1
|
Gurung S, Budden T, Mallela K, Jenkins B, von Kriegsheim A, Manrique E, Millán-Esteban D, Romero-Camarero I, Amaral F, Craig S, Durao P, Pozniak J, Stennett L, Smith D, Ashton G, Baker A, Zeng K, Fruhwirth G, Sanz-Moreno V, Marques J, Koulman A, Marine JC, Somervaille TCP, Motta L, Gaudy-Marqueste C, Nagore E, Virós A. Stromal lipid species dictate melanoma metastasis and tropism. Cancer Cell 2025:S1535-6108(25)00138-2. [PMID: 40280124 DOI: 10.1016/j.ccell.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/14/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
Cancer cells adapt to signals in the tumor microenvironment (TME), but the TME cues that impact metastasis and tropism are still incompletely understood. We show that abundant stromal lipids from young subcutaneous adipocytes, including phosphatidylcholines, are taken up by melanoma cells, where they upregulate melanoma PI3K-AKT signaling, fatty acid oxidation, oxidative phosphorylation (OXPHOS) leading to oxidative stress, resulting in decreased metastatic burden. High OXPHOS melanoma cells predominantly seed the lung and brain; decreasing oxidative stress with antioxidants shifts tropism from the lung to the liver. By contrast, the aged TME provides fewer total lipids but is rich in ceramides, leading to lower OXPHOS and high metastatic burden. Aged TME ceramides taken up by melanoma cells activate the S1P-STAT3-IL-6 signaling axis and promote liver tropism. Inhibiting OXPHOS in the young TME or blocking the IL-6 receptor in the aged TME reduces the age-specific patterns of metastasis imposed by lipid availability.
Collapse
Affiliation(s)
- Shilpa Gurung
- Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, the University of Manchester, Manchester, UK
| | - Timothy Budden
- Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, the University of Manchester, Manchester, UK
| | - Karthik Mallela
- Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, the University of Manchester, Manchester, UK
| | - Benjamin Jenkins
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK; Medical Research Council Human Nutrition Research, Elsie Widdowson Laboratory, University of Cambridge, Fulbourn Road, Cambridge CB1 9NL, UK
| | - Alex von Kriegsheim
- CRUK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Esperanza Manrique
- School of Medicine, Universidad Católica de Valencia, San Vicente Mártir, 46001 Valencia, Spain; Department of Dermatology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain
| | - David Millán-Esteban
- School of Medicine, Universidad Católica de Valencia, San Vicente Mártir, 46001 Valencia, Spain; Department of Dermatology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain
| | - Isabel Romero-Camarero
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4GJ, UK
| | - Fabio Amaral
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4GJ, UK
| | - Sarah Craig
- Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, the University of Manchester, Manchester, UK
| | - Pedro Durao
- Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, the University of Manchester, Manchester, UK
| | - Joanna Pozniak
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Laura Stennett
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College London, Guy's Campus, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London, UK
| | - Duncan Smith
- Proteomics, Cancer Research UK Manchester Institute, The University of Manchester, Manchester SK10 4TG, UK
| | - Garry Ashton
- Histology, Cancer Research UK Manchester Institute, The University of Manchester, Manchester SK10 4TG, UK
| | - Alex Baker
- Visualisation, Irradiation & Analysis, Cancer Research UK Manchester Institute, The University of Manchester, Manchester SK10 4TG, UK
| | - Kang Zeng
- Visualisation, Irradiation & Analysis, Cancer Research UK Manchester Institute, The University of Manchester, Manchester SK10 4TG, UK
| | - Gilbert Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College London, Guy's Campus, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London, UK
| | - Victoria Sanz-Moreno
- Cytoskeleton and Metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London SW3 6JB, UK; Centre for Tumour Microenvironment at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK
| | - Jair Marques
- CRUK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Albert Koulman
- Wellcome Trust - MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK; Medical Research Council Human Nutrition Research, Elsie Widdowson Laboratory, University of Cambridge, Fulbourn Road, Cambridge CB1 9NL, UK
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium; Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4GJ, UK
| | - Luisa Motta
- Department of Histopathology, Salford Royal NHS Foundation Trust, The University of Manchester, Manchester, UK
| | - Caroline Gaudy-Marqueste
- Aix Marseille University, APHM, CRCM Inserm U1068, CNRS U7258, CHU Timone, Dermatology and Skin Cancer Department, Marseille, France
| | - Eduardo Nagore
- School of Medicine, Universidad Católica de Valencia, San Vicente Mártir, 46001 Valencia, Spain; Department of Dermatology, Fundación Instituto Valenciano de Oncología, 46009 Valencia, Spain
| | - Amaya Virós
- Skin Cancer and Ageing Lab, Cancer Research UK Manchester Institute, the University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester, UK; Department of Dermatology, Salford Royal NHS Foundation Trust, The University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Beeghly GF, Pincus MI, Varshney RR, Giri DD, Falcone DJ, Rudolph MC, Antonyak MA, Iyengar NM, Fischbach C. Hypertrophic adipocytes increase extracellular vesicle-mediated lipid release and reprogram breast cancer cell metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645549. [PMID: 40236195 PMCID: PMC11996363 DOI: 10.1101/2025.03.28.645549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Obesity worsens cancer-specific survival and all-cause mortality for women diagnosed with breast cancer. Rich in adipose tissue, the breast exhibits increased adipocyte size in obesity, which correlates with poor prognosis. However, adipocyte size is highly heterogeneous as adipose tissue expands through both hyperplasia and hypertrophy; and adipocyte size can increase independently of weight gain. Despite these observations, the impact of adipocyte size on breast cancer cell behavior remains unclear due to insufficient approaches to isolate adipocytes based on size and maintain them in culture for mechanistic studies. Here, we develop strategies to culture size-sorted adipocytes from two mouse models of obesity and test their functional impact on tumor cell malignancy. We find that large adipocytes are transcriptionally distinct from small adipocytes and are enriched for gene sets related to adipose tissue dysfunction, including altered lipid processing. In coculture studies, large adipocytes promote lipid accumulation in breast cancer cells, and enhance their migration, proliferation, and aerobic metabolism in a manner dependent on fatty acid oxidation. These changes coincide with increased release of extracellular vesicles by large versus small adipocytes, which transfer lipid to recipient tumor cells. Moving forward, our findings suggest that adipocyte size could serve as a prognostic biomarker for women with breast cancer and help identify new therapeutic targets to advance clinical outcomes for these patients.
Collapse
|
3
|
Eduardo MB, Cottone G, McCloskey CW, Liu S, Palma FR, Zappia MP, Islam AB, Gao P, Setya J, Dennis S, Gao H, Zhang Q, Xuei X, Luo Y, Locasale J, Bonini MG, Khokha R, Frolov MV, Benevolenskaya EV, Chandel NS, Khan SA, Clare SE. A metabolic shift to the serine pathway induced by lipids fosters epigenetic reprogramming in nontransformed breast cells. SCIENCE ADVANCES 2025; 11:eads9182. [PMID: 40117373 PMCID: PMC11927636 DOI: 10.1126/sciadv.ads9182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025]
Abstract
Lipid metabolism and the serine, one-carbon, glycine (SOG) and methionine pathways are independently and significantly correlated with estrogen receptor-negative breast cancer (ERneg BC). Here, we propose a link between lipid metabolism and ERneg BC through phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in the de novo serine pathway. We demonstrate that the metabolism of the paradigmatic medium-chain fatty acid octanoic acid leads to a metabolic shift toward the SOG and methionine pathways. PHGDH plays a role in both the forward direction, contributing to the production of S-adenosylmethionine, and the reverse direction, generating the oncometabolite 2-hydroxyglutarate, leading to epigenomic reprogramming and phenotypic plasticity. The methionine cycle is closely linked to the transsulfuration pathway. Consequently, we observe that the shift increases the antioxidant glutathione, which mitigates reactive oxygen species (ROS), enabling survival of a subset of cells that have undergone DNA damage. These metabolic changes contribute to several hallmarks of cancer.
Collapse
Affiliation(s)
| | - Gannon Cottone
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Curtis W. McCloskey
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Shiyu Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Flavio R. Palma
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maria Paula Zappia
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Abul B.M.M.K. Islam
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joel Setya
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Saya Dennis
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Hongyu Gao
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qian Zhang
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaoling Xuei
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Jason Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Marcelo G. Bonini
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Maxim V. Frolov
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Elizaveta V. Benevolenskaya
- Department of Medicine/Division of Hematology Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Navdeep S. Chandel
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Seema A. Khan
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| | - Susan E. Clare
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
4
|
Guo Y, Shen K, Ge S, Zheng Q, Gao J, He X. Exploring the potential causal relationship between fatty acid metabolism ratios and major salivary gland carcinomas: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41802. [PMID: 40101084 PMCID: PMC11922437 DOI: 10.1097/md.0000000000041802] [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: 03/20/2025] Open
Abstract
Major salivary gland carcinomas (MSGCs) is a rare but aggressive cancer, with limited understanding of its metabolic underpinnings. Lipid metabolism, particularly fatty acid metabolism ratios (FAMRs), has been implicated in various cancers, but its role in MSGCs remains unclear. This study aims to explore the potential causal relationships between specific FAMRs and MSGCs using Mendelian randomization (MR) analysis. A 2-sample MR analysis was conducted using summary data from genome-wide association studies. Three FAMRs, including the ratio of diacylglycerol to triglycerides (DAG/TG), total cholesterol (TC) to total lipids (TL) ratio in large very low-density lipoprotein (VLDL; TC/TL in large VLDL), and triglycerides to total lipids ratio in medium VLDL (TG/TL in medium VLDL), were investigated for their potential causal relationships with MSGCs. Sensitivity analyses, including MR-Egger and leave-one-out tests, were performed to assess pleiotropy and the robustness of the results. The DAG/TG and TC/TL ratios in large VLDL were significantly positively associated with an increased risk of MSGCs (OR = 10.921, P = .004 and OR = 2.651, P = .047, respectively). In contrast, the TG/TL ratio in medium VLDL showed a significant negative association with MSGCs risk (OR = 0.460, P = .041). Sensitivity analyses confirmed the robustness of these associations, with no evidence of significant pleiotropy in 2 of the ratios. This study reveals novel insights into the metabolic basis of MSGCs, demonstrating significant associations between specific FAMRs and MSGCs risk. These findings highlight the potential clinical relevance of FAMRs as biomarkers or therapeutic targets in MSGCs. Future studies should focus on diverse populations and mechanistic research to validate these associations and explore their clinical implications.
Collapse
Affiliation(s)
- Yongjie Guo
- Department of Maxillofacial Surgery, Wei Fang People's Hospital, Wei Fang, China
| | - Kailin Shen
- Department of Intervention, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuqing Ge
- Department of Maxillofacial Surgery, Wei Fang People's Hospital, Wei Fang, China
| | - Qi Zheng
- School of Stomatology, Shandong Second Medical University, Wei Fang, China
| | - Jingchao Gao
- School of Stomatology, Shandong Second Medical University, Wei Fang, China
| | - Xiaopo He
- Department of Maxillofacial Surgery, Wei Fang People's Hospital, Wei Fang, China
| |
Collapse
|
5
|
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
|
6
|
Snoke DB, Atwood GS, Bellefleur ER, Stokes AM, Toth MJ. Body composition alterations in patients with lung cancer. Am J Physiol Cell Physiol 2025; 328:C872-C886. [PMID: 39887975 DOI: 10.1152/ajpcell.01048.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Most patients with lung cancer experience cancer cachexia (CC), a syndrome of skeletal muscle and adipose tissue wasting. Knowledge of body composition changes in patients is limited, however, because most studies have been cross-sectional, comparing patients with noncancer controls or patients with and without CC. Few studies, in contrast, have evaluated body composition in patients with lung cancer over time. This review examines our current understanding of longitudinal body composition changes in patients with lung cancer and identifies modifying factors contributing to variation in muscle and adipose tissue wasting, focusing on biological sex. We identified 32 studies conducting longitudinal measurements of body composition by computed tomography, bioelectrical impedance, dual X-ray absorptiometry, or total body nitrogen, with a total of n = 3,951 patients (35% female). All studies evaluated changes following diagnosis while patients were receiving treatment. Most studies reporting muscle-specific outcomes show decreased skeletal muscle mass, with more pronounced muscle wasting in males and male-enriched populations. In a small number of studies reporting muscle density, the majority show increased myosteatosis. Adiposity changes are less frequently reported, although wasting appears more prevalent in late-stage disease. Further studies are needed to define adipose changes along the lung cancer continuum. Our review emphasizes the need for balanced recruitment based on biological sex and sex-based analyses. In addition, consensus reporting of relevant patient data and outcomes in future studies will allow for meta-analysis and assist in the development of effective treatments for lung CC.
Collapse
Affiliation(s)
- Deena B Snoke
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States
| | - Gary S Atwood
- Dana Health Sciences Library, University of Vermont, Burlington, Vermont, United States
| | - Emma R Bellefleur
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States
| | - Alice M Stokes
- Dana Health Sciences Library, University of Vermont, Burlington, Vermont, United States
| | - Michael J Toth
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States
| |
Collapse
|
7
|
Bernhardt SM, House CD. Bisphenol A and DDT disrupt adipocyte function in the mammary gland: implications for breast cancer risk and progression. Front Oncol 2025; 15:1490898. [PMID: 40034592 PMCID: PMC11873108 DOI: 10.3389/fonc.2025.1490898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
As breast cancer incidence continues to rise worldwide, there is a pressing need to understand the environmental factors that contribute to its development. Obesogens, including Bisphenol A (BPA) and Dichlorodiphenyltrichloroethane (DDT), are highly prevalent in the environment, and have been associated with obesity and metabolic dysregulation. BPA and DDT, known to disrupt hormone signaling in breast epithelial cells, also promote adipogenesis, lipogenesis, and adipokine secretion in adipose tissue, directly contributing to the pathogenesis of obesity. While the adipose-rich mammary gland may be particularly vulnerable to environmental obesogens, there is a scarcity of research investigating obesogen-mediated changes in adipocytes that drive oncogenic transformation of breast epithelial cells. Here, we review the preclinical and clinical evidence linking BPA and DDT to impaired mammary gland development and breast cancer risk. We discuss how the obesogen-driven mechanisms that contribute to obesity, including changes in adipogenesis, lipogenesis, and adipokine secretion, could provide a pro-inflammatory, nutrient-rich environment that promotes activation of oncogenic pathways in breast epithelial cells. Understanding the role of obesogens in breast cancer risk and progression is essential for informing public health guidelines aimed at minimizing obesogen exposure, to ultimately reduce breast cancer incidence and improve outcomes for women.
Collapse
Affiliation(s)
- Sarah M. Bernhardt
- Department of Biology, San Diego State University, San Diego, CA, United States
| | - Carrie D. House
- Department of Biology, San Diego State University, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
8
|
He X, Guan XY, Li Y. Clinical significance of the tumor microenvironment on immune tolerance in gastric cancer. Front Immunol 2025; 16:1532605. [PMID: 40028336 PMCID: PMC11868122 DOI: 10.3389/fimmu.2025.1532605] [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: 11/22/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
In the realm of oncology, the tumor microenvironment (TME)-comprising extracellular matrix components, immune cells, fibroblasts, and endothelial cells-plays a pivotal role in tumorigenesis, progression, and response to therapeutic interventions. Initially, the TME exhibits tumor-suppressive properties that can inhibit malignant transformation. However, as the tumor progresses, various factors induce immune tolerance, resulting in TME behaving in a state that promotes tumor growth and metastasis in later stages. This state of immunosuppression is crucial as it enables TME to change from a role of killing tumor cells to a role of promoting tumor progression. Gastric cancer is a common malignant tumor of the gastrointestinal tract with an alarmingly high mortality rate. While chemotherapy has historically been the cornerstone of treatment, its efficacy in prolonging survival remains limited. The emergence of immunotherapy has opened new therapeutic pathways, yet the challenge of immune tolerance driven by the gastric cancer microenvironment complicates these efforts. This review aims to elucidate the intricate role of the TME in mediating immune tolerance in gastric cancer and to spotlight innovative strategies and clinical trials designed to enhance the efficacy of immunotherapeutic approaches. By providing a comprehensive theoretical framework, this review seeks to advance the understanding and application of immunotherapy in the treatment of gastric cancer, ultimately contributing to improved patient outcomes.
Collapse
Affiliation(s)
- Xiangyang He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Clinical Oncology, The University of Hongkong, Hong Kong, Hong Kong SAR, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
9
|
Wu P, Chen J, Li H, Lu H, Li Y, Zhang J. Interactions between ferroptosis and tumour development mechanisms: Implications for gynaecological cancer therapy (Review). Oncol Rep 2025; 53:18. [PMID: 39635847 PMCID: PMC11638741 DOI: 10.3892/or.2024.8851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Ferroptosis is a form of programmed cell death that is distinct from apoptosis. The mechanism involves redox‑active metallic iron and is characterized by an abnormal increase in iron‑dependent lipid reactive oxygen species, which results in high levels of membrane lipid peroxides. The relationship between ferroptosis and gynaecological tumours is complex. Ferroptosis can regulate tumour proliferation, metastasis and chemotherapy resistance, and targeting ferroptosis is a promising antitumour approach. Ferroptosis interacts with mechanisms related to tumorigenesis and development, such as macrophage polarization, the neutrophil trap network, mitochondrial autophagy and cuproptosis. The present review examines recent information on the interaction between the molecular mechanism of ferroptosis and other tumour‑related mechanisms, as well as the involvement of ferroptosis in gynaecological tumours, to identify implications for gynaecological cancer therapy.
Collapse
Affiliation(s)
- Peiting Wu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Jianlin Chen
- Department of Assisted Reproductive Centre, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Haiyuan Lu
- Department of Clinical Laboratory Department, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
- Department of Hunan Vigorzoe Biotechnology Co., Ltd., Hunan 417700, P.R. China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan 410013, P.R. China
| |
Collapse
|
10
|
Wang Y, Bo Y, Liu Y, Zhou J, Nguyen D, Baskaran D, Liu Y, Wang H. Metabolic labeling and targeted modulation of adipocytes. Biomater Sci 2025; 13:434-445. [PMID: 39648977 PMCID: PMC11758917 DOI: 10.1039/d4bm01352b] [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] [Indexed: 12/10/2024]
Abstract
Adipocytes play a critical role in energy storage and endocrine signaling and are associated with various diseases such as cancer and diabetes. Facile strategies to engineer adipocytes have long been pursued for elucidating adipocyte biology and developing adipocyte-based therapies. Herein, we report metabolic glycan labeling of adipocytes and subsequent targeted modulation of adipocytes via click chemistry. We show that azido tags expressed on the surface of adipocytes can persist for over 4 days. By conjugating dibenzocyclooctyne (DBCO)-cargos onto azido-labeled adipocytes via click chemistry, the cargos can be retained on the adipocyte membrane for over 12 hours. We further show that signaling molecules including adiponectin, calreticulin, mannose-binding lectin 2, and milk fat globule-EGF factor 8 protein can be conjugated to adipocytes to orchestrate their phagocytosis by macrophages. The azido-labeled adipocytes grafted into mice can also mediate targeted conjugation of DBCO-cargos in vivo. This adipocyte labeling and targeting technology will facilitate the development of adipocyte-based therapies and provides a new platform for manipulating the interaction between adipocytes and other types of cells.
Collapse
Affiliation(s)
- Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Daniel Nguyen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Dhyanesh Baskaran
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yuan Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
11
|
Torabinejad S, Miro C, Nappi A, Del Giudice F, Cicatiello AG, Sagliocchi S, Acampora L, Restolfer F, Murolo M, Di Cicco E, Capone F, Imbimbo C, Dentice M, Crocetto F. Obesity alters the fitness of peritumoral adipose tissue, exacerbating tumor invasiveness in renal cancer through the induction of ADAM12 and CYP1B1. Mol Oncol 2025. [PMID: 39806854 DOI: 10.1002/1878-0261.13782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity exacerbates the risk and aggressiveness of many types of cancer. Adipose tissue (AT) represents a prevalent component of the tumor microenvironment (TME) and contributes to cancer development and progression. Reciprocal communication between cancer and adipose cells leads to the generation of cancer-associated adipocytes (CAAs), which in turn foster tumor invasiveness by producing paracrine metabolites, adipocytokines, and growth factors. Interfering with the crosstalk between CAAs and cancer cells is of key relevance in the prevention of tumor progression. The present study aimed to analyze the contribution of peritumoral AT in renal cell carcinoma (RCC) progression in lean versus overweight or obese patients. By isolating human adipose-derived stromal/stem cells from the three groups of patients and performing conditioned medium studies with RCC cells along with in vivo xenograft experiments, we found that peritumoral adipocytes from the three groups show a distinct expression profile of genes. In particular, ADAM metallopeptidase domain 12 (ADAM12) and cytochrome P450 family 1 subfamily B member 1 (CYP1B1) were found to be upregulated in obesity and their silencing reduced RCC cell invasiveness. In conclusion, high ADAM12 and CYP1B1 expressions in the peritumoral adipocytes boost tumor invasiveness and may serve as an indicator of poor prognosis in RCC.
Collapse
Affiliation(s)
- Sepehr Torabinejad
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Francesco Del Giudice
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | | | - Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Emery Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Federico Capone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| |
Collapse
|
12
|
Li Y, Ming R, Zhang T, Gao Z, Wang L, Yang Y, Shen K, Wei C, Zhu Y, Li J, Zheng S, Luo Z, Ding Y, Xuan J, Hu Q, Yang Y, Gu J, Wei C. TCTN1 Induces Fatty Acid Oxidation to Promote Melanoma Metastasis. Cancer Res 2025; 85:84-100. [PMID: 39325960 DOI: 10.1158/0008-5472.can-24-0158] [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: 01/15/2024] [Revised: 06/12/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
Metabolic reprogramming promotes and sustains multiple steps of melanoma metastasis. Identification of key regulators of metabolic reprogramming could lead to the development of treatments for preventing and treating metastatic melanoma. In this study, we identified that tectonic family member 1 (TCTN1) promotes melanoma metastasis by increasing fatty acid oxidation (FAO). In clinical melanoma samples, high expression of TCTN1 correlated with increased metastasis and shorter patient survival. Functionally, TCTN1 promoted melanoma invasion and migration in vitro and distant metastasis in vivo and induced a mesenchymal-like phenotype switch. Mechanistically, TCTN1 acted as a protein scaffold to promote the binding of HADHA and HADHB, subunits of the mitochondrial trifunctional protein complex, thus leading to FAO activation. TCTN1-mediated FAO activated the p38/MAPK signaling pathway in melanoma cells, promoting tumor epithelial-mesenchymal transition and stemness. Molecular docking indicated that the prostaglandin F receptor agonist fluprostenol can block HADHA/HADHB binding, which was confirmed experimentally. Treatment with fluprostenol was able to inhibit TCTN1-induced melanoma invasion and metastasis. Taken together, these findings elucidate the mechanism of TCTN1-mediated promotion of melanoma metastasis and support the potential application of fluprostenol for targeted therapy of metastatic melanoma. Significance: TCTN1 activates fatty acid oxidation to induce melanoma mesenchymal phenotype switching and invasion by promoting the binding of the subunits of MTP, which can be targeted with fluprostenol to inhibit melanoma metastasis.
Collapse
Affiliation(s)
- Yinlam Li
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Ren Ming
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Tianyi Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Zixu Gao
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yang Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Kangjie Shen
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Chenlu Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yu Zhu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Jianrui Li
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Shaoluan Zheng
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Shanghai, P. R. China
| | - Zucheng Luo
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yiteng Ding
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Jiangying Xuan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Qianrong Hu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Yanwen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Jianying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| | - Chuanyuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
13
|
Jiang Z, Tabuchi C, Gayer SG, Bapat SP. Immune Dysregulation in Obesity. ANNUAL REVIEW OF PATHOLOGY 2025; 20:483-509. [PMID: 39854190 DOI: 10.1146/annurev-pathmechdis-051222-015350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
The immune system plays fundamental roles in maintaining physiological homeostasis. With the increasing prevalence of obesity-a state characterized by chronic inflammation and systemic dyshomeostasis-there is growing scientific and clinical interest in understanding how obesity reshapes immune function. In this review, we propose that obesity is not merely an altered metabolic state but also a fundamentally altered immunological state. We summarize key seminal and recent findings that elucidate how obesity influences immune function, spanning its classical role in microbial defense, its contribution to maladaptive inflammatory diseases such as asthma, and its impact on antitumor immunity. We also explore how obesity modulates immune function within tissue parenchyma, with a particular focus on the role of T cells in adipose tissue. Finally, we consider areas for future research, including investigation of the durable aspects of obesity on immunological function even after weight loss, such as those observed with glucagon-like peptide-1 (GLP-1) receptor agonist treatment. Altogether, this review emphasizes the critical role of systemic metabolism in shaping immune cell functions, with profound implications for tissue homeostasis across various physiological contexts.
Collapse
Affiliation(s)
- Zewen Jiang
- Diabetes Center and Department of Laboratory Medicine, University of California, San Francisco, California, USA;
| | - Chihiro Tabuchi
- Diabetes Center and Department of Laboratory Medicine, University of California, San Francisco, California, USA;
| | - Sarah G Gayer
- Diabetes Center and Department of Laboratory Medicine, University of California, San Francisco, California, USA;
| | - Sagar P Bapat
- Diabetes Center and Department of Laboratory Medicine, University of California, San Francisco, California, USA;
| |
Collapse
|
14
|
Gu J, Zhang H, Qian K, Ye X, Wu G. Who is Focusing on Women's Health: A Bibliometric Analysis of Global Research Trends on Overweight in Female Patients with Gynecologic or Breast Cancer. Int J Womens Health 2024; 16:2157-2171. [PMID: 39713093 PMCID: PMC11662632 DOI: 10.2147/ijwh.s496718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Objective Obesity has become a significant public health concern, strongly linked to various diseases, particularly gynecologic and breast cancers. This bibliometric review aims to analyze global research trends on overweight women, particularly those with gynecologic and breast cancers, to identify research hotspots, key contributors, and emerging areas of study. Methods A comprehensive bibliometric analysis was conducted using the Web of Science Core Collection (WoSCC) database, covering the period from January 2013 to September 2024. Articles were screened and analyzed using tools such as VOSviewer and Biblioshiny platform, with metrics including publication volume, citation analysis, and co-authorship networks. Key areas of focus were global research trends, leading countries, institutions, authors, journals, and keyword analysis. Results A total of 1452 publications were analyzed. Research activity on the association between obesity and gynecologic/breast cancer has steadily increased, with the United States leading in publications and citations, followed by China and Italy. Core journals included Breast Cancer Research and Treatment and Gynecologic Oncology. Key research areas identified through keyword analysis include the relationship between body mass index (BMI) and cancer risk, survival rates in cancer patients, physical activity, and the role of adipose tissue inflammation in tumor progression. Emerging topics include extracellular vesicles and cancer-associated fibroblasts. Conclusion Global research on the relationship between obesity and female-specific cancers has shown significant growth. The findings highlight BMI, survival, and physical activity as central themes. Future research should explore the molecular mechanisms linking obesity to cancer and evaluate weight loss interventions for cancer prevention and treatment.
Collapse
Affiliation(s)
- Jialin Gu
- Department of TCM, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Hongqun Zhang
- Hunan Engineering Research Center of TCM Real-World Clinical Practice, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, Hunan, 418000, People’s Republic of China
| | - Kang Qian
- Department of TCM, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, People’s Republic of China
| | - Xunwen Ye
- Department of Oncology, Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, Zhejiang, 315500, People’s Republic of China
| | - Guolin Wu
- Department of TCM, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, People’s Republic of China
| |
Collapse
|
15
|
Nayak A, Panda SS, Dwivedi I, Meena S, Aich P. Role of gut microbial-derived metabolites and other select agents on adipocyte browning. Biochem Biophys Res Commun 2024; 737:150518. [PMID: 39142136 DOI: 10.1016/j.bbrc.2024.150518] [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: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
AIMS Metabolic disease is a multifaceted condition characterized by the disruption of numerous metabolic parameters within the host. Its prevalence has surged significantly in recent years and it has become a prominent non-communicable disease worldwide. The effect of gut microbiota on various beige fat induction is well studied, while the mechanisms behind the link remain unclear. Given that gut microbiota-derived metabolites (meta-metabolites) secreted in the gut serve as a key mode of communication with their host through direct circulation or indirect host physiology modification, understanding the effect of meta-metabolites on adipose tissue is essential. METHODOLOGY In our previous in-vivo studies, we observed a correlation between gut microbiota and the formation of beige fat. In this study, we further aimed to validate this correlation by treating the adipocyte cell line (3T3-L1) with meta-metabolites collected from the cecum of mice exhibiting beige adipose tissue formation. Additionally, we treated the adipocyte cell line with known beige fat inducers (L-Rhamnose and Ginsenoside) to assess meta-metabolites' efficacy on beige fat formation. KEY FINDINGS Upon treatment with the meta-metabolites from the antibiotic-treated mice, we observed a significant increase in lipid metabolism and beige-specific gene expression. Analyzing the metabolites in these cells revealed that a set of metabolites potentially govern adipocytes, contributing to a metabolically active state. These effects were at par or even better than those of cells treated with L-Rhamnose or Ginsenoside. SIGNIFICANCE This research sheds light on the intricate interplay between microbial metabolites and adipose tissue, offering valuable clues for understanding and potentially manipulating these processes for therapeutic purposes.
Collapse
Affiliation(s)
- Akankshya Nayak
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Jatni, 752050, India; Homi Bhabha National Institute, Training School Complex, Mumbai, 400094, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), Jatni, 752050, India
| | - Swati Sagarika Panda
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Jatni, 752050, India; Homi Bhabha National Institute, Training School Complex, Mumbai, 400094, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), Jatni, 752050, India
| | - Isha Dwivedi
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Jatni, 752050, India; Homi Bhabha National Institute, Training School Complex, Mumbai, 400094, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), Jatni, 752050, India
| | - Shivani Meena
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Jatni, 752050, India; Homi Bhabha National Institute, Training School Complex, Mumbai, 400094, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), Jatni, 752050, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Jatni, 752050, India; Homi Bhabha National Institute, Training School Complex, Mumbai, 400094, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), Jatni, 752050, India.
| |
Collapse
|
16
|
Kisar Tunca S, Unal R. Adipocyte-derived fatty acid uptake induces obesity-related breast cancer progression: a review. Mol Biol Rep 2024; 52:39. [PMID: 39644365 DOI: 10.1007/s11033-024-10139-x] [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] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Obesity is a metabolic disorder that occurs when excess energy taken into the body is stored as fat. It is known that this metabolic imbalance affects the development of other diseases such as cancer, cardiovascular diseases, insulin resistance, and diabetes. The main cellular component of adipose tissue is adipocytes, and the environmental interactions of adipocytes are important to study the mechanism of disorder formation. Breast tissue is rich in adipose tissue and obesity is known to be an important risk factor in the development of breast cancer. Altered adipogenesis and lipogenesis processes in adipocytes in breast tissue support tumor development through the transfer of fatty acids released from adipocytes. We believe that blending adipocyte biology with breast cancer development is important for investigating the mechanisms that regulate breast tumor malignant behavior and providing new targets for treatment. Fatty acids, which are an energy source for breast cancer cells, are discussed from molecular perspectives in this review.
Collapse
Affiliation(s)
- Selin Kisar Tunca
- Faculty of Science, Department of Molecular Biology and Genetics, Mugla Sitki Kocman University, Mugla, Turkey
| | - Resat Unal
- Faculty of Science, Department of Molecular Biology and Genetics, Mugla Sitki Kocman University, Mugla, Turkey.
| |
Collapse
|
17
|
Chang Y, Du R, Xia F, Xu X, Wang H, Chen X. Dysregulation of Fatty Acid Metabolism in Breast Cancer and Its Targeted Therapy. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:825-844. [PMID: 39628960 PMCID: PMC11614585 DOI: 10.2147/bctt.s496322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/13/2024] [Indexed: 12/06/2024]
Abstract
Breast cancer has become the number one cancer worldwide, there are challenges in its prevention, diagnosis and treatment, especially the pathogenesis of triple negative breast cancer has not been clear and the treatment dilemma of metastatic breast cancer. Metabolic reprogramming is currently considered to be one of the hallmarks of cancer, and metabolic alterations in breast cancer, including enhanced glycolysis, tricarboxylic acid cycle activity, glutamine catabolism, and fatty acid biosynthesis, are manifested differently in different breast cancer subtypes and have a complex relationship with tumor growth, metastasis, death, and drug resistance. At present, inhibitors of fatty acid synthesis and oxidation related enzymes have a certain effect in the treatment of breast cancer. In this paper, we review the studies on fatty acid metabolism in breast cancer to better understand the mechanism of fatty acid metabolism in breast cancer pathogenesis and hope to provide new ideas for targeting fatty acid metabolism in the treatment of breast cancer.
Collapse
Affiliation(s)
- Yue Chang
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Rui Du
- Department of Anorectal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei First People’s Hospital, Hefei, Anhui, People’s Republic of China
| | - Fan Xia
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Xiuli Xu
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Hongzhi Wang
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Xueran Chen
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| |
Collapse
|
18
|
Miracle CE, McCallister CL, Egleton RD, Salisbury TB. Mechanisms by which obesity regulates inflammation and anti-tumor immunity in cancer. Biochem Biophys Res Commun 2024; 733:150437. [PMID: 39074412 PMCID: PMC11455618 DOI: 10.1016/j.bbrc.2024.150437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Obesity is associated with an increased risk for 13 different cancers. The increased risk for cancer in obesity is mediated by obesity-associated changes in the immune system. Obesity has distinct effects on different types of inflammation that are tied to tumorigenesis. For example, obesity promotes chronic inflammation in adipose tissue that is tumor-promoting in peripheral tissues. Conversely, obesity inhibits acute inflammation that rejects tumors. Obesity therefore promotes cancer by differentially regulating chronic versus acute inflammation. Given that obesity is chronic, the initial inflammation in adipose tissue will lead to systemic inflammation that could induce compensatory anti-inflammatory reactions in peripheral tissues to suppress chronic inflammation. The overall effect of obesity in peripheral tissues is therefore dependent on the duration and severity of obesity. Adipose tissue is a complex tissue that is composed of many cell types in addition to adipocytes. Further, adipose tissue cellularity is different at different anatomical sites throughout the body. Consequently, the sensitivity of adipose tissue to obesity is dependent on the anatomical location of the adipose depot. For example, obesity induces more inflammation in visceral than subcutaneous adipose tissue. Based on these studies, the mechanisms by which obesity promotes tumorigenesis are multifactorial and immune cell type-specific. The objective of our paper is to discuss the cellular mechanisms by which obesity promotes tumorigenesis by regulating distinct types of inflammation in adipose tissue and the tumor microenvironment.
Collapse
Affiliation(s)
- Cora E Miracle
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Chelsea L McCallister
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Richard D Egleton
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| | - Travis B Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1 John Marshall Drive, Huntington, WV, 25755, USA.
| |
Collapse
|
19
|
Tovar-Hernández K, Salinas-Vera YM, Carlos-Reyes Á, García-Hernández AP, Marchat LA, Mandujano-Lázaro G, Ríos-Castro E, Velasco-Suárez A, Mendez-Gómez I, Tecalco-Cruz ÁC, Ibarra-Sierra E, López-Camarillo C. Adipocytes reprogram the proteome of breast cancer cells in organotypic three-dimensional cell cultures. Sci Rep 2024; 14:27029. [PMID: 39505903 PMCID: PMC11542085 DOI: 10.1038/s41598-024-76053-1] [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: 03/02/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
While epidemiological evidence has long linked obesity with an increased risk of breast cancer, the intricate interactions between adipocytes and cancer cells within the tumor microenvironment remain largely uncharted territory. The use of organotypic three-dimensional (3D) cell cultures that more accurately mimic the spatial architecture of tumors represents an innovative approach to this complex issue. In the present study, we investigated the effects of adipocytes on the proteome of Hs578t breast cancer cells cultured in a 3D microenvironment. Using different treatments, we rigorously optimized the experimental conditions to induce the optimal differentiation of 3T3-L1 fibroblasts into mature adipocytes. Then, we grow the Hs578t cells in a simulated microenvironment using an on-top model for organotypic 3D cultures. Our data showed that cancer cells formed 3D stellate-like architectures when grown over an extracellular matrix proteins-enriched scaffold for 48 h. Proteomic profiling using LC-MS/MS mass spectrometry of Hs578t cells grown in 3D conditions with or without the adipocyte-enriched culture discovered 916 unique proteins. Of these, 605 showed no significant changes in abundance, whereas 87 proteins were significantly upregulated and 224 downregulated after interaction with fat cells (p < 0.05, FC > 2.0). Bioinformatic analysis of upregulated proteins indicated that the most enriched GO terms and molecular functions were related to lipids transport, cell differentiation, hypoxia response, and cell junctions. In addition, several modulated proteins have been previously associated with breast cancer progression. Interestingly, lipid transport proteins, including PITPNM2, ATP2C1, ABCA12, HDLBP, and APOB, showed perturbations in their expression, which were also associated with low overall survival in breast cancer patients. Functional studies showed that the knockdown of apolipoprotein B (APOB) expression in Hs578t cells reduced the size of 3D cellular structures. Moreover, APOB-knocked cells cocultured with adipocytes for 48 h exhibited a significant decrease of intracellular lipids, whereas an increase in the adipocytes was found. Our results indicate that the 3D microenvironment and the adipocytes crosstalk reprogram the proteome of breast cancer cells. These data help us understand the environmental effects in gene expression and contribute to discovering novel tumor proteins with potential intervention in breast cancer therapy.
Collapse
Affiliation(s)
- Karla Tovar-Hernández
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México
| | - Yarely M Salinas-Vera
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México
| | - Ángeles Carlos-Reyes
- Laboratorio de Oncoinmunobiologia, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Ciudad de México, México.
- , Calzada de Tlalpan 4502, Belisario Domínguez Sección 16, Tlalpan, Ciudad de México, 14080, México.
| | | | - Laurence A Marchat
- Laboratorio de Biomedicina Molecular II. Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de México, México
| | - Gilberto Mandujano-Lázaro
- Laboratorio de Biomedicina Molecular II. Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de México, México
| | - Emmanuel Ríos-Castro
- Unidad de Genómica, Proteómica y Metabolómica. CINVESTAV-IPN, Ciudad de México, México
| | - Andrea Velasco-Suárez
- Unidad de Genómica, Proteómica y Metabolómica. CINVESTAV-IPN, Ciudad de México, México
| | - Ivonne Mendez-Gómez
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México
| | - Ángeles C Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México
| | - Eloisa Ibarra-Sierra
- Departamento de Investigación, Instituto Estatal de Cancerologia "Dr. Arturo Beltrán Ortega", Acapulco, Guerrero, México
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México.
- San Lorenzo 290, Colonia del Valle, Ciudad de México, 03100, México.
| |
Collapse
|
20
|
Solsona-Vilarrasa E, Vousden KH. Obesity, white adipose tissue and cancer. FEBS J 2024. [PMID: 39496581 DOI: 10.1111/febs.17312] [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: 04/20/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
White adipose tissue (WAT) is crucial for whole-body energy homeostasis and plays an important role in metabolic and hormonal regulation. While healthy WAT undergoes controlled expansion and contraction to meet the body's requirements, dysfunctional WAT in conditions like obesity is characterized by excessive tissue expansion, alterations in lipid homeostasis, inflammation, hypoxia, and fibrosis. Obesity is strongly associated with an increased risk of numerous cancers, with obesity-induced WAT dysfunction influencing cancer development through various mechanisms involving both systemic and local interactions between adipose tissue and tumors. Unhealthy obese WAT affects circulating levels of free fatty acids and factors like leptin, adiponectin, and insulin, altering systemic lipid metabolism and inducing inflammation that supports tumor growth. Similar mechanisms are observed locally in an adipose-rich tumor microenvironment (TME), where WAT cells can also trigger extracellular matrix remodeling, thereby enhancing the TME's ability to promote tumor growth. Moreover, tumors reciprocally interact with WAT, creating a bidirectional communication that further enhances tumorigenesis. This review focuses on the complex interplay between obesity, WAT dysfunction, and primary tumor growth, highlighting potential targets for therapeutic intervention.
Collapse
|
21
|
Ambrosio MR, Adriaens M, Derks K, Migliaccio T, Costa V, Liguoro D, Cataldi S, D'Esposito V, Maneli G, Bassolino R, Di Paola S, Pirozzi M, Schonauer F, D'Andrea F, Beguinot F, Arts I, Formisano P. Glucose impacts onto the reciprocal reprogramming between mammary adipocytes and cancer cells. Sci Rep 2024; 14:24674. [PMID: 39433816 PMCID: PMC11494089 DOI: 10.1038/s41598-024-76522-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
An established hallmark of cancer cells is metabolic reprogramming, largely consisting in the exacerbated glucose uptake. Adipocytes in the tumor microenvironment contribute toward breast cancer (BC) progression and are highly responsive to metabolic fluctuations. Metabolic conditions characterizing obesity and/or diabetes associate with increased BC incidence and mortality. To explore BC-adipocytes interaction and define the impact of glucose in such dialogue, Mammary Adipose-derived Mesenchymal Stem Cells (MAd-MSCs) were differentiated into adipocytes and co-cultured with ER+ BC cells while exposed to glucose concentration resembling hyperglycemia or normoglycemia in humans (25mM or 5.5mM). The transcriptome of both cell types in co-culture as in mono-culture was profiled by RNA-Seq to define the impact of adipocytes on BC cells and viceversa (i), the action of glucose on BC cells, adipocytes (ii) and their crosstalk (iii). Noteworthy, we provided evidence that co-culture with adipocytes in a glucose-rich environment determined a re-program of BC cell transcriptome driving lipid accumulation, a hallmark of BC aggressiveness, promoting stem-like properties and reducing Tamoxifen responsiveness. Moreover, our data point out to a transcriptional effect through which BC cells induce adipocytes de-lipidation, paralleled by pluripotency gain, as source of lipids when glucose lowering occurs. Thus, modulating plasticity of peri-tumoral adipocytes may represent a key point for halting BC progression in metabolically unbalanced patients.
Collapse
Affiliation(s)
- Maria Rosaria Ambrosio
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy.
| | - Michiel Adriaens
- Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - Kasper Derks
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Teresa Migliaccio
- Department of Translational Medicine (DiSMeT), University of Naples "Federico II", Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics "A. Buzzati Traverso", National Council of Research (IGB-CNR), Naples, Italy
| | - Domenico Liguoro
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy
| | - Simona Cataldi
- Institute of Genetics and Biophysics "A. Buzzati Traverso", National Council of Research (IGB-CNR), Naples, Italy
| | - Vittoria D'Esposito
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy
| | - Giovanni Maneli
- Department of Translational Medicine (DiSMeT), University of Naples "Federico II", Naples, Italy
| | - Rita Bassolino
- Department of Translational Medicine (DiSMeT), University of Naples "Federico II", Naples, Italy
| | - Simone Di Paola
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy
| | - Marinella Pirozzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy
| | - Fabrizio Schonauer
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Francesco D'Andrea
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Francesco Beguinot
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy
- Department of Translational Medicine (DiSMeT), University of Naples "Federico II", Naples, Italy
| | - Ilja Arts
- Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, The Netherlands
| | - Pietro Formisano
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research (IEOS-CNR), Naples, Italy.
- Department of Translational Medicine (DiSMeT), University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
22
|
Wolf M, Brochhausen C, Ramakrishnan V, Iberl S, Roth J, Seitz S, Burkhardt R, Stadler SC. Histologic Characterization of Tumor-Adjacent Mammary Adipose Tissue in Normal-Weight and Overweight/Obese Patients with Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3515. [PMID: 39456610 PMCID: PMC11506523 DOI: 10.3390/cancers16203515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Obesity is a risk factor of several types of cancer, including breast cancer. In this study, we aimed to histologically characterize the adipose tissue of the tumor microenvironment (TME) of triple-negative breast cancer (TNBC) in overweight/obese versus normal-weight patients. Methods: TNBC tissue sections from normal-weight (BMI<25) and overweight/obese patients (BMI≥25) were stained with antibodies against CD68, CD163, CD31, CD34, and vimentin. At the invasive tumor front, positive cells were counted in tumor adjacent adipose tissue (AT) and within cancer tissue (CT). Further, the size of the tumor-adjacent and distant mammary adipocytes was determined in perilipin stained sections. Expression of ANGPTL4, CD36 and FABP4, proteins involved in fatty acid metabolism, was analyzed in marginal tumor cells using an immune reactive score. Results: Overweight/obese TNBC patients had significantly larger adipocytes, higher numbers of CD163+ macrophages (BMI<25: 2.80 vs. BMI≥25: 10.45; p = 0.011) and lower numbers of CD31+ (BMI<25: 4.20 vs. BMI≥25: 2.40; p = 0.018) and CD34+ (BMI<25: 14.60 vs. BMI≥25: 5.20; p = 0.045) cells as markers of angiogenesis in the AT as well as a higher frequency of cancer-associated-fibroblast-like cells in the AT and CT (BMI<25: 7.60 vs. BMI≥25: 25.39 in total; p = 0.001). Moreover, expression of CD36 (BMI<25: 2.15 vs. BMI≥25: 2.60; p = 0.041) and ANGPTL4 (BMI<25: 6.00 vs. BMI≥25: 9.80; p = 0.026) was elevated in the TNBC cells of overweight/obese patients. Conclusions: Our data suggest BMI-related changes in the TME of overweight/obese TNBC patients, including hypertrophied adipocytes, reduced vascularization, more M2-like macrophages and CAF-like cells, and an increase in the expression of fatty acid metabolizing proteins in marginal tumor cells, all contributing to a more tumor-promoting, immunosuppressive environment.
Collapse
Affiliation(s)
- Marietta Wolf
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, University Heidelberg, 69120 Mannheim, Germany
- Institute of Pathology, Regensburg University, 93053 Regensburg, Germany
| | | | - Sabine Iberl
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| | - Jonas Roth
- Department of Gynecology and Obstetrics, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Centre Regensburg, 93053 Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| | - Sonja C. Stadler
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany (R.B.)
| |
Collapse
|
23
|
Miracle CE, McCallister CL, Denning KL, Russell R, Allen J, Lawrence L, Legenza M, Krutzler-Berry D, Salisbury TB. High BMI Is Associated with Changes in Peritumor Breast Adipose Tissue That Increase the Invasive Activity of Triple-Negative Breast Cancer Cells. Int J Mol Sci 2024; 25:10592. [PMID: 39408921 PMCID: PMC11476838 DOI: 10.3390/ijms251910592] [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: 08/28/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Breast cancer is the most common cancer in women with multiple risk factors including smoking, genetics, environmental factors, and obesity. Smoking and obesity are the top two risk factors for the development of breast cancer. The effect of obesity on adipose tissue mediates the pathogenesis of breast cancer in the context of obesity. Triple-negative breast cancer (TNBC) is a breast cancer subtype within which the cells lack estrogen, progesterone, and HER2 receptors. TNBC is the deadliest breast cancer subtype. The 5-year survival rates for patients with TNBC are 8-16% lower than the 5-year survival rates for patients with estrogen-receptor-positive breast tumors. In addition, TNBC patients have early relapse rates (3-5 years after diagnosis). Obesity is associated with an increased risk for TNBC, larger TNBC tumors, and increased breast cancer metastasis compared with lean women. Thus, novel therapeutic approaches are warranted to treat TNBC in the context of obesity. In this paper, we show that peritumor breast adipose-derived secretome (ADS) from patients with a high (>30) BMI is a stronger inducer of TNBC cell invasiveness and JAG1 expression than peritumor breast ADS from patients with low (<30) BMI. These findings indicate that patient BMI-associated changes in peritumor AT induce changes in peritumor ADS, which in turn acts on TNBC cells to stimulate JAG1 expression and cancer cell invasiveness.
Collapse
Affiliation(s)
- Cora E. Miracle
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (C.E.M.); (C.L.M.)
| | - Chelsea L. McCallister
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (C.E.M.); (C.L.M.)
| | - Krista L. Denning
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Rebecca Russell
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Jennifer Allen
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Logan Lawrence
- Cabell Huntington Hospital Laboratory, Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (K.L.D.); (R.R.); (J.A.); (L.L.)
| | - Mary Legenza
- Edwards Comprehensive Cancer Center, Department of Oncology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (M.L.); (D.K.-B.)
| | - Diane Krutzler-Berry
- Edwards Comprehensive Cancer Center, Department of Oncology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (M.L.); (D.K.-B.)
| | - Travis B. Salisbury
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (C.E.M.); (C.L.M.)
| |
Collapse
|
24
|
Mo YY, Han YX, Xu SN, Jiang HL, Wu HX, Cai JM, Li L, Bu YH, Xiao F, Liang HD, Wen Y, Liu YZ, Yin YL, Zhou HD. Adipose Tissue Plasticity: A Comprehensive Definition and Multidimensional Insight. Biomolecules 2024; 14:1223. [PMID: 39456156 PMCID: PMC11505740 DOI: 10.3390/biom14101223] [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: 08/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Adipose tissue is composed of adipocytes, stromal vascular fraction, nerves, surrounding immune cells, and the extracellular matrix. Under various physiological or pathological conditions, adipose tissue shifts cellular composition, lipid storage, and organelle dynamics to respond to the stress; this remodeling is called "adipose tissue plasticity". Adipose tissue plasticity includes changes in the size, species, number, lipid storage capacity, and differentiation function of adipocytes, as well as alterations in the distribution and cellular composition of adipose tissue. This plasticity has a major role in growth, obesity, organismal protection, and internal environmental homeostasis. Moreover, certain thresholds exist for this plasticity with significant individualized differences. Here, we comprehensively elaborate on the specific connotation of adipose tissue plasticity and the relationship between this plasticity and the development of many diseases. Meanwhile, we summarize possible strategies for treating obesity in response to adipose tissue plasticity, intending to provide new insights into the dynamic changes in adipose tissue and contribute new ideas to relevant clinical problems.
Collapse
Affiliation(s)
- Yu-Yao Mo
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Xin Han
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Shi-Na Xu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Jun-Min Cai
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yan-Hong Bu
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha 410012, China;
| | - Fen Xiao
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Han-Dan Liang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Ying Wen
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| | - Yu-Ze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
| | - Yu-Long Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.-Y.M.); (Y.-X.H.); (S.-N.X.); (H.-L.J.); (H.-X.W.); (J.-M.C.); (L.L.); (F.X.); (H.-D.L.); (Y.W.)
| |
Collapse
|
25
|
Yum C, Andolino C, Larrick B, Sheeley MP, Teegarden D. 1α,25-Dihydroxyvitamin D Downregulates Adipocyte Impact on Breast Cancer Cell Migration and Adipokine Release. Nutrients 2024; 16:3153. [PMID: 39339753 PMCID: PMC11434957 DOI: 10.3390/nu16183153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES Excess adiposity is associated with a higher risk of breast cancer metastasis and mortality. Evidence suggests that dietary vitamin D inhibits breast cancer metastasis. However, the mechanistic link between vitamin D's regulation of adipocyte metabolism and metastasis has not been previously investigated. Therefore, the purpose of these experiments was to examine the effect of the active form of vitamin D, 1α,25-dihydroxyvitamin D (1,25(OH)2D), on adipocyte release of bioactive compounds and whether the impact on adipocytes leads to inhibition of breast cancer cell migration, an important step of metastasis. METHODS Differentiated 3T3-L1 adipocytes were treated with 1,25(OH)2D for two days, followed by either harvesting the adipocytes or collecting adipocyte-conditioned media without 1,25(OH)2D. A transwell migration assay was conducted with vehicle- or 1,25(OH)2D-conditioned media. In order to explore the mechanism underlying effects on breast cancer metastatic capability, the mRNA expression of leptin, adiponectin, insulin-like growth factor (IGF-1), interleukin-6 (IL-6), and monocyte chemoattractant protein-1 (MCP-1) was measured in adipocytes following either vehicle or 1,25(OH)2D treatment. RESULTS Conditioned media from 1,25(OH)2D-treated adipocytes inhibited the migration of metastatic MDA-MB-231 breast cancer cells compared to conditioned media from vehicle-treated adipocytes. Treatment of adipocytes with 1,25(OH)2D decreased mRNA expression of leptin, adiponectin, IGF-1, IL-6, and MCP-1. Consistent with mRNA expression, concentrations of leptin, adiponectin, IGF-1, and IL-6 in adipocyte-conditioned media were decreased with 1,25(OH)2D treatment, although MCP-1 remained unchanged. CONCLUSIONS In summary, these results suggest that 1,25(OH)2D alters adipocyte secretions to prevent breast cancer metastasis.
Collapse
Affiliation(s)
- Chaehyun Yum
- Interdepartmental Nutrition Program, Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Chaylen Andolino
- Interdepartmental Nutrition Program, Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Brienna Larrick
- Interdepartmental Nutrition Program, Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Madeline P Sheeley
- Interdepartmental Nutrition Program, Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Dorothy Teegarden
- Interdepartmental Nutrition Program, Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
- Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
26
|
Shen T, Sun S, Li W, Wang X, Gao Y, Yang Q, Cai J. Association between body mass index and lymph node metastasis among women with cervical cancer: a systematic review and network meta-analysis. Arch Gynecol Obstet 2024; 310:1289-1301. [PMID: 38858322 DOI: 10.1007/s00404-024-07528-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/22/2024] [Indexed: 06/12/2024]
Abstract
PURPOSE Lymph node status is a determinant of survival in patients with early-stage cervical cancer. However, the relationship between obesity and lymph node status remains unclear. Therefore, this systematic review aims to evaluate the correlation between body mass index (BMI) and lymph node metastasis in cervical cancer. METHODS Cohort studies through six databases were reviewed until December 2021. Odds ratios (ORs) for lymphatic metastasis were estimated using random-effects models and network meta-analysis. BMI groups for lymph node metastasis were ranked. Heterogeneities were assessed using I2. Subgroup analyses were performed to determine possible sources of heterogeneity. RESULTS No significant difference was found between obese (BMI ≥ 25) and non-obese patients (BMI < 25) (OR = 1.01; 95% CI 0.69-1.47; P = 0.97). In subgroup analyses, obesity was associated with higher risk among the Americans and advanced-stage patients. The grouping analysis based on BMI and the rankogram values revealed that the '35 ≤ BMI' group had the highest risk of lymph node metastasis. CONCLUSION Although there were no significant differences in lymph node metastasis between obese and non-obese cervical cancer patients in overall analysis, patients with BMI ≥ 35 were at significantly higher risk of lymph node metastasis.
Collapse
Affiliation(s)
- Tiantian Shen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenhan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoman Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumei Gao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiang Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
27
|
Zakic T, Pekovic-Vaughan V, Cvoro A, Korac A, Jankovic A, Korac B. Redox and metabolic reprogramming in breast cancer and cancer-associated adipose tissue. FEBS Lett 2024; 598:2106-2134. [PMID: 38140817 DOI: 10.1002/1873-3468.14794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Redox and metabolic processes are tightly coupled in both physiological and pathological conditions. In cancer, their integration occurs at multiple levels and is characterized by synchronized reprogramming both in the tumor tissue and its specific but heterogeneous microenvironment. In breast cancer, the principal microenvironment is the cancer-associated adipose tissue (CAAT). Understanding how the redox-metabolic reprogramming becomes coordinated in human breast cancer is imperative both for cancer prevention and for the establishment of new therapeutic approaches. This review aims to provide an overview of the current knowledge of the redox profiles and regulation of intermediary metabolism in breast cancer while considering the tumor and CAAT of breast cancer as a unique Warburg's pseudo-organ. As cancer is now recognized as a systemic metabolic disease, we have paid particular attention to the cell-specific redox-metabolic reprogramming and the roles of estrogen receptors and circadian rhythms, as well as their crosstalk in the development, growth, progression, and prognosis of breast cancer.
Collapse
Affiliation(s)
- Tamara Zakic
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Vanja Pekovic-Vaughan
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, William Henry Duncan Building, University of Liverpool, UK
| | | | | | - Aleksandra Jankovic
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Bato Korac
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
- Faculty of Biology, University of Belgrade, Serbia
| |
Collapse
|
28
|
Lin Z, Hua G, Hu X. Lipid metabolism associated crosstalk: the bidirectional interaction between cancer cells and immune/stromal cells within the tumor microenvironment for prognostic insight. Cancer Cell Int 2024; 24:295. [PMID: 39174964 PMCID: PMC11342506 DOI: 10.1186/s12935-024-03481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Cancer is closely related to lipid metabolism, with the tumor microenvironment (TME) containing numerous lipid metabolic interactions. Cancer cells can bidirectionally interact with immune and stromal cells, the major components of the TME. This interaction is primarily mediated by fatty acids (FAs), cholesterol, and phospholipids. These interactions can lead to various physiological changes, including immune suppression, cancer cell proliferation, dissemination, and anti-apoptotic effects on cancer cells. The physiological modulation resulting from this lipid metabolism-associated crosstalk between cancer cells and immune/stromal cells provides valuable insights into cancer prognosis. A comprehensive literature review was conducted to examine the function of the bidirectional lipid metabolism interactions between cancer cells and immune/stromal cells within the TME, particularly how these interactions influence cancer prognosis. A novel autophagy-extracellular vesicle (EV) pathway has been proposed as a mediator of lipid metabolism interactions between cancer cells and immune cells/stromal cells, impacting cancer prognosis. As a result, different forms of lipid metabolism interactions have been described as being linked to cancer prognosis, including those mediated by the autophagy-EV pathway. In conclusion, understanding the bidirectional lipid metabolism interactions between cancer cells and stromal/immune cells in the TME can help develop more advanced prognostic approaches for cancer patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Guanxiang Hua
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Xiaojuan Hu
- Queen Mary College, Nanchang University, Nanchang, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
29
|
Chen X, Zhang H, Fang Z, Wang D, Song Y, Zhang Q, Hou J, Yang S, Xu D, Fei Y, Zhang W, Zhang J, Tang J, Li L. Adipocytes promote metastasis of breast cancer by attenuating the FOXO1 effects and regulating copper homeostasis. Cancer Cell Int 2024; 24:284. [PMID: 39135158 PMCID: PMC11320833 DOI: 10.1186/s12935-024-03433-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Obesity and the forkhead box O1(FOXO1) affect the survival of breast cancer patients, but the underlying mechanism remains unclear. We aimed to investigate the role of FOXO1 in obesity-associated-breast cancer. METHODS We screened 383 breast disease patients from the first affiliated hospital with Nanjing Medical University in 2020. We performed wound healing, transwell, matrigel assays to assess the metastatic ability of cancer cells. We adopted mRNAs sequencing to select the differentially expressed transcripts in breast cancer. We applied immunohistochemistry, western blot, tissue microarrays to assess the level of FOXO1 and epithelial-mesenchymal transition (EMT) pathways. We conducted bioinformatic analysis to investigate interactions between FOXO1 and miR-135b. We used fluorescence in situ hybridization, RT-qPCR to confirm the characteristics of circCNIH4. We conducted luciferase reporter assay, rescue experiments to investigate interactions between circCNIH4 and miR-135b. RESULTS Obesity was positively correlated with the incidence and progression of breast cancer. Adipocytes enhanced the migration of breast cancer and attenuated the effects of FOXO1. MiR-135b was a binding gene of FOXO1 and was regulated by circCNIH4. CircCNIH4 exhibited antitumor activity in vitro and in vivo. CONCLUSION Adipocytes might accelerate the progression of breast cancer by modulating FOXO1/miR-135b/ circCNIH4 /EMT axis and regulating copper homeostasis.
Collapse
Affiliation(s)
- Xiu Chen
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Heda Zhang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zheng Fang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Dandan Wang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yuxin Song
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Junchen Hou
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Sujin Yang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Di Xu
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yinjiao Fei
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wei Zhang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Zhang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Lei Li
- Department of General Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
30
|
Cui X, Cao C, Li X, Lin B, Yan A, Yang Y. Succinylation of 14-3-3 theta by CPT1A promotes survival and paclitaxel resistance in nasal type extranodal natural killer/T-cell lymphoma. Transl Oncol 2024; 46:102006. [PMID: 38823259 PMCID: PMC11176827 DOI: 10.1016/j.tranon.2024.102006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND The aggressive and refractory extranodal natural killer/T-cell lymphoma, nasal type (ENKTL-NT) is a subtype of non-Hodgkin's lymphoma. Succinylation promotes progression in a variety of tumors, but its mechanism in ENKTL-NT is unclear. METHODS Bioinformatic analysis was performed to screen differentially expressed genes in the ENKTL dataset. Cell transfection techniques were used for knockdown and overexpression of genes. The mRNA and protein expression were detected using RT-qPCR and western blot, respectively. Immunohistochemical staining was used to assess protein expression in situ. For the detection of cell proliferation activity, CCK-8, clonal formation, and EDU staining assays were used. Flow cytometry was employed to detect apoptosis. Co-immunoprecipitation was utilized for the identification of protein interactions and succinylation modifications. RESULTS Succinyltransferase CPT1A was highly elevated in ENKTL-NT and was associated with a dismal prognosis. CPT1A knockdown suppressed SNK-6 cells' proliferation and induced apoptosis, while these effects were reversed by the overexpression of 14-3-3theta. Co-immunoprecipitation results showed that CPT1A caused succinylation of 14-3-3theta at site of K85, thereby enhancing the protein stability. Suppression of CPT1A-induced succinylation of 14-3-3theta by ST1326 resulted in the inhibition of SNK-6 cell proliferation and increased apoptosis. Paclitaxel combined with knockdown of CPT1A significantly inhibited the proliferation of ENKTL-NT compared to paclitaxel alone. CONCLUSION CPT1A induces succinylation of 14-3-3theta at the K85 site, promoting ENKTL-NT proliferation. The anti-ENKTL activity of paclitaxel was improved when combined with CPT1A knockdown.
Collapse
Affiliation(s)
- Xiao Cui
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang 110003, China
| | - Chengcheng Cao
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xinyang Li
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Biyan Lin
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang 110003, China
| | - Aihui Yan
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang 110003, China.
| | - Ying Yang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| |
Collapse
|
31
|
Cai Q, Yang J, Shen H, Xu W. Cancer-associated adipocytes in the ovarian cancer microenvironment. Am J Cancer Res 2024; 14:3259-3279. [PMID: 39113876 PMCID: PMC11301307 DOI: 10.62347/xzri9189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
The tumor microenvironment (TME) plays a critical role in high energy metabolism during tumorigenesis, progression and metastasis. Among them, adipocytes, as an important component of the TME, can transform into cancer-associated adipocytes (CAAs) through dedifferentiation via interactions with tumor cells. These CAAs provide nutrients, growth factors, cytokines and metabolites to the tumor and later transdifferentiate into other stromal cells at a later stage to alter tumor growth, metastasis and the drug response and ultimately influence the treatment and prognosis of ovarian cancer. This review outlines the physiological functions of CAAs and discusses the progress in the use of CAAs as therapeutic targets in ovarian cancer.
Collapse
Affiliation(s)
- Qiuling Cai
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Jing Yang
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Huiling Shen
- Department of Oncology, The First Affiliated People’s Hospital of Jiangsu UniversityZhenjiang, Jiangsu, China
| | - Wenlin Xu
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu UniversityZhenjiang, Jiangsu, China
| |
Collapse
|
32
|
Habanjar O, Nehme R, Goncalves-Mendes N, Cueff G, Blavignac C, Aoun J, Decombat C, Auxenfans C, Diab-Assaf M, Caldefie-Chézet F, Delort L. The obese inflammatory microenvironment may promote breast DCIS progression. Front Immunol 2024; 15:1384354. [PMID: 39072314 PMCID: PMC11272476 DOI: 10.3389/fimmu.2024.1384354] [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: 02/09/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Ductal carcinoma in situ (DCIS), characterized by a proliferation of neoplastic cells confined within the mammary ducts, is distinctly isolated from the surrounding stroma by an almost uninterrupted layer of myoepithelial cells (MECs) and by the basement membrane. Heightened interactions within the adipose microenvironment, particularly in obese patients, may play a key role in the transition from DCIS to invasive ductal carcinoma (IDC), which is attracting growing interest in scientific research. Adipose tissue undergoes metabolic changes in obesity, impacting adipokine secretion and promoting chronic inflammation. This study aimed to assess the interactions between DCIS, including in situ cancer cells and MECs, and the various components of its inflammatory adipose microenvironment (adipocytes and macrophages). Methods To this end, a 3D co-culture model was developed using bicellular bi-fluorescent DCIS-like tumoroids, adipose cells, and macrophages to investigate the influence of the inflammatory adipose microenvironment on DCIS progression. Results The 3D co-culture model demonstrated an inhibition of the expression of genes involved in apoptosis (BAX, BAG1, BCL2, CASP3, CASP8, and CASP9), and an increase in genes related to cell survival (TP53, JUN, and TGFB1), inflammation (TNF-α, PTGS2, IL-6R), invasion and metastasis (TIMP1 and MMP-9) in cancer cells of the tumoroids under inflammatory conditions versus a non-inflammatory microenvironment. On the contrary, it confirmed the compromised functionality of MECs, resulting in the loss of their protective effects against cancer cells. Adipocytes from obese women showed a significant increase in the expression of all studied myofibroblast-associated genes (myoCAFs), such as FAP and α-SMA. In contrast, adipocytes from normal-weight women expressed markers of inflammatory fibroblast phenotypes (iCAF) characterized by a significant increase in the expression of LIF and inflammatory cytokines such as TNF-α, IL-1β, IL-8, and CXCL-10. These changes also influenced macrophage polarization, leading to a pro-inflammatory M1 phenotype. In contrast, myoCAF-associated adipocytes, and the cancer-promoting microenvironment polarized macrophages towards an M2 phenotype, characterized by high CD163 receptor expression and IL-10 and TGF-β secretion. Discussion Reciprocal interactions between the tumoroid and its microenvironment, particularly in obesity, led to transcriptomic changes in adipocytes and macrophages, may participate in breast cancer progression while disrupting the integrity of the MEC layer. These results underlined the importance of adipose tissue in cancer progression.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Rawan Nehme
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Gwendal Cueff
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Christelle Blavignac
- Université Clermont-Auvergne, Centre d’Imagerie Cellulaire Santé (CCIS), Clermont-Ferrand, France
| | - Jessy Aoun
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | | | - Céline Auxenfans
- Banque de tissus et de cellules, Hôpital Edouard-Herriot, Lyon, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Moléculaire et Pharmacologie Anticancéreuse, Faculté des Sciences II, Université libanaise Fanar, Beirut, Lebanon
| | | | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Clermont-Ferrand, France
| |
Collapse
|
33
|
Pliszka M, Szablewski L. Associations between Diabetes Mellitus and Selected Cancers. Int J Mol Sci 2024; 25:7476. [PMID: 39000583 PMCID: PMC11242587 DOI: 10.3390/ijms25137476] [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: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the major causes of mortality and is the second leading cause of death. Diabetes mellitus is a serious and growing problem worldwide, and its prevalence continues to grow; it is the 12th leading cause of death. An association between diabetes mellitus and cancer has been suggested for more than 100 years. Diabetes is a common disease diagnosed among patients with cancer, and evidence indicates that approximately 8-18% of patients with cancer have diabetes, with investigations suggesting an association between diabetes and some particular cancers, increasing the risk for developing cancers such as pancreatic, liver, colon, breast, stomach, and a few others. Breast and colorectal cancers have increased from 20% to 30% and there is a 97% increased risk of intrahepatic cholangiocarcinoma or endometrial cancer. On the other hand, a number of cancers and cancer therapies increase the risk of diabetes mellitus. Complications due to diabetes in patients with cancer may influence the choice of cancer therapy. Unfortunately, the mechanisms of the associations between diabetes mellitus and cancer are still unknown. The aim of this review is to summarize the association of diabetes mellitus with selected cancers and update the evidence on the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Monika Pliszka
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
34
|
Park WY, Song G, Park JY, Jung SJ, Kim S, Ahn KS, Choe SK, Kwak HJ, Park J, Um JY. Epigallocatechin gallate protects against fat and muscle atrophy in B16BL6 melanoma-bearing mice on a high-fat diet. Life Sci 2024; 348:122677. [PMID: 38702026 DOI: 10.1016/j.lfs.2024.122677] [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: 04/12/2023] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
AIMS Epidemiological evidence indicates that there is a substantial association between body mass index (BMI) and at least ten forms of cancer, including melanoma, and BMI imbalance contributes to the poor survival rate of cancer patients before and after therapy. Nevertheless, few pharmacological studies on models of obesity and cancer have been reported. In this study, we administered epigallocatechin gallate (EGCG) to B16BL6 tumor-bearing mice that received a high-fat diet (HFD) to examine its impact. METHODS B16BL6 tumor-bearing mice were fed a HFD. Body weight and food intake were documented every week. We conducted a Western blot analysis to examine the protein levels in the tumor, gastrocnemius (GAS), and tibialis anterior (TA) muscles, as well as the inguinal and epididymal white adipose tissues (iWAT and eWAT). KEY FINDINGS EGCG has been shown to have anti-cancer effects equivalent to those of cisplatin, a chemotherapy drug. Furthermore, EGCG protected against the loss of epidydimal white adipose tissue by regulating protein levels of lipolysis factors of adipose triglyceride lipase and hormone-sensitive lipase as well as WAT browning factors of uncoupling protein 1, as opposed to cisplatin. EGCG was shown to reduce the protein levels of muscular atrophy factors of muscle RING-finger protein-1, whereas cisplatin did not contribute to rescuing the atrophy of TA and GAS muscles. CONCLUSION Taken together, our findings indicate that EGCG has a preventive effect against cachexia symptoms and has anti-cancer effects similar to those of cisplatin in tumor-bearing mice fed a high-fat diet.
Collapse
Affiliation(s)
- Woo Yong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gahee Song
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja Yeon Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Se Jin Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seokwoo Kim
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae-Young Um
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
35
|
Marlin S, Goepp M, Desiderio A, Rougé S, Aldekwer S, Le Guennec D, Goncalves-Mendes N, Talvas J, Farges MC, Rossary A. Long-Term High-Fat Diet Limits the Protective Effect of Spontaneous Physical Activity on Mammary Carcinogenesis. Int J Mol Sci 2024; 25:6221. [PMID: 38892407 PMCID: PMC11172547 DOI: 10.3390/ijms25116221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancer is influenced by factors such as diet, a sedentary lifestyle, obesity, and postmenopausal status, which are all linked to prolonged hormonal and inflammatory exposure. Physical activity offers protection against breast cancer by modulating hormones, immune responses, and oxidative defenses. This study aimed to assess how a prolonged high-fat diet (HFD) affects the effectiveness of physical activity in preventing and managing mammary tumorigenesis. Ovariectomised C57BL/6 mice were provided with an enriched environment to induce spontaneous physical activity while being fed HFD. After 44 days (short-term, ST HFD) or 88 days (long-term, LT HFD), syngenic EO771 cells were implanted into mammary glands, and tumour growth was monitored until sacrifice. Despite similar physical activity and food intake, the LT HFD group exhibited higher visceral adipose tissue mass and reduced skeletal muscle mass. In the tumour microenvironment, the LT HFD group showed decreased NK cells and TCD8+ cells, with a trend toward increased T regulatory cells, leading to a collapse of the T8/Treg ratio. Additionally, the LT HFD group displayed decreased tumour triglyceride content and altered enzyme activities indicative of oxidative stress. Prolonged exposure to HFD was associated with tumour growth despite elevated physical activity, promoting a tolerogenic tumour microenvironment. Future studies should explore inter-organ exchanges between tumour and tissues.
Collapse
MESH Headings
- Animals
- Diet, High-Fat/adverse effects
- Female
- Mice
- Mice, Inbred C57BL
- Physical Conditioning, Animal
- Tumor Microenvironment
- Oxidative Stress
- Carcinogenesis
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/prevention & control
- Cell Line, Tumor
- Mammary Neoplasms, Animal/pathology
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/prevention & control
- Intra-Abdominal Fat/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
Collapse
Affiliation(s)
- Sébastien Marlin
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Marie Goepp
- Resolution Therapeutics, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Adrien Desiderio
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Stéphanie Rougé
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Sahar Aldekwer
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Delphine Le Guennec
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Nicolas Goncalves-Mendes
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Jérémie Talvas
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Marie-Chantal Farges
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| | - Adrien Rossary
- UNH—Unité de Nutrition Humaine, CRNH-Auvergne, Université Clermont-Auvergne, INRAe, F-63000 Clermont-Ferrand, France; (S.M.); (A.D.); (S.R.); (S.A.); (D.L.G.); (N.G.-M.); (J.T.)
| |
Collapse
|
36
|
Chen X, Tian J, Zhao C, Wu Y, Li J, Ji Z, Lian D, Jia Z, Chen X, Zhou Z, Zhu B, Hua Z. Resveratrol, a novel inhibitor of fatty acid binding protein 5, inhibits cervical cancer metastasis by suppressing fatty acid transport into nucleus and downstream pathways. Br J Pharmacol 2024; 181:1614-1634. [PMID: 38158217 DOI: 10.1111/bph.16308] [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: 07/27/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Because of cervical cancer (CC) metastasis, the prognosis of diagnosed patients is poor. However, the molecular mechanisms and therapeutic approach for metastatic CC remain elusive. EXPERIMENTAL APPROACH In this study, we first evaluated the effect of resveratrol (RSV) on CC cell migration and metastasis. Via an activity-based protein profiling (ABPP) approach, a photoaffinity probe of RSV (RSV-P) was synthesized, and the protein targets of RSV in HeLa cells were identified. Based on target information and subsequent in vivo and in vitro validation experiments, we finally elucidated the mechanism of RSV corresponding to its antimetastatic activity. KEY RESULTS The results showed that RSV concentration-dependently suppressed CC cell migration and metastasis. A list of proteins was identified as the targets of RSV, through the ABPP approach with RSV-P, among which fatty acid binding protein 5 (FABP5) attracted our attention based on The Cancer Genome Atlas (TCGA) database analysis. Subsequent knockout and overexpression experiments confirmed that RSV directly interacted with FABP5 to inhibit fatty acid transport into the nucleus, thereby suppressing downstream matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9) expression, thus inhibiting CC metastasis. CONCLUSIONS AND IMPLICATIONS Our study confirmed the key role of FABP5 in CC metastasis and provided important target information for the design of therapeutic lead compounds for metastatic CC.
Collapse
Affiliation(s)
- Xiao Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jing Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chunyuan Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yanhui Wu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiahuang Li
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zehan Ji
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Danchen Lian
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhibo Jia
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Xingyu Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zixin Zhou
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Bo Zhu
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zichun Hua
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
37
|
Langer HT, Rohm M, Goncalves MD, Sylow L. AMPK as a mediator of tissue preservation: time for a shift in dogma? Nat Rev Endocrinol 2024:10.1038/s41574-024-00992-y. [PMID: 38760482 DOI: 10.1038/s41574-024-00992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Ground-breaking discoveries have established 5'-AMP-activated protein kinase (AMPK) as a central sensor of metabolic stress in cells and tissues. AMPK is activated through cellular starvation, exercise and drugs by either directly or indirectly affecting the intracellular AMP (or ADP) to ATP ratio. In turn, AMPK regulates multiple processes of cell metabolism, such as the maintenance of cellular ATP levels, via the regulation of fatty acid oxidation, glucose uptake, glycolysis, autophagy, mitochondrial biogenesis and degradation, and insulin sensitivity. Moreover, AMPK inhibits anabolic processes, such as lipogenesis and protein synthesis. These findings support the notion that AMPK is a crucial regulator of cell catabolism. However, studies have revealed that AMPK's role in cell homeostasis might not be as unidirectional as originally thought. This Review explores emerging evidence for AMPK as a promoter of cell survival and an enhancer of anabolic capacity in skeletal muscle and adipose tissue during catabolic crises. We discuss AMPK-activating interventions for tissue preservation during tissue wasting in cancer-associated cachexia and explore the clinical potential of AMPK activation in wasting conditions. Overall, we provide arguments that call for a shift in the current dogma of AMPK as a mere regulator of cell catabolism, concluding that AMPK has an unexpected role in tissue preservation.
Collapse
Affiliation(s)
- Henning Tim Langer
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riβ, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus DaSilva Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lykke Sylow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Song Y, Na H, Lee SE, Kim YM, Moon J, Nam TW, Ji Y, Jin Y, Park JH, Cho SC, Lee J, Hwang D, Ha SJ, Park HW, Kim JB, Lee HW. Dysfunctional adipocytes promote tumor progression through YAP/TAZ-dependent cancer-associated adipocyte transformation. Nat Commun 2024; 15:4052. [PMID: 38744820 PMCID: PMC11094189 DOI: 10.1038/s41467-024-48179-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity has emerged as a prominent risk factor for the development of malignant tumors. However, the existing literature on the role of adipocytes in the tumor microenvironment (TME) to elucidate the correlation between obesity and cancer remains insufficient. Here, we aim to investigate the formation of cancer-associated adipocytes (CAAs) and their contribution to tumor growth using mouse models harboring dysfunctional adipocytes. Specifically, we employ adipocyte-specific BECN1 KO (BaKO) mice, which exhibit lipodystrophy due to dysfunctional adipocytes. Our results reveal the activation of YAP/TAZ signaling in both CAAs and BECN1-deficient adipocytes, inducing adipocyte dedifferentiation and formation of a malignant TME. The additional deletion of YAP/TAZ from BaKO mice significantly restores the lipodystrophy and inflammatory phenotypes, leading to tumor regression. Furthermore, mice fed a high-fat diet (HFD) exhibit decreased BECN1 and increased YAP/TAZ expression in their adipose tissues. Treatment with the YAP/TAZ inhibitor, verteporfin, suppresses tumor progression in BaKO and HFD-fed mice, highlighting its efficacy against mice with metabolic dysregulation. Overall, our findings provide insights into the key mediators of CAA and their significance in developing a TME, thereby suggesting a viable approach targeting adipocyte homeostasis to suppress cancer growth.
Collapse
Affiliation(s)
- Yaechan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Heeju Na
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - You Min Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jihyun Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Tae Wook Nam
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yul Ji
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Hyung Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seok Chan Cho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaehoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Gemcro, Inc, Seoul, 03722, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Bum Kim
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Gemcro, Inc, Seoul, 03722, Republic of Korea.
| |
Collapse
|
39
|
Niu N, Shen X, Wang Z, Chen Y, Weng Y, Yu F, Tang Y, Lu P, Liu M, Wang L, Sun Y, Yang M, Shen B, Jin J, Lu Z, Jiang K, Shi Y, Xue J. Tumor cell-intrinsic epigenetic dysregulation shapes cancer-associated fibroblasts heterogeneity to metabolically support pancreatic cancer. Cancer Cell 2024; 42:869-884.e9. [PMID: 38579725 DOI: 10.1016/j.ccell.2024.03.005] [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: 08/15/2023] [Revised: 02/01/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
The tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) involves a significant accumulation of cancer-associated fibroblasts (CAFs) as part of the host response to tumor cells. The origins and functions of transcriptionally diverse CAF populations in PDAC remain poorly understood. Tumor cell-intrinsic genetic mutations and epigenetic dysregulation may reshape the TME; however, their impacts on CAF heterogeneity remain elusive. SETD2, a histone H3K36 trimethyl-transferase, functions as a tumor suppressor. Through single-cell RNA sequencing, we identify a lipid-laden CAF subpopulation marked by ABCA8a in Setd2-deficient pancreatic tumors. Our findings reveal that tumor-intrinsic SETD2 loss unleashes BMP2 signaling via ectopic gain of H3K27Ac, leading to CAFs differentiation toward lipid-rich phenotype. Lipid-laden CAFs then enhance tumor progression by providing lipids for mitochondrial oxidative phosphorylation via ABCA8a transporter. Together, our study links CAF heterogeneity to epigenetic dysregulation in tumor cells, highlighting a previously unappreciated metabolic interaction between CAFs and pancreatic tumor cells.
Collapse
Affiliation(s)
- Ningning Niu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuqing Shen
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yueyue Chen
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yawen Weng
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feier Yu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Tang
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Lu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingzhu Liu
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Jin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zipeng Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Yufeng Shi
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer, Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
40
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [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: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
41
|
Liu H, Liu Y, Zhou Y, Chen X, Pan S, Zhou Q, Ji H, Zhu X. TM7SF2-induced lipid reprogramming promotes cell proliferation and migration via CPT1A/Wnt/β-Catenin axis in cervical cancer cells. Cell Death Discov 2024; 10:207. [PMID: 38693136 PMCID: PMC11063194 DOI: 10.1038/s41420-024-01975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
Cervical cancer poses a serious threat to women's health globally. Our previous studies found that upregulation of TM7SF2, which works as an enzyme involved in the process of cholesterol biosynthesis expression, was highly correlated with cervical cancer. However, the mechanistic basis of TM7SF2 promoting cervical cancer progression via lipid metabolism remains poorly understood. Therefore, quantification of fatty acids and lipid droplets were performed in vitro and in vivo. The protein-protein interaction was verified by Co-IP technique. The mechanism and underlying signaling pathway of TM7SF2 via CPT1A associated lipid metabolism in cervical cancer development were explored using Western blotting, IHC, colony formation, transwell assay, and wound healing assay. This study reported that overexpression of TM7SF2 increased fatty acids content and lipid droplets both in vivo and in vitro experiments. While knockout of TM7SF2 obviously attenuated this process. Moreover, TM7SF2 directly bonded with CPT1A, a key enzyme in fatty acid oxidation, and regulated CPT1A protein expression in cervical cancer cells. Notably, the proliferation and metastasis of cervical cancer cells were elevated when their CPT1A expression was upregulated. Then, rescue assay identified that CPT1A overexpressed could enhance the cell viability and migration in TM7SF2-knockout cells. Furthermore, depletion of TM7SF2 significantly inhibited WNT and β-catenin proteins expression, which was enhanced by CPT1A-overexpressed. The proliferation and migration of cervical cancer cells were reversed in CPT1A-overexpressed cells with the treatment of MSAB, an inhibitor of Wnt/β-Catenin pathway. This study put forward an idea that TM7SF2-induced lipid reprogramming promotes proliferation and migration via CPT1A/Wnt/β-Catenin axis in cervical cancer, underlying the progression of cervical cancer.
Collapse
Affiliation(s)
- Hejing Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yujia Zhou
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Chen
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingfeng Zhou
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Huihui Ji
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
42
|
Lefevre C, Thibaut MM, Loumaye A, Thissen JP, Neyrinck AM, Navez B, Delzenne NM, Feron O, Bindels LB. Tumoral acidosis promotes adipose tissue depletion by fostering adipocyte lipolysis. Mol Metab 2024; 83:101930. [PMID: 38570069 PMCID: PMC11027574 DOI: 10.1016/j.molmet.2024.101930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE Tumour progression drives profound alterations in host metabolism, such as adipose tissue depletion, an early event of cancer cachexia. As fatty acid consumption by cancer cells increases upon acidosis of the tumour microenvironment, we reasoned that fatty acids derived from distant adipose lipolysis may sustain tumour fatty acid craving, leading to the adipose tissue loss observed in cancer cachexia. METHODS To evaluate the pro-lipolytic capacities of acid-exposed cancer cells, primary mouse adipocytes from subcutaneous and visceral adipose tissue were exposed to pH-matched conditioned medium from human and murine acid-exposed cancer cells (pH 6.5), compared to naive cancer cells (pH 7.4). To further address the role of tumoral acidosis on adipose tissue loss, a pH-low insertion peptide was injected into tumour-bearing mice, and tumoral acidosis was neutralised with a sodium bicarbonate buffer. Prolipolytic mediators were identified by transcriptomic approaches and validated on murine and human adipocytes. RESULTS Here, we reveal that acid-exposed cancer cells promote lipolysis from subcutaneous and visceral adipocytes and that dampening acidosis in vivo inhibits adipose tissue depletion. We further found a set of well-known prolipolytic factors enhanced upon acidosis adaptation and unravelled a role for β-glucuronidase (GUSB) as a promising new actor in adipocyte lipolysis. CONCLUSIONS Tumoral acidosis promotes the mobilization of fatty acids derived from adipocytes via the release of soluble factors by cancer cells. Our work paves the way for therapeutic approaches aimed at tackling cachexia by targeting the tumour acidic compartment.
Collapse
Affiliation(s)
- Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Morgane M Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey Loumaye
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Jean-Paul Thissen
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Benoit Navez
- Department of Abdominal Surgery and Transplantation, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
43
|
Jiang M, Chen R, Hu B, Xiong S, Li S, Fu B, Liu X. FATP2 activates PI3K/Akt/mTOR pathway by inhibiting ATF3 and promotes the occurrence and development of bladder cancer. Cell Signal 2024; 117:111087. [PMID: 38316266 DOI: 10.1016/j.cellsig.2024.111087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/21/2024] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Bladder cancer (BLCA) is ranked among the main causes of mortality in male cancer patients, and research into targeted therapies guided by its genomics and molecular biology has been a prominent focus in BLCA studies. Fatty acid transporter protein 2 (FATP2), a member of the FATPs family,is a key contributor to the progression of cancers such as hepatocellular carcinomas and melanomas.However,its role in BLCA remains poorly understand. This study delved into the function of FATP2 in BLCA through a succession of experiments in vivo and in vitro, employing techniques as quantitative real-time polymerase chain reaction (qRT-PCR), RNA sequencing, transwell assays, immunofluorescence, western blot,and others to dissect its mechanistic actions. The findings revealed that an oncogenic function is executed by FATP2 in bladder cancer, significantly impacting the proliferation and migration capabilities, thereby affecting the prognosis of BLCA patients. Furthermore, A suppression that relies on both time and concentration of BLCA proliferation and migration, trigger of apoptosis, and blockage of the cell cycle at the G2/M phase were observed when the inhibitor of FATP2, Lipofermata, was applied. It was unveiled through subsequent investigations that ATF3 expression is indirectly promoted by Lipofermata through the inhibition of FATP2, ultimately inhibiting the signal transduction of the PI3K/Akt/mTOR pathway. This effect was also responsible for the inhibitory impact on BLCA proliferation. Therefore, FATP2 emerges as an auspicious and emerging molecular target with potential applications in precision therapy in BLCA.
Collapse
Affiliation(s)
- Ming Jiang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi,China; Department of Anesthesiology, Affiliated Sanming First Hospital of Fujian Medical Unerversity, Sanming, Fujian, China
| | - Ru Chen
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Bing Hu
- Department of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Situ Xiong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi,China
| | - Sheng Li
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi,China
| | - Bin Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi,China.
| | - Xiaoqiang Liu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi,China.
| |
Collapse
|
44
|
Taborda Ribas H, Sogayar MC, Dolga AM, Winnischofer SMB, Trombetta-Lima M. Lipid profile in breast cancer: From signaling pathways to treatment strategies. Biochimie 2024; 219:118-129. [PMID: 37993054 DOI: 10.1016/j.biochi.2023.11.008] [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: 08/25/2023] [Revised: 11/02/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023]
Abstract
Breast cancer is the most prevalent cancer in women. Metabolic abnormalities, particularly increased lipid synthesis and uptake, impact the onset and progression of the disease. However, the influence of lipid metabolism in breast cancer varies according to the disease stage and patient's hormone status. In postmenopausal patients, obesity is associated with a higher risk and poor prognosis of luminal tumors, while in premenopausal individuals, it is correlated to BRCA mutated tumors. In fact, the tumor's lipid profile may be used to distinguish between HER2+, luminal and BRCA-mutated tumors. Moreover, drug resistance was associated with increased fatty acid synthesis and alterations in membrane composition, impacting its fluidity and spatial subdomains such as lipid rafts. Here, we discuss the subtype-specific lipid metabolism alterations found in breast cancer and the potentiality of its modulation in a clinical setting.
Collapse
Affiliation(s)
- Hennrique Taborda Ribas
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, Netherlands; Postgraduate Program in Biochemistry Sciences, Sector of Biological Sciences, Federal University of Paraná, Curitiba, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy Center (NUCEL), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, Brazil
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, Netherlands
| | - Sheila M B Winnischofer
- Postgraduate Program in Biochemistry Sciences, Sector of Biological Sciences, Federal University of Paraná, Curitiba, Brazil; Biochemistry and Molecular Biology Department, Federal University of Paraná, Curitiba, Brazil; Postgraduate Program in Cellular and Molecular Biology, Biological Sciences Sector, UFPR, Curitiba, Brazil.
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Pharmaceutical Technology and Biopharmacy, Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, Netherlands.
| |
Collapse
|
45
|
Hagberg CE, Spalding KL. White adipocyte dysfunction and obesity-associated pathologies in humans. Nat Rev Mol Cell Biol 2024; 25:270-289. [PMID: 38086922 DOI: 10.1038/s41580-023-00680-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 02/10/2024]
Abstract
The prevalence of obesity and associated chronic diseases continues to increase worldwide, negatively impacting on societies and economies. Whereas the association between excess body weight and increased risk for developing a multitude of diseases is well established, the initiating mechanisms by which weight gain impairs our metabolic health remain surprisingly contested. In order to better address the myriad of disease states associated with obesity, it is essential to understand adipose tissue dysfunction and develop strategies for reinforcing adipocyte health. In this Review we outline the diverse physiological functions and pathological roles of human white adipocytes, examining our current knowledge of why white adipocytes are vital for systemic metabolic control, yet poorly adapted to our current obesogenic environment.
Collapse
Affiliation(s)
- Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
46
|
Grunt TW, Wagner R, Ries A, Berghoff AS, Preusser M, Grusch M, Valent P. Targeting endogenous fatty acid synthesis stimulates the migration of ovarian cancer cells to adipocytes and promotes the transport of fatty acids from adipocytes to cancer cells. Int J Oncol 2024; 64:24. [PMID: 38214315 PMCID: PMC10807641 DOI: 10.3892/ijo.2024.5612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024] Open
Abstract
Despite significant advances in oncology, 1 of 108 female patients succumb to ovarian cancer (OC) each year. Improved novel treatments against this aggressive disease would be a major improvement. The growth of OC cells has been demonstrated to be highly dependent on lipids. OC cells are abundantly present in the abdominal cavity and omentum, the main sites of OC expansion. Accordingly, it has been attempted not only to block the hyperactive synthesis of fatty acids (FAs) in cancer cells, but also to disrupt lipid supply. While either strategy has yielded promising results as monotherapy, the induction of resistance pathways diminishing the anticancer effects is yet conceivable. The endogenous regulation of lipid biosynthesis in OC has been extensively studied. However, the role of stromal cells in the modulation of the effects of anti‑lipogenic drugs has not yet been well documented. The present study thus examined the interaction between OC cells and associated stromal cells, when de novo FA synthesis was blocked. It has recently been revealed by the authors that when FA are provided to OC cells in monoculture, the lipid deficiency induced by pharmacological inhibition of FA synthase (FASN), the key enzyme of endogenous FA synthesis, cannot be compensated through an increased FA uptake by OC cells. In the present study, OC cells were co‑cultured with adipocytes preloaded with fluorescent FA and the effects of FASN‑inhibition on OC homing to adipocytes and the transcellular delivery of fluorescent FA from adipocytes to OC cells were examined. The FASN inhibitors, G28UCM and Fasnall, stimulated the spontaneous migration of A2780 OC cells in a concentration‑dependent manner and stimulated the transfer of FA from adipocytes to OC cells. Similar effects were observed with all types of adipocytes tested. The models applied in the present study demonstrated that co‑cultured cancer‑associated adipocytes may attenuate the anticancer effects of FASN inhibitors by attracting tumor cells and by supplying the cells with FA. This lipid‑mediated dependency may provide a rationale for the design of new treatment approaches for the treatment of OC.
Collapse
Affiliation(s)
- Thomas W. Grunt
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Renate Wagner
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
| | - Alexander Ries
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Anna S. Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, A-1090 Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Grusch
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Peter Valent
- Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, A-1090 Vienna, Austria
- Division of Hematology, Department of Medicine I, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
47
|
Mistry T, Nath A, Pal R, Ghosh S, Mahata S, Kumar Sahoo P, Sarkar S, Choudhury T, Nath P, Alam N, Nasare VD. Emerging Futuristic Targeted Therapeutics: A Comprising Study Towards a New Era for the Management of TNBC. Am J Clin Oncol 2024; 47:132-148. [PMID: 38145412 DOI: 10.1097/coc.0000000000001071] [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: 12/26/2023]
Abstract
Triple-negative breast cancer is characterized by high lethality attributed to factors such as chemoresistance, transcriptomic, and genomic heterogeneity, leading to a poor prognosis and limiting available targeted treatment options. While the identification of molecular targets remains pivotal for therapy involving chemo drugs, the current challenge lies in the poor response rates, low survival rates, and frequent relapses. Despite various clinical investigations exploring molecular targeted therapies in conjunction with conventional chemo treatment, the outcomes have been less than optimal. The critical need for more effective therapies underscores the urgency to discover potent novel treatments, including molecular and immune targets, as well as emerging strategies. This review provides a comprehensive analysis of conventional treatment approaches and explores emerging molecular and immune-targeted therapeutics, elucidating their mechanisms to address the existing obstacles for a more effective management of triple-negative breast cancer.
Collapse
Affiliation(s)
- Tanuma Mistry
- Departments of Pathology and Cancer Screening
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal
| | - Arijit Nath
- Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, School of Biotechnology, Bhubaneswar, Odisha, India
| | - Ranita Pal
- Departments of Pathology and Cancer Screening
| | | | | | | | | | | | | | - Neyaz Alam
- Surgical Oncology, Chittaranjan National Cancer Institute
| | | |
Collapse
|
48
|
Kim S, Oh J, Park C, Kim M, Jo W, Kim CS, Cho SW, Park J. FAM3C in Cancer-Associated Adipocytes Promotes Breast Cancer Cell Survival and Metastasis. Cancer Res 2024; 84:545-559. [PMID: 38117489 DOI: 10.1158/0008-5472.can-23-1641] [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: 06/02/2023] [Revised: 09/26/2023] [Accepted: 12/12/2023] [Indexed: 12/21/2023]
Abstract
Adipose tissue within the tumor microenvironment (TME) plays a critical role in supporting breast cancer progression. In this study, we identified FAM3 metabolism-regulating signaling molecule C (FAM3C) produced by cancer-associated adipocytes (CAA) as a key regulator of tumor progression. FAM3C overexpression in cultured adipocytes significantly reduced cell death in both adipocytes and cocultured breast cancer cells while suppressing markers of fibrosis. Conversely, FAM3C depletion in CAAs resulted in adipocyte-mesenchymal transition (AMT) and increased fibrosis within the TME. Adipocyte FAM3C expression was driven by TGFβ signaling from breast cancer cells and was reduced upon treatment with a TGFβ-neutralizing antibody. FAM3C knockdown in CAAs early in tumorigenesis in a genetically engineered mouse model of breast cancer significantly inhibited primary and metastatic tumor growth. Circulating FAM3C levels were elevated in patients with metastatic breast cancer compared with those with nonmetastatic breast cancer. These results suggest that therapeutic inhibition of FAM3C expression levels in CAAs during early tumor development could be a promising approach in the treatment of patients with breast cancer. SIGNIFICANCE High FAM3C levels in cancer-associated adipocytes contribute to tumor-supportive niches and are tightly associated with metastatic growth, indicating that FAM3C inhibition could be beneficial for treating patients with breast cancer.
Collapse
Affiliation(s)
- Sahee Kim
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jiyoung Oh
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chanho Park
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Min Kim
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Woobeen Jo
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chu-Sook Kim
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Sun Wook Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
49
|
Li S, Yuan J, Cheng Z, Li Y, Cheng S, Liu X, Huang S, Xu Z, Wu A, Liu L, Dong J. Hsa_circ_0021205 enhances lipolysis via regulating miR-195-5p/HSL axis and drives malignant progression of glioblastoma. Cell Death Discov 2024; 10:71. [PMID: 38341418 DOI: 10.1038/s41420-024-01841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Abnormal lipid metabolism is an essential hallmark of glioblastoma. Hormone sensitive lipase (HSL), an important rate-limiting enzyme contributed to lipolysis, which was involved in aberrant lipolysis of glioblastoma, however, its definite roles and the relevant regulatory pathway have not been fully elucidated. Our investigations disclosed high expression of HSL in glioblastoma. Knock-down of HSL restrained proliferation, migration, and invasion of glioblastoma cells while adding to FAs could significantly rescue the inhibitory effect of si-HSL on tumor cells. Overexpression of HSL further promoted tumor cell proliferation and invasion. Bioinformatics analysis and dual-luciferase reporter assay were performed to predict and verify the regulatory role of ncRNAs on HSL. Mechanistically, hsa_circ_0021205 regulated HSL expression by sponging miR-195-5p, which further promoted lipolysis and drove the malignant progression of glioblastoma. Besides, hsa_circ_0021205/miR-195-5p/HSL axis activated the epithelial-mesenchymal transition (EMT) signaling pathway. These findings suggested that hsa_circ_0021205 promoted tumorigenesis of glioblastoma through regulation of HSL, and targeting hsa_circ_0021205/miR-195-5p/HSL axis can serve as a promising new strategy against glioblastoma.
Collapse
Affiliation(s)
- Suwen Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaqi Yuan
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, the Zhangjiagang Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Zhe Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, the Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yongdong Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shan Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinglei Liu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shilu Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhipeng Xu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Anyi Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Dong
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
50
|
Chocarro-Calvo A, Jociles-Ortega M, García-Martinez JM, Louphrasitthiphol P, Garcia YV, Ramírez-Sánchez A, Chauhan J, Fiuza MC, Duran M, García-Jiménez C, Goding CR. Phenotype-specific melanoma uptake of fatty acid from human adipocytes activates AXL and CAV1-dependent β-catenin nuclear accumulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.21.576568. [PMID: 38328032 PMCID: PMC10849526 DOI: 10.1101/2024.01.21.576568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Phenotypic diversity of cancer cells within tumors generated through bi-directional interactions with the tumor microenvironment has emerged as a major driver of disease progression and therapy resistance. Nutrient availability plays a critical role in determining phenotype, but whether specific nutrients elicit different responses on distinct phenotypes is poorly understood. Here we show, using melanoma as a model, that only MITF Low undifferentiated cells, but not MITF High cells, are competent to drive lipolysis in human adipocytes. In contrast to MITF High melanomas, adipocyte-derived free fatty acids are taken up by undifferentiated MITF Low cells via a fatty acid transporter (FATP)-independent mechanism. Importantly, oleic acid (OA), a monounsaturated long chain fatty acid abundant in adipose tissue and lymph, reprograms MITF Low undifferentiated melanoma cells to a highly invasive state by ligand-independent activation of AXL, a receptor tyrosine kinase associated with therapy resistance in a wide range of cancers. AXL activation by OA then drives SRC-dependent formation and nuclear translocation of a β-catenin-CAV1 complex. The results highlight how a specific nutritional input drives phenotype-specific activation of a pro-metastasis program with implications for FATP-targeted therapies.
Collapse
|