overexpression. DHA significantly reduced the mitochondrial ATP production and altered the arrangement of microfilaments in liver cancer cells. In addition, DHA significantly decreased the expression of CaMKK2, NCLX, ATP1A1 and ATP5H. Furthermore, by knockdown experiments of NCLX the results demonstrated that CaMKK2 downregulated the expression of ATP1A1 and ATP5H in liver cancer cells through NCLX. In conclusion, DHA may reduce ATP synthase production via the CaMKK2/NCLX signaling pathway to inhibit the invasive phenotype of liver cancer cells. It is essential to further investigate the effectiveness of DHA in the anticancer mechanism of liver cancer cells.
Collapse
Affiliation(s)
- Jiang Chang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Chengyi Xin
- Department of Pharmacy, Bayannur Hospital, Bayannur, Inner Mongolia Autonomous Region 015000, P.R. China
| | - Yong Wang
- Department of Neurosurgery, Hainan West Central Hospital, Danzhou, Hainan 571700, P.R. China
| | - Ying Wang
- Department of General Practice, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| |
Collapse
|
10
|
Augimeri G, Bonofiglio D. Promising Effects of N-Docosahexaenoyl Ethanolamine in Breast Cancer: Molecular and Cellular Insights. Molecules 2023; 28:molecules28093694. [PMID: 37175104 PMCID: PMC10180201 DOI: 10.3390/molecules28093694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
Unhealthy dietary habits have been identified as a risk factor for the development and progression of cancer. Therefore, adopting a healthy eating pattern is currently recommended to prevent the onset of different types of cancers, including breast carcinoma. In particular, the Mediterranean diet, based on high consumption of omega-3 polyunsaturated fatty acids (N-3 PUFAs), such as those found in cold-water fish and other seafood, nuts, and seeds, is recommended to reduce the incidence of several chronic-degenerative diseases. Indeed, the consumption of N-3 PUFAs, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), reduced the risk of different types of cancer, including breast cancer. Moreover, they can counteract breast cancer progression and reduce the side effects of chemotherapy in breast cancer survival. Studies have demonstrated that DHA, exhibiting greater antitumor activity than EPA in breast cancer, can be attributed to its direct impact on breast cancer cells and also due to its conversion into various metabolites. N-docosahexaenoyl ethanolamine, DHEA, is the most studied DHA derivative for its therapeutic potential in breast cancer. In this review, we emphasize the significance of dietary habits and the consumption of N-3 polyunsaturated fatty acids, particularly DHA, and we describe the current knowledge on the antitumoral action of DHA and its derivative DHEA in the treatment of breast cancer.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, CS, Italy
- Centro Sanitario, University of Calabria, 87036 Rende, CS, Italy
| |
Collapse
|
11
|
Munkácsy G, Santarpia L, Győrffy B. Therapeutic Potential of Tumor Metabolic Reprogramming in Triple-Negative Breast Cancer. Int J Mol Sci 2023; 24:ijms24086945. [PMID: 37108109 PMCID: PMC10138520 DOI: 10.3390/ijms24086945] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with clinical features of high metastatic potential, susceptibility to relapse, and poor prognosis. TNBC lacks the expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). It is characterized by genomic and transcriptional heterogeneity and a tumor microenvironment (TME) with the presence of high levels of stromal tumor-infiltrating lymphocytes (TILs), immunogenicity, and an important immunosuppressive landscape. Recent evidence suggests that metabolic changes in the TME play a key role in molding tumor development by impacting the stromal and immune cell fractions, TME composition, and activation. Hence, a complex inter-talk between metabolic and TME signaling in TNBC exists, highlighting the possibility of uncovering and investigating novel therapeutic targets. A better understanding of the interaction between the TME and tumor cells, and the underlying molecular mechanisms of cell-cell communication signaling, may uncover additional targets for better therapeutic strategies in TNBC treatment. In this review, we aim to discuss the mechanisms in tumor metabolic reprogramming, linking these changes to potential targetable molecular mechanisms to generate new, physical science-inspired clinical translational insights for the cure of TNBC.
Collapse
Affiliation(s)
- Gyöngyi Munkácsy
- National Laboratory for Drug Research and Development, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary
- Oncology Biomarker Research Group, Research Centre for Natural Sciences, Institute of Enzymology, Magyar Tudósok Körútja 2, 1117 Budapest, Hungary
| | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Tűzoltó u. 5-7, 1094 Budapest, Hungary
- Department of Pediatrics, Semmelweis University, Tűzoltó u. 5-7, 1094 Budapest, Hungary
| |
Collapse
|