1
|
Bessot A, Gunter J, McGovern J, Bock N. Bone marrow adipocytes in cancer: Mechanisms, models, and therapeutic implications. Biomaterials 2025; 322:123341. [PMID: 40315628 DOI: 10.1016/j.biomaterials.2025.123341] [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: 11/21/2024] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 05/04/2025]
Abstract
Adipose tissue is the primary site of energy storage in the body and a key regulator of metabolism. However, different adipose depots exhibit distinct molecular and phenotypic characteristics that have yet to be fully unraveled. While initially considered inert, bone marrow adipocytes (BMAs) have been recognized as key regulators of bone homeostasis, and more recently bone pathologies, although many unknowns remain. In this review, we summarize the current knowledge on BMAs, focusing on their distinct characteristics, functional significance in bone physiology and metabolism, as well as their emerging role in cancer pathogenesis. We present and discuss the current methodologies for investigating BMA-cancer interactions, encompassing both in vitro 3D culture systems and in vivo models, and their limitations in accurately replicating the phenotypes and biological processes of the human species. We highlight the imperative for advancing towards humanized models to better mimic the complexities of human physiology and disease progression. Finally, therapeutic strategies targeting metabolism or BMA-secreted factors, such as anti-cholesterol drugs, hold considerable promise in cancer treatment. We present the synergistic avenue of combining conventional cancer therapies with agents targeting adipocyte signaling to amplify treatment efficacy. Developing preclinical models that more faithfully replicate human pathological and physiological processes will lead to more accurate mechanistic understanding of the role of BMAs in bone metastasis and lead to more relevant preclinical drug screening.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia
| | - Jennifer Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia; Centre for Genomics and Personalised Health, QUT, Brisbane, QLD, 4102, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia; Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD, 4000, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia; Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Brisbane, QLD, 4000, Australia; Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia; Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| |
Collapse
|
2
|
Yang L, Wang X, Wang S, Shen J, Li Y, Wan S, Xiao Z, Wu Z. Targeting lipid metabolism in regulatory T cells for enhancing cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189259. [PMID: 39798823 DOI: 10.1016/j.bbcan.2025.189259] [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: 06/11/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
As immunosuppressive cells, Regulatory T cells (Tregs) exert their influence on tumor immune escape within the tumor microenvironment (TME) by effectively suppressing the activity of other immune cells, thereby significantly impeding the anti-tumor immune response. In recent years, the metabolic characteristics of Tregs have become a focus of research, especially the important role of lipid metabolism in maintaining the function of Tregs. Consequently, targeted interventions aimed at modulating lipid metabolism in Tregs have been recognized as an innovative and promising approach to enhance the effectiveness of tumor immunotherapy. This review presents a comprehensive overview of the pivotal role of lipid metabolism in regulating the function of Tregs, with a specific focus on targeting Tregs lipid metabolism as an innovative approach to augment anti-tumor immune responses. Furthermore, we discuss potential opportunities and challenges associated with this strategy, aiming to provide novel insights for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Liu Yang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shurong Wang
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yaling Li
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shengli Wan
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Zhigui Wu
- Department of Pharmacy, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China; Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646000, China; Laboratory of Personalised Cell Therapy and Cell Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
3
|
Bessot A, Röhl J, Emmerich M, Klotz A, Ravichandran A, Meinert C, Waugh D, McGovern J, Gunter J, Bock N. ECM-mimicking hydrogel models of human adipose tissue identify deregulated lipid metabolism in the prostate cancer-adipocyte crosstalk under antiandrogen therapy. Mater Today Bio 2025; 30:101424. [PMID: 39866784 PMCID: PMC11764633 DOI: 10.1016/j.mtbio.2024.101424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025] Open
Abstract
Antiandrogen therapies are effectively used to treat advanced prostate cancer, but eventually cancer adaptation drives unresolved metastatic castration-resistant prostate cancer (mCRPC). Adipose tissue influences metabolic reprogramming in cancer and was proposed as a contributor to therapy resistance. Using extracellular matrix (ECM)-mimicking hydrogel coculture models of human adipocytes and prostate cancer cells, we show that adipocytes from subcutaneous or bone marrow fat have dissimilar responses under the antiandrogen Enzalutamide. We demonstrate that androgen receptor (AR)-dependent cancer cells (LNCaP) are more influenced by human adipocytes than AR-independent cells (C4-2B), with altered lipid metabolism and adipokine secretion. This response changes under Enzalutamide, with increased AR expression and adipogenic and lipogenic genes in cancer cells and decreased lipid content and gene dysregulation associated with insulin resistance in adipocytes. This is in line with the metabolic syndrome that men with mCRPC under Enzalutamide experience. The all-human, all-3D, models presented here provide a significant advance to dissect the role of fat in therapy response for mCRPC.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Joan Röhl
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, 4226, Australia
| | - Maria Emmerich
- School of Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
| | - Anton Klotz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, QUT, Brisbane, QLD 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | | | - David Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5005, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | - Jenni Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
4
|
Zhang Y, Yang Z, Liu Y, Pei J, Li R, Yang Y. Targeting lipid metabolism: novel insights and therapeutic advances in pancreatic cancer treatment. Lipids Health Dis 2025; 24:12. [PMID: 39806478 PMCID: PMC11727729 DOI: 10.1186/s12944-024-02426-0] [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: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid metabolism in cancer is characterized by dysregulated lipid regulation and utilization, critical for promoting tumor growth, survival, and resistance to therapy. Pancreatic cancer (PC) is a highly aggressive malignancy of the gastrointestinal tract that has a dismal 5-year survival rate of less than 10%. Given the essential function of the pancreas in digestion, cancer progression severely disrupts its function. Standard treatments for PC such as surgical resection, chemotherapy, and radiotherapy. However, these therapies often face significant challenges, including biochemical recurrence and drug resistance.Given these limitations, new therapeutic approaches are being developed to target tumor metabolism. Dysregulation of cholesterol biosynthesis and alterations in fatty acids (FAs), such as palmitate, stearate, omega-3, and omega-6, have been observed in pancreatic cancer. These lipids serve as energy sources, signaling molecules, and essential components of cell membranes. Their accumulation fosters an immunosuppressive tumor microenvironment that supports cancer cell proliferation and metastasis.Moreover, lipid metabolism dysregulation within immune cells, particularly T cells, impairs immune surveillance and weakens the body's defenses against cancer. Abnormal lipid metabolism also contributes to drug resistance in PC. Despite these challenges, targeting lipid metabolism may offer a promising therapeutic strategy. By enhancing lipid peroxidation, the induction of ferroptosis-a form of regulated cell death-could impair the survival of PC cells and hinder disease progression.
Collapse
Affiliation(s)
- Yanyan Zhang
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Qinling, Hanzhong, 723001, China
| | - Zhichao Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Dalian Medical University, Dalian, China
| | - Yuchen Liu
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Qinling, Hanzhong, 723001, China
| | - Jinjin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Bashan Mountains Bioresources Comprehensive Development C.I.C, Shaanxi University of Technology, Qinling, Hanzhong, 723001, China
| | - Ruojie Li
- Interventional Therapy Department, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, P.R. China.
| | - Yanhui Yang
- Emergency surgery Dapartment (Trauma center), The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, Henan, China.
| |
Collapse
|
5
|
Zwara A, Hellmann A, Czapiewska M, Korczynska J, Sztendel A, Mika A. The influence of cancer on the reprogramming of lipid metabolism in healthy thyroid tissues of patients with papillary thyroid carcinoma. Endocrine 2025; 87:273-280. [PMID: 39145825 PMCID: PMC11739254 DOI: 10.1007/s12020-024-03993-z] [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: 06/19/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND AND OBJECTIVES Over the years we observed changes in the metabolism of glucose, amino acids, fatty acids (FA) and nucleotides in cancer cells in order to maintain their viability and proliferate. Moreover, as the latest data show, cancer also forces a complete change in the behavior of other tissues. For instance, fat-filled adipocytes are often found in the vicinity of invasive solid human tumors. We investigated the effects of papillary thyroid carcinoma (PTC) on the lipid metabolism of healthy tissue distant from the tumor. METHOD Thyroid tissue was collected from female patients immediately after surgical removal of the entire thyroid gland. Blood samples were collected from PTC patients and healthy volunteers. Real-time PCR assays were performed to analyze the expression of lipogenic genes and a broad panel of FA was determined using the gas chromatography-mass spectrometry method. RESULTS The concentration of lipids was higher in paratumor tissue than in healthy thyroid tissue (p = 0.005). The lipogenic genes tested were significantly increased in paratumor tissue compared to healthy tissue, especially enzymes related to the synthesis of very long-chain saturated and polyunsaturated FAs (VLCSFAs and PUFAs, respectively) (p < 0.001). The FA profile also showed increased levels of C22-C26, VLCSFAs and almost all PUFAs in paratumor tissue (p < 0.05). CONCLUSION Our study suggests that a restructuring of lipid metabolism occurs in the adjacent healthy thyroid gland and that the metabolism of VLCSFAs and PUFAs is higher in the paratumor tissue than in the distant tissue of the healthy thyroid gland.
Collapse
Affiliation(s)
- Agata Zwara
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
| | - Andrzej Hellmann
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland.
| | - Monika Czapiewska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Justyna Korczynska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Alicja Sztendel
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| | - Adriana Mika
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
6
|
Fallone F, Rebeaud M, Bouche C, Fontaine J, Arellano C, Ducoux-Petit M, Orgerit L, Deudon R, Nicolle R, Franchet C, Estève D, Mouton-Barbosa E, Dauvillier S, Moutahir M, Burlet-Schiltz O, Bouloumié A, Vaysse C, Muller C. Lack of fibro-inflammatory response in human mammary adipose tissue in obesity. Int J Obes (Lond) 2024:10.1038/s41366-024-01705-1. [PMID: 39738492 DOI: 10.1038/s41366-024-01705-1] [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] [Received: 04/29/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Understanding how obesity impacts human mammary adipose tissue (MAT) biology is crucial for deciphering its role in mammary epithelium during both physiological and pathophysiological processes, including breast cancer. Hypertrophic mammary adipocytes and Crown-Like Structures are present in MAT of patients with obesity but whether these changes initiate a fibro-inflammatory response at the tissue level remains insufficiently explored. OBJECTIVE We investigated the markers of adipose tissue dysfunction (immune cell infiltration, secretion pattern and fibrosis) in tumor-free MAT of patients with obesity versus patients who are lean. METHODS Tumor-free MAT were obtained from 96 women with (n = 43) or without (n = 53) obesity who underwent mastectomy for breast cancer risk reduction or treatment. Immune and non-immune cell infiltration were determined using flow cytometry. Bulk transcriptomic was used to characterize the phenotype of CD206+ macrophages whose infiltration is increased in patients with obesity. Conditioned-medium were prepared from MAT to characterize their secretome and dose adipokines and cytokines by ELISA assay. The extra-cellular matrix (ECM) deposition was evaluated by Masson trichrome staining on cross-stained sections, 3D imaging of red picrosirius-stained tissues and measure of hydroxyproline content. RESULTS We observed an increase of CD206+/HLA-DR+ macrophages in the stromal vascular fraction of MAT from patients with obesity compared to patients who are lean. Other immune cell infiltration and endothelial or adipose progenitor cell numbers were similar between groups. Bulk transcriptomics on CD206+ macrophages revealed a significant decrease in ECM component expression and processing in obesity. In addition, no heightened secretion of pro-inflammatory cytokines, TGF-β1 or MCP-1 was observed in the samples from patients with obesity. ECM characterization revealed an absence of fibrosis, with MAT of patients with obesity showing even a slightly reduced collagen secretion and deposition compared with their lean counterparts. CONCLUSIONS Obesity is not associated with inflammation nor fibrosis in MAT, highlighting its unique behavior.
Collapse
Affiliation(s)
- Frédérique Fallone
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France.
| | - Marie Rebeaud
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Caroline Bouche
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Jessica Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Carlo Arellano
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Manuelle Ducoux-Petit
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Lucyle Orgerit
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Rémi Deudon
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Rémy Nicolle
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018, Paris, France
| | - Camille Franchet
- Département d'Anatomo-Pathologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - David Estève
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Stéphanie Dauvillier
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Mohamed Moutahir
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Infrastructure Nationale de Protéomique, ProFI, FR 2048, Toulouse, France
| | - Anne Bouloumié
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université de Toulouse, INSERM, UPS, Toulouse, France
| | - Charlotte Vaysse
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France
- Département de Chirurgie Gynécologique Oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III-Paul Sabatier (UT3), Toulouse, France.
| |
Collapse
|
7
|
Stanisławowski M. Effect of adipose tissue on the development of multiple myeloma. Mol Biol Rep 2024; 52:74. [PMID: 39708277 DOI: 10.1007/s11033-024-10174-8] [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: 09/17/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Multiple myeloma (MM), also referred to as Kahler's disease, is a cancer characterized by the uncontrolled growth of abnormal plasma cells and is associated with alterations in the bone tissue microenvironment. Bone marrow adipose tissue (BMAT), which comprises approximately ten percent of total body fat, can influence the progression, survival, and drug resistance of MM cells through paracrine, hormonal, and metabolic pathways. Obesity can lead to an increase in BMAT mass, which not only disrupts bone metabolism but also reduces bone density, potentially progressing from monoclonal gammopathy of undetermined significance, a benign condition, to MM. A range of factors, including impaired fatty acid metabolism, increased production of adipokines that support myeloma, and heightened expression of oncogenic microRNAs in multiple myeloma, contribute to the progression of this incurable blood cancer. To better understand the relationship between excess adipose tissue accumulation and the risk of developing multiple myeloma, a comprehensive review of published data was conducted.
Collapse
Affiliation(s)
- Marcin Stanisławowski
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland.
| |
Collapse
|
8
|
Calabrese C, Miserocchi G, De Vita A, Spadazzi C, Cocchi C, Vanni S, Gabellone S, Martinelli G, Ranallo N, Bongiovanni A, Liverani C. Lipids and adipocytes involvement in tumor progression with a focus on obesity and diet. Obes Rev 2024; 25:e13833. [PMID: 39289899 DOI: 10.1111/obr.13833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The adipose tissue is a complex organ that can play endocrine, metabolic, and immune regulatory roles in cancer. In particular, adipocytes provide metabolic substrates for cancer cell proliferation and produce signaling molecules that can stimulate cell adhesion, migration, invasion, angiogenesis, and inflammation. Cancer cells, in turn, can reprogram adipocytes towards a more inflammatory state, resulting in a vicious cycle that fuels tumor growth and evolution. These mechanisms are enhanced in obesity, which is associated with the risk of developing certain tumors. Diet, an exogenous source of lipids with pro- or anti-inflammatory functions, has also been connected to cancer risk. This review analyzes how adipocytes and lipids are involved in tumor development and progression, focusing on the relationship between obesity and cancer. In addition, we discuss how diets with varying lipid intakes can affect the disease outcomes. Finally, we introduce novel metabolism-targeted treatments and adipocyte-based therapies in oncology.
Collapse
Affiliation(s)
- Chiara Calabrese
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sofia Gabellone
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicoletta Ranallo
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alberto Bongiovanni
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
9
|
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
|
10
|
Abduljabbar MK, Merza M, Aziz A, Menon SV, Kaur M, Aminov Z, Rab SO, Hjazi A, Mustafa YF, Gabel BC. Lipid metabolism reprogramming in renal cell carcinomas. Med Oncol 2024; 41:243. [PMID: 39240415 DOI: 10.1007/s12032-024-02484-5] [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: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024]
Abstract
This study investigates the intricate mechanisms underlying the correlation between elevated consumption of harmful fats and the onset of kidney malignancies. The rise in global obesity rates has been accompanied by an increased prevalence of renal cancers, prompting an exploration into the molecular pathways and biological processes linking these phenomena. Through an extensive review of current literature and clinical studies, we identify potential key factors contributing to the carcinogenic influence of harmful fats on renal tissues. Our analysis highlights the role of adipose tissue-derived factors, inflammatory mediators, and lipid metabolism dysregulation in fostering a microenvironment conducive to renal tumorigenesis. Furthermore, we delve into the impact of harmful fats on signaling pathways associated with cell proliferation, apoptosis evasion, and angiogenesis within the renal parenchyma. This review underscores the importance of elucidating the molecular intricacies linking lipid metabolism and kidney malignancies, offering a foundation for future research and the development of targeted preventive and therapeutic interventions. The findings discussed herein contribute to our understanding of the complex relationship between lipid mediators and renal cancer, providing a basis for public health strategies aimed at mitigating the impact of harmful fats on kidney health.
Collapse
Affiliation(s)
| | - Mohammed Merza
- Clinical Analysis Department, Hawler Medical University, Kurdistan Regional Government, Erbil, Iraq.
- Medical Biochemical Analysis Department, College of Health Technology, Cihan University, Erbil, Kurdistan Region, Iraq.
| | - Abdulqader Aziz
- Faculty of Pharmacy, Tishk International University, Kurdistan Region of Iraq, Erbil, Iraq.
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul-41001, Iraq
| | - Benien C Gabel
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
11
|
Märkl B, Reitsam NG, Grochowski P, Waidhauser J, Grosser B. The SARIFA biomarker in the context of basic research of lipid-driven cancers. NPJ Precis Oncol 2024; 8:165. [PMID: 39085485 PMCID: PMC11291993 DOI: 10.1038/s41698-024-00662-2] [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: 02/27/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
SARIFA was very recently introduced as a histomorphological biomarker with strong prognostic power for colorectal, gastric, prostate, and pancreatic cancer. It is characterized by the direct contact between tumor cells and adipocytes due to a lack of stromal reaction. This can be easily evaluated on routinely available H&E-slides with high interobserver agreement. SARIFA also reflects a specific tumor biology driven by metabolic reprogramming. Tumor cells in SARIFA-positive tumors benefit from direct interaction with adipocytes as an external source of lipids. Numerous studies have shown that lipid metabolism is crucial in carcinogenesis and cancer progression. We found that the interaction between tumor cells and adipocytes was not triggered by obesity, as previously assumed. Instead, we believe that this is due to an immunological mechanism. Knowledge about lipid metabolism in cancer from basic experiments can be transferred to develop strategies targeting this reprogramed metabolism.
Collapse
Affiliation(s)
- Bruno Märkl
- Pathology, Medical Faculty Augsburg, University of Augsburg, Augsburg, Germany.
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany.
- WERA Comprehensive Cancer Center, Augsburg, Germany.
| | - Nic G Reitsam
- Pathology, Medical Faculty Augsburg, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- WERA Comprehensive Cancer Center, Augsburg, Germany
| | - Przemyslaw Grochowski
- Pathology, Medical Faculty Augsburg, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- WERA Comprehensive Cancer Center, Augsburg, Germany
| | - Johanna Waidhauser
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- WERA Comprehensive Cancer Center, Augsburg, Germany
- Hematology and Oncology, Medical Faculty Augsburg, University of Augsburg, Augsburg, Germany
| | - Bianca Grosser
- Pathology, Medical Faculty Augsburg, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
- WERA Comprehensive Cancer Center, Augsburg, Germany
| |
Collapse
|
12
|
Offermans K, Reitsam NG, Simons CCJM, Grosser B, Zimmermann J, Grabsch HI, Märkl B, van den Brandt PA. The relationship between Stroma AReactive Invasion Front Areas (SARIFA), Warburg-subtype and survival: results from a large prospective series of colorectal cancer patients. Cancer Metab 2024; 12:21. [PMID: 38992781 PMCID: PMC11241902 DOI: 10.1186/s40170-024-00349-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Stroma AReactive Invasion Front Areas (SARIFA) is a recently identified haematoxylin & eosin (H&E)based histopathologic biomarker in gastrointestinal cancers, including colorectal cancer (CRC), defined as direct contact between tumour cells and adipocytes at the tumour invasion front. The current study aimed at validating the prognostic relevance of SARIFA in a large population-based CRC series as well as at investigating the relationship between SARIFA-status and previously established Warburg-subtypes, both surrogates of the metabolic state of the tumour cells. METHODS SARIFA-status (positive versus negative) was determined on H&E slides of 1,727 CRC specimens. Warburg-subtype (high versus moderate versus low) data was available from our previous study. The associations between SARIFA-status, Warburg-subtype, clinicopathological characteristics and CRC-specific as well as overall survival were investigated. RESULTS 28.7% (n=496) CRC were SARIFA-positive. SARIFA-positivity was associated with more advanced disease stage, higher pT category, and more frequent lymph node involvement (all p<0.001). SARIFA-positivity was more common in Warburg-high CRC. 44.2% (n=219) of SARIFA-positive CRCs were Warburg-high compared to 22.8% (n=113) being Warburg-low and 33.1% (n=164) being Warburg-moderate (p<0.001). In multivariable-adjusted analysis, patients with SARIFA-positive CRCs had significantly poorer CRC-specific (HRCRC-specific 1.65; 95% CI 1.41-1.93) and overall survival (HRoverall survival 1.46; 95% CI 1.28-1.67) independent of clinically known risk factors and independent of Warburg-subtype. Combining the SARIFA-status and the Warburg-subtype to a combination score (SARIFA-negative/Warburg-high versus SARIFA-positive/Warburg-low versus SARIFA-positive/Warburg-high, and so on) did not improve the survival prediction compared to the use of SARIFA-status alone (SARIFA-negative + Warburg-high: HRCRC-specific 1.08; 95% CI 0.84-1.38; SARIFA-positive + Warburg-low: HRCRC-specific 1.79; 95% CI 1.32-2.41; SARIFA-positive + Warburg-high: HRCRC-specific 1.58; 95% CI 1.23-2.04). CONCLUSIONS Our current study is the by far largest external validation of SARIFA-positivity as a novel independent negative prognostic H&E-based biomarker in CRC. In addition, our study shows that SARIFA-positivity is associated with the Warburg-high subtype. Further research is warranted to provide a more mechanistic understanding of the underlying tumour biology. Based on our data, we conclude SARIFA-status should be implemented in pathologic routine practice to stratify CRC patients.
Collapse
Affiliation(s)
- Kelly Offermans
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Nic G Reitsam
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Colinda C J M Simons
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Bianca Grosser
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | | | - Heike I Grabsch
- Department of Pathology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands.
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.
| | - Bruno Märkl
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany.
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany.
| | - Piet A van den Brandt
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands.
- Department of Epidemiology, Care and Public Health Research Institute (CAPHRI), Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
13
|
Garlisi B, Lauks S, Aitken C, Ogilvie LM, Lockington C, Petrik D, Eichhorn JS, Petrik J. The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Curr Oncol 2024; 31:3826-3844. [PMID: 39057155 PMCID: PMC11275383 DOI: 10.3390/curroncol31070283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) in ovarian cancer (OC) has much greater complexity than previously understood. In response to aggressive pro-angiogenic stimulus, blood vessels form rapidly and are dysfunctional, resulting in poor perfusion, tissue hypoxia, and leakiness, which leads to increased interstitial fluid pressure (IFP). Decreased perfusion and high IFP significantly inhibit the uptake of therapies into the tumor. Within the TME, there are numerous inhibitor cells, such as myeloid-derived suppressor cells (MDSCs), tumor association macrophages (TAMs), regulatory T cells (Tregs), and cancer-associated fibroblasts (CAFs) that secrete high numbers of immunosuppressive cytokines. This immunosuppressive environment is thought to contribute to the lack of success of immunotherapies such as immune checkpoint inhibitor (ICI) treatment. This review discusses the components of the TME in OC, how these characteristics impede therapeutic efficacy, and some strategies to alleviate this inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (B.G.); (S.L.); (C.A.); (L.M.O.); (C.L.); (D.P.); (J.S.E.)
| |
Collapse
|
14
|
Rebeaud M, Lacombe M, Fallone F, Milhas D, Roumiguié M, Vaysse C, Attané C, Muller C. Specificities of mammary and periprostatic adipose tissues: A perspective from cancer research. ANNALES D'ENDOCRINOLOGIE 2024; 85:220-225. [PMID: 38871505 DOI: 10.1016/j.ando.2024.05.016] [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: 06/15/2024]
Abstract
In addition to the major subcutaneous and visceral adipose tissues (AT), other adipose depots are dispersed throughout the body and are found in close interaction with proximal organs such as mammary and periprostatic AT (MAT and PPAT respectively). These ATs have an effect on proximal organ function during physiological processes and diseases such as cancer. We highlighted here some of their most distinctive features in terms of tissular organization and responses to external stimuli and discussed how obesity affects them based on our current knowledge.
Collapse
Affiliation(s)
- Marie Rebeaud
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Mathilde Lacombe
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Frédérique Fallone
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Delphine Milhas
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Mathieu Roumiguié
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France; Département d'urologie, CHU de Toulouse, 1, avenue du Professeur-Jean-Poulhès, 31400 Toulouse, France
| | - Charlotte Vaysse
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France; Département de chirurgie gynécologique-oncologique, institut universitaire du cancer de Toulouse-Oncopole, CHU de Toulouse, 1, avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France
| | - Camille Attané
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France
| | - Catherine Muller
- UMR 5089, CNRS, équipe labélisée ligue nationale contre le cancer, institut de pharmacologie et de biologie structurale, université de Toulouse, 205, route de Narbonne, BP 64182, 31077 Toulouse, France.
| |
Collapse
|
15
|
Andele PK, Palazzolo S, Corona G, Caligiuri I, Kamensek U, Cemazar M, Canzonieri V, Rizzolio F. Human Omental Mature Adipocytes used as Paclitaxel Reservoir for Cell-Based Therapy in Ovarian Cancer. Adv Healthc Mater 2024; 13:e2304206. [PMID: 38334216 DOI: 10.1002/adhm.202304206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/20/2024] [Indexed: 02/10/2024]
Abstract
Primary human omental adipocytes and ovarian cancer(OC) cells establish a bidirectional communication in which tumor driven lipolysis is induced in adipocytes and the resulting fatty acids are delivered to cancer cells within the tumor microenvironment. Despite meaningful improvement in the treatment of OC, its efficacy is still limited by hydrophobicity and untargeted effects related to chemotherapeutics. Herein, omental adipocytes are firstly used as a reservoir for paclitaxel, named Living Paclitaxel Bullets (LPB) and secondly benefit from the established dialogue between adipocytes and cancer cells to engineer a drug delivery process that target specifically cancer cells. These results show that mature omental adipocytes can successfully uptake paclitaxel and deliver it to OC cells in a transwell coculture based in vitro model. In addition, the efficacy of this proof-of-concept has been demonstrated in vivo and induces a significant inhibition of tumor growth on a xenograft tumor model. The use of mature adipocytes can be suitable for clinical prospection in a cell-based therapy system, due to their mature and differentiated state, to avoid risks related to uncontrolled cell de novo proliferation capacity after the delivery of the antineoplastic drug as observed with other cell types when employed as drug carriers.
Collapse
Affiliation(s)
- Pacome K Andele
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Venice, 30172, Italy
- Department of Pathology, IRCCS CRO Aviano National Cancer Institute, Aviano, 33081, Italy
| | - Stefano Palazzolo
- Department of Pathology, IRCCS CRO Aviano National Cancer Institute, Aviano, 33081, Italy
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, 33081, Italy
| | - Isabella Caligiuri
- Department of Pathology, IRCCS CRO Aviano National Cancer Institute, Aviano, 33081, Italy
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Vincenzo Canzonieri
- Department of Pathology, IRCCS CRO Aviano National Cancer Institute, Aviano, 33081, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, 34127, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Venice, 30172, Italy
- Department of Pathology, IRCCS CRO Aviano National Cancer Institute, Aviano, 33081, Italy
| |
Collapse
|
16
|
Halpert MM, Burns BA, Rosario SR, Withers HG, Trivedi AJ, Hofferek CJ, Gephart BD, Wang H, Vazquez-Perez J, Amanya SB, Hyslop ST, Yang J, Kemnade JO, Sandulache VC, Konduri V, Decker WK. Multifactoral immune modulation potentiates durable remission in multiple models of aggressive malignancy. FASEB J 2024; 38:e23644. [PMID: 38738472 PMCID: PMC11155525 DOI: 10.1096/fj.202302675r] [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: 12/25/2023] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
Tumors typically lack canonical danger signals required to activate adaptive immunity and also frequently employ substantial immunomodulatory mechanisms that downregulate adaptive responses and contribute to escape from immune surveillance. Given the variety of mechanisms involved in shielding tumors from immune recognition, it is not surprising that single-agent immunomodulatory approaches have been largely unsuccessful in generating durable antitumor responses. Here we report a unique combination of immunomodulatory and cytostatic agents that recondition the tumor microenvironment and eliminate complex and/or poor-prognosis tumor types including the non-immunogenic 4T-1 model of TNBC, the aggressive MOC-2 model of HNSCC, and the high-risk MYCN-amplified model of neuroblastoma. A course of therapy optimized for TNBC cured a majority of tumors in both ectopic and orthotopic settings and eliminated metastatic spread in all animals tested at the highest doses. Immune responses were transferable between therapeutic donor and naïve recipient through adoptive transfer, and a sizeable abscopal effect on distant, untreated lesions could be demonstrated experimentally. Similar results were observed in HNSCC and neuroblastoma models, with characteristic remodeling of the tumor microenvironment documented in all model systems. scRNA-seq analysis implicated upregulation of innate immune responses and antigen presentation in tumor cells and the myeloid cell compartment as critical early events. This analysis also highlighted the potential importance of the autonomic nervous system in the governance of inflammatory processes. The data indicate that the targeting of multiple pathways and mechanisms of action can result in substantial synergistic antitumor effects and suggest follow-up in the neoadjuvant setting may be warranted.
Collapse
Affiliation(s)
- MM Halpert
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - BA Burns
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - SR Rosario
- Department of Biostatistics and Bioinformatics, Baylor College of Medicine, Houston, TX 77030 United States
- Acquired Resistance to Therapy Network (ARTNet) U24/U54 Investigator, Baylor College of Medicine, Houston, TX 77030 United States
| | - HG Withers
- Department of Biostatistics and Bioinformatics, Baylor College of Medicine, Houston, TX 77030 United States
| | - AJ Trivedi
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - CJ Hofferek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - BD Gephart
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - H Wang
- Department of Medicine, Section of Hematology & Oncology, Baylor College of Medicine, Houston, TX 77030 United States
| | - J Vazquez-Perez
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - SB Amanya
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - ST Hyslop
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
| | - J Yang
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030 United States
| | - JO Kemnade
- Department of Medicine, Section of Hematology & Oncology, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
| | - VC Sandulache
- Acquired Resistance to Therapy Network (ARTNet) U24/U54 Investigator, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
- Bobby R. Alford Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, TX 77030 United States
| | - V Konduri
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
| | - WK Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 United States
- Acquired Resistance to Therapy Network (ARTNet) U24/U54 Investigator, Baylor College of Medicine, Houston, TX 77030 United States
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030 United States
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030 United States
| |
Collapse
|
17
|
Arumugam M, Tovar EA, Essenburg CJ, Dischinger PS, Beddows I, Wolfrum E, Madaj ZB, Turner L, Feenstra K, Gallik KL, Cohen L, Nichols M, Sheridan RTC, Esquibel CR, Mouneimne G, Graveel CR, Steensma MR. Nf1 deficiency modulates the stromal environment in the pretumorigenic rat mammary gland. Front Cell Dev Biol 2024; 12:1375441. [PMID: 38799507 PMCID: PMC11116614 DOI: 10.3389/fcell.2024.1375441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024] Open
Abstract
Background Neurofibromin, coded by the NF1 tumor suppressor gene, is the main negative regulator of the RAS pathway and is frequently mutated in various cancers. Women with Neurofibromatosis Type I (NF1)-a tumor predisposition syndrome caused by a germline NF1 mutation-have an increased risk of developing aggressive breast cancer with poorer prognosis. The mechanism by which NF1 mutations lead to breast cancer tumorigenesis is not well understood. Therefore, the objective of this work was to identify stromal alterations before tumor formation that result in the increased risk and poorer outcome seen among NF1 patients with breast cancer. Approach To accurately model the germline monoallelic NF1 mutations in NF1 patients, we utilized an Nf1-deficient rat model with accelerated mammary development before presenting with highly penetrant breast cancer. Results We identified increased collagen content in Nf1-deficient rat mammary glands before tumor formation that correlated with age of tumor onset. Additionally, gene expression analysis revealed that Nf1-deficient mature adipocytes in the rat mammary gland have increased collagen expression and shifted to a fibroblast and preadipocyte expression profile. This alteration in lineage commitment was also observed with in vitro differentiation, however, flow cytometry analysis did not show a change in mammary adipose-derived mesenchymal stem cell abundance. Conclusion Collectively, this study uncovered the previously undescribed role of Nf1 in mammary collagen deposition and regulating adipocyte differentiation. In addition to unraveling the mechanism of tumor formation, further investigation of adipocytes and collagen modifications in preneoplastic mammary glands will create a foundation for developing early detection strategies of breast cancer among NF1 patients.
Collapse
Affiliation(s)
- Menusha Arumugam
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Elizabeth A. Tovar
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Curt J. Essenburg
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Patrick S. Dischinger
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Ian Beddows
- Biostatistics ad Bioinformatics Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Emily Wolfrum
- Biostatistics ad Bioinformatics Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Zach B. Madaj
- Biostatistics ad Bioinformatics Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Lisa Turner
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Kristin Feenstra
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Kristin L. Gallik
- Optical Imaging Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Lorna Cohen
- Optical Imaging Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Madison Nichols
- Flow Cytometry Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | | | - Corinne R. Esquibel
- Optical Imaging Core, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Ghassan Mouneimne
- University of Arizona Cancer Center, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Carrie R. Graveel
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
| | - Matthew R. Steensma
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, United States
- Helen DeVos Children’s Hospital, Spectrum Health System, Grand Rapids, MI, United States
- Michigan State University College of Human Medicine, Grand Rapids, MI, United States
| |
Collapse
|
18
|
Yang Y, Li H, Liu K, Zou L, Xiang S, Geng Y, Li X, Qiu S, Yang J, Cui X, Li L, Li Y, Li W, Yan S, Liu L, Wu X, Liu F, Wu W, Chen S, Liu Y. Acylcarnitines promote gallbladder cancer metastasis through lncBCL2L11-THOC5-JNK axis. J Transl Med 2024; 22:299. [PMID: 38519939 PMCID: PMC10958842 DOI: 10.1186/s12967-024-05091-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The progression of gallbladder cancer (GBC) is accompanied by abnormal fatty acid β-oxidation (FAO) metabolism. Different types of lipids perform various biological functions. This study aimed to determine the role of acyl carnitines in the molecular mechanisms of GBC progression. METHODS Distribution of lipids in GBC was described by LC-MS-based lipidomics. Cellular localization, expression level and full-length of lncBCL2L11 were detected using fluorescence in situ hybridization (FISH) assays, subcellular fractionation assay and 5' and 3' rapid amplification of the cDNA ends (RACE), respectively. In vitro and in vivo experiments were used to verify the biological function of lncBCL2L11 in GBC cells. Methylated RNA Immunoprecipitation (MeRIP) was performed to detect the methylation levels of lncBCL2L11. RNA pull-down assay and RNA immunoprecipitation (RIP) assay were used to identify lncBCL2L11 interacting proteins. Co-Immunoprecipitation (Co-IP) and Western blot assay were performed to validate the regulatory mechanism of lncBCL2L11 and THO complex. RESULTS Acylcarnitines were significantly up-regulated in GBC tissues. High serum triglycerides correlated to decreased survival in GBC patients and promoted tumor migration. LncBCL2L11 was identified in the joint analysis of highly metastatic cells and RNA sequencing data. LncBCl2L11 prevented the binding of THOC6 and THOC5 and causes the degradation of THOC5, thus promoting the accumulation of acylcarnitines in GBC cells, leading to the malignant progression of cancer cells. In addition, highly expressed acylcarnitines stabilized the expression of lncBCL2L11 through N6-methyladenosine methylation (m6A), forming a positive feedback regulation in tumor dissemination. CONCLUSIONS LncBCL2L11 is involved in gallbladder cancer metastasis through FAO metabolism. High lipid intake is associated with poor prognosis of GBC. Therefore, targeting lncBCL2L11 and its pathway-related proteins or reducing lipid intake may be significant for the treatment of GBC patients.
Collapse
Affiliation(s)
- Yang Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Huaifeng Li
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ke Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lu Zou
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shanshan Xiang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yajun Geng
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xuechuan Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shimei Qiu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Jiahua Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xuya Cui
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lin Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yang Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Weijian Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Siyuan Yan
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Liguo Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiangsong Wu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Fatao Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Wenguang Wu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Shili Chen
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
19
|
Sun S, Xu H, Zhao W, Li Q, Yuan Y, Zhang G, Li S, Wang B, Zhang W, Gao X, Zheng J, Zhang Q. PA suppresses antitumor immunity of T cells by disturbing mitochondrial activity through Akt/mTOR-mediated Ca 2+ flux. Cancer Lett 2024; 581:216511. [PMID: 38013049 DOI: 10.1016/j.canlet.2023.216511] [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: 07/25/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Deciphering the mechanisms behind how T cells become exhausted and regulatory T cells (Tregs) differentiate in a tumor microenvironment (TME) will significantly benefit cancer immunotherapy. A common metabolic alteration feature in TME is lipid accumulation, associated with T cell exhaustion and Treg differentiation. However, the regulatory role of free fatty acids (FFA) on T cell antitumor immunity has yet to be clearly illustrated. Our study observed that palmitic acid (PA), the most abundant saturated FFA in mouse plasma, enhanced T cell exhaustion and Tregs population in TME and increased tumor growth. In contrast, oleic acid (OA), a monounsaturated FFA, rescued PA-induced T cell exhaustion, decreased Treg population, and ameliorated T cell antitumor immunity in an obese mouse model. Mechanistically, mitochondrial metabolic activity is critical in maintaining T cell function, which PA attenuated. PA-induced T cell exhaustion and Treg formation depended on CD36 and Akt/mTOR-mediated calcium signaling. The study described a new mechanism of PA-induced downregulation of antitumor immunity of T cells and the therapeutic potential behind its restoration by targeting PA.
Collapse
Affiliation(s)
- Shishuo Sun
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Heng Xu
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Wanxin Zhao
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Qihong Li
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Yifan Yuan
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Guopeng Zhang
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Shuyu Li
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Bixi Wang
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Wei Zhang
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Xiaoge Gao
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| | - Qing Zhang
- Cancer Institute, the First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| |
Collapse
|
20
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [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: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
21
|
Hassin O, Sernik M, Seligman A, Vogel FCE, Wellenstein MD, Smollich J, Halperin C, Pirona AC, Toledano LN, Caballero CD, Schlicker L, Salame TM, Sarusi Portuguez A, Aylon Y, Scherz-Shouval R, Geiger T, de Visser KE, Schulze A, Oren M. p53 deficient breast cancer cells reprogram preadipocytes toward tumor-protective immunomodulatory cells. Proc Natl Acad Sci U S A 2023; 120:e2311460120. [PMID: 38127986 PMCID: PMC10756271 DOI: 10.1073/pnas.2311460120] [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: 07/09/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
The TP53 gene is mutated in approximately 30% of all breast cancer cases. Adipocytes and preadipocytes, which constitute a substantial fraction of the stroma of normal mammary tissue and breast tumors, undergo transcriptional, metabolic, and phenotypic reprogramming during breast cancer development and play an important role in tumor progression. We report here that p53 loss in breast cancer cells facilitates the reprogramming of preadipocytes, inducing them to acquire a unique transcriptional and metabolic program that combines impaired adipocytic differentiation with augmented cytokine expression. This, in turn, promotes the establishment of an inflammatory tumor microenvironment, including increased abundance of Ly6C+ and Ly6G+ myeloid cells and elevated expression of the immune checkpoint ligand PD-L1. We also describe a potential gain-of-function effect of common p53 missense mutations on the inflammatory reprogramming of preadipocytes. Altogether, our study implicates p53 deregulation in breast cancer cells as a driver of tumor-supportive adipose tissue reprogramming, expanding the network of non-cell autonomous mechanisms whereby p53 dysfunction may promote cancer. Further elucidation of the interplay between p53 and adipocytes within the tumor microenvironment may suggest effective therapeutic targets for the treatment of breast cancer patients.
Collapse
Affiliation(s)
- Ori Hassin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Miriam Sernik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Adi Seligman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Felix C. E. Vogel
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg69120, Germany
| | - Max D. Wellenstein
- Division of Tumour Biology and Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam1066CX, The Netherlands
| | - Joachim Smollich
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Coral Halperin
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Anna Chiara Pirona
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Liron Nomi Toledano
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Carolina Dehesa Caballero
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg69120, Germany
| | - Lisa Schlicker
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg69120, Germany
| | - Tomer-Meir Salame
- Mass Cytometry Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Avital Sarusi Portuguez
- The Mantoux Bioinformatics Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Yael Aylon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Tamar Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Karin E. de Visser
- Division of Tumour Biology and Immunology, Oncode Institute, Netherlands Cancer Institute, Amsterdam1066CX, The Netherlands
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg69120, Germany
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot7610001, Israel
| |
Collapse
|
22
|
Roux A, Winnard PT, Van Voss MH, Muller L, Jackson SN, Hoffer B, Woods AS, Raman V. MALDI-MSI of lipids in a model of breast cancer brain metastasis provides a surrogate measure of ischemia/hypoxia. Mol Cell Biochem 2023; 478:2567-2580. [PMID: 36884151 DOI: 10.1007/s11010-023-04685-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023]
Abstract
Breast cancer brain metastasis (BCBM) has an incidence of 10-30%. It is incurable and the biological mechanisms that promote its progression remain largely undefined. Consequently, to gain insights into BCBM processes, we have developed a spontaneous mouse model of BCBM and in this study found a 20% penetrance of macro-metastatic brain lesion formation. Considering that lipid metabolism is indispensable to metastatic progression, our goal was the mapping of lipid distributions throughout the metastatic regions of the brain. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) of lipids revealed that, relative to surrounding brain tissue, seven long-chain (13-21 carbons long) fatty acylcarnitines, as well as two phosphatidylcholines, two phosphatidylinositols two diacylglycerols, a long-chain phosphatidylethanolamine, and a long-chain sphingomyelin were highly concentrated in the metastatic brain lesion In broad terms, lipids known to be enriched in brain tissues, such as very long-chain (≥ 22 carbons in length) polyunsaturated fatty acid of phosphatidylcholines, phosphatidylethanolamine, sphingomyelins, sulfatides, phosphatidylinositol phosphates, and galactosylceramides, were not found or only found in trace amounts in the metastatic lesion and instead consistently detected in surrounding brain tissues. The data, from this mouse model, highlights an accumulation of fatty acylcarnitines as possible biological makers of a chaotic inefficient vasculature within the metastasis, resulting in relatively inadequate blood flow and disruption of fatty acid β-oxidation due to ischemia/hypoxia.
Collapse
Affiliation(s)
- Aurelie Roux
- Structural Biology Unit, Cellular Neurobiology Branch, Integrative Neuroscience NIDA-IRP, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Paul T Winnard
- Division of Cancer Imaging Research, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Marise Heerma Van Voss
- Division of Cancer Imaging Research, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ludovic Muller
- Structural Biology Unit, Cellular Neurobiology Branch, Integrative Neuroscience NIDA-IRP, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Shelley N Jackson
- Structural Biology Unit, Cellular Neurobiology Branch, Integrative Neuroscience NIDA-IRP, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Barry Hoffer
- Case Western Reserve University, Cleveland, OH, USA
| | - Amina S Woods
- Structural Biology Unit, Cellular Neurobiology Branch, Integrative Neuroscience NIDA-IRP, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA.
- Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Venu Raman
- Division of Cancer Imaging Research, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Zhang M, Bai X, Du Q, Xu J, Wang D, Chen L, Dong K, Chen Z, Yang J. The Different Mechanisms of Lipid Accumulation in Hepatocytes Induced by Oleic Acid/Palmitic Acid and High-Fat Diet. Molecules 2023; 28:6714. [PMID: 37764494 PMCID: PMC10536454 DOI: 10.3390/molecules28186714] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the primary chronic liver disease worldwide, mainly manifested by hepatic steatosis. Hepatic lipids may be derived from dietary intake, plasma free fatty acid (FFA) uptake, or hepatic de novo lipogenesis (DNL). Currently, cellular and animal models of hepatocellular steatosis are widely used to study the pathogenesis of NAFLD and to investigate therapeutic agents. However, whether there are differences between the in vivo and in vitro models of the mechanisms that cause lipid accumulation has not been reported. We used OA/PA-induced NCTC 1469 cells and high-fat-diet-fed C57BL/6J mice to simulate a hepatocyte steatosis model of NAFLD and to detect indicators related to FFA uptake and DNL. In addition, when serological indicators were analysed in the mouse model, it was found that serum FASN levels decreased. The results revealed that, in the cellular model, indicators related to DNL were decreased, FASN enzyme activity was unchanged, and indicators related to FFA uptake were increased, including the high expression of CD36; while, in the animal model, indicators related to both FFA uptake and de novo synthesis were increased, including the high expression of CD36 and the increased protein levels of FASN with enhanced enzyme activity. In addition, after an analysis of the serological indicators in the mouse model, it was found that the serum levels of FASN were reduced. In conclusion, the OA/PA-induced cellular model can be used to study the mechanism of FFA uptake, whereas the high-fat-diet-induced mouse model can be used to study the mechanism of FFA uptake and DNL. Combined treatment with CD36 and FASN may be more effective against NAFLD. FASN in the serum can be used as one of the indicators for the clinical diagnosis of NAFLD.
Collapse
Affiliation(s)
- Miao Zhang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Xue Bai
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Qian Du
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Jiaojiao Xu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Danqing Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Lei Chen
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Keting Dong
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Ziyue Chen
- School of Nursing, Capital Medical University, Beijing 100069, China;
| | - Jianhong Yang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| |
Collapse
|
24
|
Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, Yang JL. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J Hematol Oncol 2023; 16:103. [PMID: 37700339 PMCID: PMC10498649 DOI: 10.1186/s13045-023-01498-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Lipid metabolic reprogramming is an emerging hallmark of cancer. In order to sustain uncontrolled proliferation and survive in unfavorable environments that lack oxygen and nutrients, tumor cells undergo metabolic transformations to exploit various ways of acquiring lipid and increasing lipid oxidation. In addition, stromal cells and immune cells in the tumor microenvironment also undergo lipid metabolic reprogramming, which further affects tumor functional phenotypes and immune responses. Given that lipid metabolism plays a critical role in supporting cancer progression and remodeling the tumor microenvironment, targeting the lipid metabolism pathway could provide a novel approach to cancer treatment. This review seeks to: (1) clarify the overall landscape and mechanisms of lipid metabolic reprogramming in cancer, (2) summarize the lipid metabolic landscapes within stromal cells and immune cells in the tumor microenvironment, and clarify their roles in tumor progression, and (3) summarize potential therapeutic targets for lipid metabolism, and highlight the potential for combining such approaches with other anti-tumor therapies to provide new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Hao-Ran Jin
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zi-Jing Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ming-Jia Xi
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bi-Han Xia
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jin-Lin Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
25
|
Cremer J, Brohée L, Dupont L, Lefevre C, Peiffer R, Saarinen AM, Peulen O, Bindels L, Liu J, Colige A, Deroanne CF. Acidosis-induced regulation of adipocyte G0S2 promotes crosstalk between adipocytes and breast cancer cells as well as tumor progression. Cancer Lett 2023:216306. [PMID: 37442366 DOI: 10.1016/j.canlet.2023.216306] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023]
Abstract
Bidirectional interactions between cancer cells and their microenvironment govern tumor progression. Among the stromal cells in this microenvironment, adipocytes have been reported to upregulate cancer cell migration and invasion by producing fatty acids. Conversely, cancer cells alter adipocyte phenotype notably via increased lipolysis. We aimed to identify the mechanisms through which cancer cells trigger adipocyte lipolysis and evaluate the functional consequences on cancer progression. Here, we show that cancer cell-induced acidification of the extracellular medium strongly promotes preadipocyte lipolysis through a mechanism that does not involve lipophagy but requires adipose triglyceride lipase (ATGL) activity. This increased lipolysis is triggered mainly by attenuation of the G0/G1 switch gene 2 (G0S2)-induced inhibition of ATGL. G0S2-mediated regulation in preadipocytes affects their communication with breast cancer cells, modifying the phenotype of the cancer cells and increasing their resistance to chemotherapeutic agents in vitro. Furthermore, we demonstrate that the adipocyte-specific overexpression of G0S2 impairs mammary tumor growth and lung metastasis formation in vivo. Our results highlight the importance of acidosis in cancer cell-adipocyte crosstalk and identify G0S2 as the main regulator of cancer-induced lipolysis, regulating tumor establishment and spreading.
Collapse
Affiliation(s)
- Julie Cremer
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Laura Brohée
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Laura Dupont
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Avenue Mounier 73, B1.73.11, 1200, Brussels, Belgium
| | - Raphaël Peiffer
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Alicia M Saarinen
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona Scottsdale, AZ, USA
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Laure Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Avenue Mounier 73, B1.73.11, 1200, Brussels, Belgium
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium
| | - Christophe F Deroanne
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University of Liège, Avenue Hippocrate 13, 4000, Liège, Belgium.
| |
Collapse
|
26
|
Olszańska J, Pietraszek-Gremplewicz K, Domagalski M, Nowak D. Mutual impact of adipocytes and colorectal cancer cells growing in co-culture conditions. Cell Commun Signal 2023; 21:130. [PMID: 37316878 DOI: 10.1186/s12964-023-01155-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/29/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common malignancy worldwide. CRC cells are situated in an adipocyte-rich microenvironment, which leads to interactions between adipocytes and CRC cells. Upon exposure to cancer cells, adipocytes transform into cancer-associated adipocytes (CAAs), and as a result, they gain features that promote tumor progression. The aim of this research was to shed more light on the detailed role of interactions between adipocytes and CRC cells associated with cancer progression in the context of these alterations. METHODS To implement adipocyte-CRC cell interaction, a co-culture model was applied. The analyses mainly focused on the metabolic modifications within CAAs and CRC cells, as well as the proliferation and migration potential of CRC cells. The impact of CRC on adipocytes was investigated by qRT-PCR analysis and Oil Red O staining. Proliferation and migration of CRC cells upon co-culture were tested with videomicroscopy, XTT, and a wound healing assay. Metabolic changes within CAAs and CRC cells were investigated based on lipid droplet formation, cell cycle analysis, gene and protein expression by qRT-PCR, and western blotting techniques. RESULTS CRC cells induced reprogramming of adipocytes into CAAs, which was connected with downregulation of lipid droplet formation in CAAs and alteration in adipocyte features. CAAs showed decreased metabolism-related gene expression, phosphorylation of Akt, ERK kinases, STAT3, and lactate secretion in comparison to the control. CAAs also promoted the migration, proliferation, and lipid droplet accumulation of CRC cells. After co-culturing with adipocytes, there was a shift to the G2/M phase of the cell cycle according to the differences in cyclin expression. CONCLUSION There are complex bidirectional interactions between adipocytes and CRC cells that may be connected with the induction of CRC cell progression. Video Abstract.
Collapse
Affiliation(s)
- Joanna Olszańska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | | | - Mikołaj Domagalski
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| |
Collapse
|
27
|
Qiao X, Hu Z, Xiong F, Yang Y, Peng C, Wang D, Li X. Lipid metabolism reprogramming in tumor-associated macrophages and implications for therapy. Lipids Health Dis 2023; 22:45. [PMID: 37004014 PMCID: PMC10064535 DOI: 10.1186/s12944-023-01807-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
The tumormicroenvironment (TME) plays a key role in tumor progression. Tumor-associated macrophages (TAMs), which are natural immune cells abundantin the TME, are mainly divided into the anti-tumor M1 subtype and pro-tumor M2 subtype. Due to the high plasticity of TAMs, the conversion of the M1 to M2 phenotype in hypoxic and hypoglycemic TME promotes cancer progression, which is closely related to lipid metabolism. Key factors of lipid metabolism in TAMs, including peroxisome proliferator-activated receptor and lipoxygenase, promote the formation of a tumor immunosuppressive microenvironment and facilitate immune escape. In addition, tumor cells promote lipid accumulation in TAMs, causing TAMs to polarize to the M2 phenotype. Moreover, other factors of lipid metabolism, such as abhydrolase domain containing 5 and fatty acid binding protein, have both promoting and inhibiting effects on tumor cells. Therefore, further research on lipid metabolism in tumors is still required. In addition, statins, as core drugs regulating cholesterol metabolism, can inhibit lipid rafts and adhesion of tumor cells, which can sensitize them to chemotherapeutic drugs. Clinical studies on simvastatin and lovastatin in a variety of tumors are underway. This article provides a comprehensive review of the role of lipid metabolism in TAMs in tumor progression, and provides new ideas for targeting lipid metabolism in tumor therapy.
Collapse
Affiliation(s)
- Xuehan Qiao
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhangmin Hu
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Fen Xiong
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yufei Yang
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chen Peng
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Deqiang Wang
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
28
|
Rebeaud M, Bouche C, Dauvillier S, Attané C, Arellano C, Vaysse C, Fallone F, Muller C. A novel 3D culture model for human primary mammary adipocytes to study their metabolic crosstalk with breast cancer in lean and obese conditions. Sci Rep 2023; 13:4707. [PMID: 36949082 PMCID: PMC10033714 DOI: 10.1038/s41598-023-31673-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023] Open
Abstract
Obesity is a negative prognosis factor for breast cancer. Yet, the biological mechanisms underlying this effect are still largely unknown. An emerging hypothesis is that the transfer of free fatty acids (FFA) between adipocytes and tumor cells might be altered under obese conditions, contributing to tumor progression. Currently there is a paucity of models to study human mammary adipocytes (M-Ads)-cancer crosstalk. As for other types of isolated white adipocytes, herein, we showed that human M-Ads die within 2-3 days by necrosis when grown in 2D. As an alternative, M-Ads were grown in a fibrin matrix, a 3D model that preserve their distribution, integrity and metabolic function for up to 5 days at physiological glucose concentrations (5 mM). Higher glucose concentrations frequently used in in vitro models promote lipogenesis during M-Ads culture, impairing their lipolytic function. Using transwell inserts, the matrix embedded adipocytes were cocultured with breast cancer cells. FFA transfer between M-Ads and cancer cells was observed, and this event was amplified by obesity. Together these data show that our 3D model is a new tool for studying the effect of M-Ads on tumor cells and beyond with all the components of the tumor microenvironment including the immune cells.
Collapse
Affiliation(s)
- Marie Rebeaud
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Caroline Bouche
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
- Département de Chirurgie Gynécologique oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France
| | - Stéphanie Dauvillier
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Camille Attané
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Carlo Arellano
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
- Département de Chirurgie Gynécologique oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France
| | - Charlotte Vaysse
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
- Département de Chirurgie Gynécologique oncologique, CHU-Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, 1 avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France
| | - Frédérique Fallone
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie et de Biologie Structurale, CNRS/Université de Toulouse UMR 5089, 205 route de Narbonne, BP 64182, 31077, Toulouse, France.
| |
Collapse
|
29
|
Mei S, Chen X, Wang K, Chen Y. Tumor microenvironment in ovarian cancer peritoneal metastasis. Cancer Cell Int 2023; 23:11. [PMID: 36698173 PMCID: PMC9875479 DOI: 10.1186/s12935-023-02854-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common gynecological malignancies with high morbidity and mortality. The peritoneum is one of the most common metastatic sites in ovarian cancer, involving large amounts of ascites. However, its mechanism is unclear. The peritoneal microenvironment composed of peritoneal effusion and peritoneum creates favorable conditions for ovarian cancer progression and metastasis. Here, we reviewed the peritoneal metastasis patterns and molecular mechanisms of ovarian cancer, as well as major components of the peritoneal microenvironment, peritoneal effusion, and immune microenvironment, and investigated the relationship between the peritoneal microenvironment and ovarian cancer metastasis.
Collapse
Affiliation(s)
- Shuangshuang Mei
- grid.469636.8Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Xi Men Road, Taizhou, 317000 Zhejiang China
| | - Xing Chen
- grid.469636.8Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Xi Men Road, Taizhou, 317000 Zhejiang China
| | - Kai Wang
- grid.469636.8Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Xi Men Road, Taizhou, 317000 Zhejiang China
| | - Yuxin Chen
- grid.469636.8Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University (Enze Hospital, Taizhou Enze Medical Center Group), Tong Yang Road, Taizhou, 318053 Zhejiang China
| |
Collapse
|
30
|
Na H, Song Y, Lee HW. Emphasis on Adipocyte Transformation: Anti-Inflammatory Agents to Prevent the Development of Cancer-Associated Adipocytes. Cancers (Basel) 2023; 15:cancers15020502. [PMID: 36672449 PMCID: PMC9856688 DOI: 10.3390/cancers15020502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Of the various cell types in the tumor microenvironment (TME), adipocytes undergo a dynamic transformation when activated by neighboring cancer cells. Although these adipocytes, known as cancer-associated adipocytes (CAAs), have been reported to play a crucial role in tumor progression, the factors that mediate their transformation remain elusive. In this review, we discuss the hypothesis that inflammatory signals involving NF-ĸB activation can induce lipolysis and adipocyte dedifferentiation. This provides a mechanistic understanding of CAA formation and introduces the concept of preventing adipocyte transformation via anti-inflammatory agents. Indeed, epidemiological studies indicate a higher efficacy of nonsteroidal anti-inflammatory drugs (NSAIDs) in obese patients with cancer, suggesting that NSAIDs can modulate the TME. Inhibition of cyclooxygenase-2 (COX-2) and prostaglandin production leads to the suppression of inflammatory signals such as NF-ĸB. Thus, we suggest the use of NSAIDs in cancer patients with metabolic disorders to prevent the transformation of TME components. Moreover, throughout this review, we attempt to expand our knowledge of CAA transformation to improve the clinical feasibility of targeting CAAs.
Collapse
Affiliation(s)
- Heeju Na
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yaechan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Gemcro Corporation, Seoul 03722, Republic of Korea
- Correspondence: ; Tel.: +82-2-2123-7642
| |
Collapse
|
31
|
Lipid Metabolism Heterogeneity and Crosstalk with Mitochondria Functions Drive Breast Cancer Progression and Drug Resistance. Cancers (Basel) 2022; 14:cancers14246267. [PMID: 36551752 PMCID: PMC9776509 DOI: 10.3390/cancers14246267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is a heterogeneous disease that can be triggered by genetic alterations in mammary epithelial cells, leading to diverse disease outcomes in individual patients. The metabolic heterogeneity of BC enhances its ability to adapt to changes in the tumor microenvironment and metabolic stress, but unfavorably affects the patient's therapy response, prognosis and clinical effect. Extrinsic factors from the tumor microenvironment and the intrinsic parameters of cancer cells influence their mitochondrial functions, which consequently alter their lipid metabolism and their ability to proliferate, migrate and survive in a harsh environment. The balanced interplay between mitochondria and fatty acid synthesis or fatty acid oxidation has been attributed to a combination of environmental factors and to the genetic makeup, oncogenic signaling and activities of different transcription factors. Hence, understanding the mechanisms underlying lipid metabolic heterogeneity and alterations in BC is gaining interest as a major target for drug resistance. Here we review the major recent reports on lipid metabolism heterogeneity and bring to light knowledge on the functional contribution of diverse lipid metabolic pathways to breast tumorigenesis and therapy resistance.
Collapse
|
32
|
Ennis CS, Llevenes P, Qiu Y, Dries R, Denis GV. The crosstalk within the breast tumor microenvironment in type II diabetes: Implications for cancer disparities. Front Endocrinol (Lausanne) 2022; 13:1044670. [PMID: 36531496 PMCID: PMC9751481 DOI: 10.3389/fendo.2022.1044670] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
Obesity-driven (type 2) diabetes (T2D), the most common metabolic disorder, both increases the incidence of all molecular subtypes of breast cancer and decreases survival in postmenopausal women. Despite this clear link, T2D and the associated dysfunction of diverse tissues is often not considered during the standard of care practices in oncology and, moreover, is treated as exclusion criteria for many emerging clinical trials. These guidelines have caused the biological mechanisms that associate T2D and breast cancer to be understudied. Recently, it has been illustrated that the breast tumor microenvironment (TME) composition and architecture, specifically the surrounding cellular and extracellular structures, dictate tumor progression and are directly relevant for clinical outcomes. In addition to the epithelial cancer cell fraction, the breast TME is predominantly made up of cancer-associated fibroblasts, adipocytes, and is often infiltrated by immune cells. During T2D, signal transduction among these cell types is aberrant, resulting in a dysfunctional breast TME that communicates with nearby cancer cells to promote oncogenic processes, cancer stem-like cell formation, pro-metastatic behavior and increase the risk of recurrence. As these cells are non-malignant, despite their signaling abnormalities, data concerning their function is never captured in DNA mutational databases, thus we have limited insight into mechanism from publicly available datasets. We suggest that abnormal adipocyte and immune cell exhaustion within the breast TME in patients with obesity and metabolic disease may elicit greater transcriptional plasticity and cellular heterogeneity within the expanding population of malignant epithelial cells, compared to the breast TME of a non-obese, metabolically normal patient. These challenges are particularly relevant to cancer disparities settings where the fraction of patients seen within the breast medical oncology practice also present with co-morbid obesity and metabolic disease. Within this review, we characterize the changes to the breast TME during T2D and raise urgent molecular, cellular and translational questions that warrant further study, considering the growing prevalence of T2D worldwide.
Collapse
Affiliation(s)
- Christina S. Ennis
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
| | - Pablo Llevenes
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Yuhan Qiu
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
| | - Ruben Dries
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, United States
| | - Gerald V. Denis
- Boston University-Boston Medical Center Cancer Center, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Shipley Prostate Cancer Research Professor, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
33
|
Schoutrop E, Moyano-Galceran L, Lheureux S, Mattsson J, Lehti K, Dahlstrand H, Magalhaes I. Molecular, cellular and systemic aspects of epithelial ovarian cancer and its tumor microenvironment. Semin Cancer Biol 2022; 86:207-223. [PMID: 35395389 DOI: 10.1016/j.semcancer.2022.03.027] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/11/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Ovarian cancer encompasses a heterogeneous group of malignancies that involve the ovaries, fallopian tubes and the peritoneal cavity. Despite major advances made within the field of cancer, the majority of patients with ovarian cancer are still being diagnosed at an advanced stage of the disease due to lack of effective screening tools. The overall survival of these patients has, therefore, not substantially improved over the past decades. Most patients undergo debulking surgery and treatment with chemotherapy, but often micrometastases remain and acquire resistance to the therapy, eventually leading to disease recurrence. Here, we summarize the current knowledge in epithelial ovarian cancer development and metastatic progression. For the most common subtypes, we focus further on the properties and functions of the immunosuppressive tumor microenvironment, including the extracellular matrix. Current and future treatment modalities are discussed and finally we provide an overview of the different experimental models used to develop novel therapies.
Collapse
Affiliation(s)
- Esther Schoutrop
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Stephanie Lheureux
- University of Toronto, Toronto, Ontario, Canada; Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jonas Mattsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; University of Toronto, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, Toronto, Ontario, Canada
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hanna Dahlstrand
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Medical unit Pelvic Cancer, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden.
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
34
|
Xiong X, Hasani S, Young LEA, Rivas DR, Skaggs AT, Martinez R, Wang C, Weiss HL, Gentry MS, Sun RC, Gao T. Activation of Drp1 promotes fatty acids-induced metabolic reprograming to potentiate Wnt signaling in colon cancer. Cell Death Differ 2022; 29:1913-1927. [PMID: 35332310 PMCID: PMC9525627 DOI: 10.1038/s41418-022-00974-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 11/09/2022] Open
Abstract
Cancer cells are known for their ability to adapt variable metabolic programs depending on the availability of specific nutrients. Our previous studies have shown that uptake of fatty acids alters cellular metabolic pathways in colon cancer cells to favor fatty acid oxidation. Here, we show that fatty acids activate Drp1 to promote metabolic plasticity in cancer cells. Uptake of fatty acids (FAs) induces mitochondrial fragmentation by promoting ERK-dependent phosphorylation of Drp1 at the S616 site. This increased phosphorylation of Drp1 enhances its dimerization and interaction with Mitochondrial Fission Factor (MFF) at the mitochondria. Consequently, knockdown of Drp1 or MFF attenuates fatty acid-induced mitochondrial fission. In addition, uptake of fatty acids triggers mitophagy via a Drp1- and p62-dependent mechanism to protect mitochondrial integrity. Moreover, results from metabolic profiling analysis reveal that silencing Drp1 disrupts cellular metabolism and blocks fatty acid-induced metabolic reprograming by inhibiting fatty acid utilization. Functionally, knockdown of Drp1 decreases Wnt/β-catenin signaling by preventing fatty acid oxidation-dependent acetylation of β-catenin. As a result, Drp1 depletion inhibits the formation of tumor organoids in vitro and xenograft tumor growth in vivo. Taken together, our study identifies Drp1 as a key mediator that connects mitochondrial dynamics with fatty acid metabolism and cancer cell signaling.
Collapse
Affiliation(s)
- Xiaopeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Sumati Hasani
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Lyndsay E A Young
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Dylan R Rivas
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Ashley T Skaggs
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Rebecca Martinez
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Heidi L Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Ramon C Sun
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, 40536-0679, USA
| | - Tianyan Gao
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536-0679, USA.
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40536-0679, USA.
| |
Collapse
|
35
|
Interplay between Prostate Cancer and Adipose Microenvironment: A Complex and Flexible Scenario. Int J Mol Sci 2022; 23:ijms231810762. [PMID: 36142673 PMCID: PMC9500873 DOI: 10.3390/ijms231810762] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Adipose tissue is part of the prostate cancer (PCa) microenvironment not only in the periprostatic area, but also in the most frequent metastatic sites, such as bone marrow and pelvic lymph nodes. The involvement of periprostatic adipose tissue (PPAT) in the aggressiveness of PCa is strongly suggested by numerous studies. Many molecules play a role in the reciprocal interaction between adipocytes and PCa cells, including adipokines, hormones, lipids, and also lipophilic pollutants stored in adipocytes. The crosstalk has consequences not only on cancer cell growth and metastatic potential, but also on adipocytes. Although most of the molecules released by PPAT are likely to promote tumor growth and the migration of cancer cells, others, such as the adipokine adiponectin and the n-6 or n-3 polyunsaturated fatty acids (PUFAs), have been shown to have anti-tumor properties. The effects of PPAT on PCa cells might therefore depend on the balance between the pro- and anti-tumor components of PPAT. In addition, genetic and environmental factors involved in the risk and/or aggressiveness of PCa, including obesity and diet, are able to modulate the interactions between PPAT and cancer cells and their consequences on the growth and the metastatic potential of PCa.
Collapse
|
36
|
Cui MY, Yi X, Cao ZZ, Zhu DX, Wu J. Targeting Strategies for Aberrant Lipid Metabolism Reprogramming and the Immune Microenvironment in Esophageal Cancer: A Review. JOURNAL OF ONCOLOGY 2022; 2022:4257359. [PMID: 36106333 PMCID: PMC9467784 DOI: 10.1155/2022/4257359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 12/24/2022]
Abstract
Esophageal cancer is of high importance to occurrence, development, and treatment resistance. As evidenced by recent studies, pathways (e.g., Wnt/β-catenin, AMPK, and Hippo) are critical to the proliferation, differentiation, and self-renewal of esophageal cancer. In addition, the above pathways play a certain role in regulating esophageal cancer and act as potential therapeutic targets. Over the past few years, the function of lipid metabolism in controlling tumor cells and immune cells has aroused extensive attention. It has been reported that there are intricate interactions between lipid metabolism reprogramming between immune and esophageal cancer cells, whereas molecular mechanisms should be studied in depth. Immune cells have been commonly recognized as a vital player in the esophageal cancer microenvironment, having complex crosstalk with cancer cells. It is increasingly evidenced that the function of immune cells in the tumor microenvironment (TME) is significantly correlated with abnormal lipid metabolism. In this review, the latest findings in lipid metabolism reprogramming in TME are summarized, and the above findings are linked to esophageal cancer progression. Aberrant lipid metabolism and associated signaling pathways are likely to serve as a novel strategy to treat esophageal cancer through lipid metabolism reprogramming.
Collapse
Affiliation(s)
- Meng-Ying Cui
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xing Yi
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Zhen-Zhen Cao
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Dan-Xia Zhu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jun Wu
- Department of Oncology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
37
|
Papakonstantinou E, Piperigkou Z, Karamanos NK, Zolota V. Altered Adipokine Expression in Tumor Microenvironment Promotes Development of Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:4139. [PMID: 36077676 PMCID: PMC9454958 DOI: 10.3390/cancers14174139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity is a remarkably important factor for breast carcinogenesis and aggressiveness. The implication of increased BMI in triple negative breast cancer (TNBC) development is also well established. A malignancy-promoting role of the adipose tissue has been supposed, where the adipocytes that constitute the majority of stromal cells release pro-inflammatory cytokines and growth factors. Alterations in adipokines and their receptors play significant roles in breast cancer initiation, progression, metastasis, and drug response. Classic adipokines, such as leptin, adiponectin, and resistin, have been extensively studied in breast cancer and connected with breast cancer risk and progression. Notably, new molecules are constantly being discovered and the list is continuously growing. Additionally, substantial progress has been made concerning their differential expression in association with clinical and pathological parameters of tumors and the prognostic and predictive value of their dysregulation in breast cancer carcinogenesis. However, evidence regarding the mechanisms by which adipose tissue is involved in the development of TNBC is lacking. In the present article we comment on current data on the suggested involvement of these mediators in breast cancer development and progression, with particular emphasis on TNBC, to draw attention to the design of novel targeted therapies and biomarkers.
Collapse
Affiliation(s)
- Efthymia Papakonstantinou
- Department of Gynecology and Obstetrics, School of Medicine, University of Patras, 26504 Patras, Greece or
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), 26504 Patras, Greece
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), 26504 Patras, Greece
| | - Vasiliki Zolota
- Department of Pathology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
38
|
Vasseur S, Guillaumond F. Lipids in cancer: a global view of the contribution of lipid pathways to metastatic formation and treatment resistance. Oncogenesis 2022; 11:46. [PMID: 35945203 PMCID: PMC9363460 DOI: 10.1038/s41389-022-00420-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/13/2022] Open
Abstract
Lipids are essential constituents for malignant tumors, as they are absolutely required for tumor growth and dissemination. Provided by the tumor microenvironment (TME) or by cancer cells themselves through activation of de novo synthesis pathways, they orchestrate a large variety of pro-tumorigenic functions. Importantly, TME cells, especially immune cells, cancer-associated fibroblasts (CAFs) and cancer-associated adipocytes (CAAs), are also prone to changes in their lipid content, which hinder or promote tumor aggressiveness. In this review, we address the significant findings for lipid contribution in tumor progression towards a metastatic disease and in the poor response to therapeutic treatments. We also highlight the benefits of targeting lipid pathways in preclinical models to slow down metastasis development and overcome chemo-and immunotherapy resistance.
Collapse
Affiliation(s)
- Sophie Vasseur
- Centre de Recherche en Cancérologie de Marseille, INSERM, Aix-Marseille Université, CNRS, Institut Paoli-Calmettes, F-13009, Marseille, France
| | - Fabienne Guillaumond
- Centre de Recherche en Cancérologie de Marseille, INSERM, Aix-Marseille Université, CNRS, Institut Paoli-Calmettes, F-13009, Marseille, France.
| |
Collapse
|
39
|
Zhang H, Yao Y, Wu J, Zhou J, Zhao C, He J, Xu B. Comprehensive Analysis Identifies and Validates the Tumor Microenvironment Subtypes to Predict Anti-Tumor Therapy Efficacy in Hepatocellular Carcinoma. Front Immunol 2022; 13:838374. [PMID: 35924239 PMCID: PMC9339643 DOI: 10.3389/fimmu.2022.838374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThe objective of this study was to explore and verify the subtypes in hepatocellular carcinoma based on the immune (lymphocyte and myeloid cells), stem, and stromal cells in the tumor microenvironment and analyze the biological characteristics and potential relevance of each cluster.MethodsWe used the xCell algorithm to calculate cell scores and got subtypes by k-means clustering. In the external validation sets, we verified the conclusion stability by a neural network model. Simultaneously, we speculated the inner connection between clusters by pseudotime trajectory analysis and confirmed it by pathway enrichment, TMB, CNV, etc., analysis.ResultAccording to the results of the consensus cluster, we chose k = 4 as the optimal value and got four different subtypes (C1, C2, C3, and C4) with different biological characteristics based on infiltrating levels of 48 cells in TME. In univariable Cox regression, the hazard ratio (HR) value of C3 versus C1 was 2.881 (95% CI: 1.572–5.279); in multivariable Cox regression, we corrected the age and TNM stage, and the HR value of C3 versus C1 was 2.510 (95% CI: 1.339–4.706). C1 and C2 belonged to the immune-active type, C3 and C4 related to the immune-insensitive type and the potential conversion relationships between clusters. We established a neural network model, and the area under the curves of the neural network model was 0.949 in the testing cohort; the same survival results were also observed in the external validation set. We compared the differences in cell infiltration, immune function, pathway enrichment, TMB, and CNV of four clusters and speculated that C1 and C2 were more likely to benefit from immunotherapy and C3 may benefit from FGF inhibitors.DiscussionOur analysis provides a new approach for the identification of four tumor microenvironment clusters in patients with liver cancer and identifies the biological differences and predicts the immunotherapy efficacy between the four subtypes.
Collapse
Affiliation(s)
- Haohan Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jin Zhou
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junju He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Bin Xu,
| |
Collapse
|
40
|
Hernandez M, Shin S, Muller C, Attané C. The role of bone marrow adipocytes in cancer progression: the impact of obesity. Cancer Metastasis Rev 2022; 41:589-605. [PMID: 35708800 DOI: 10.1007/s10555-022-10042-6] [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: 02/19/2022] [Accepted: 05/27/2022] [Indexed: 11/27/2022]
Abstract
Bone marrow adipose tissues (BMATs) and their main cellular component, bone marrow adipocytes (BMAds), are found within the bone marrow (BM), which is a niche for the development of hematological malignancies as well as bone metastasis from solid tumors such as breast and prostate cancers. In humans, BMAds are present within the hematopoietic or "red" BMAT and in the "yellow" BMAT where they are more densely packed. BMAds are emerging as new actors in tumor progression; however, there are many outstanding questions regarding their precise role. In this review, we summarized our current knowledge regarding the development, distribution, and regulation by external stimuli of the BMATs in mice and humans and addressed how obesity could affect these traits. We then discussed the specific metabolic phenotype of BMAds that appear to be different from "classical" white adipocytes, since they are devoid of lipolytic function. According to this characterization, we presented how tumor cells affect the in vitro and in vivo phenotype of BMAds and the signals emanating from BMAds that are susceptible to modulate tumor behavior with a specific emphasis on their metabolic crosstalk with cancer cells. Finally, we discussed how obesity could affect this crosstalk. Deciphering the role of BMAds in tumor progression would certainly lead to the identification of new targets in oncology in the near future.
Collapse
Affiliation(s)
- Marine Hernandez
- Institut de Pharmacologie Et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- Equipe Labellisée Ligue Contre Le Cancer, Toulouse, France
| | - Sauyeun Shin
- Institut de Pharmacologie Et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- Equipe Labellisée Ligue Contre Le Cancer, Toulouse, France
| | - Catherine Muller
- Institut de Pharmacologie Et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- Equipe Labellisée Ligue Contre Le Cancer, Toulouse, France.
| | - Camille Attané
- Institut de Pharmacologie Et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- Equipe Labellisée Ligue Contre Le Cancer, Toulouse, France.
| |
Collapse
|
41
|
Liu Y, Li C, Fang L, Wang L, Liu H, Tian H, zheng Y, Fan T, He J. Lipid metabolism-related lncRNA SLC25A21-AS1 promotes the progression of oesophageal squamous cell carcinoma by regulating the NPM1/c-Myc axis and SLC25A21 expression. Clin Transl Med 2022; 12:e944. [PMID: 35735113 PMCID: PMC9218933 DOI: 10.1002/ctm2.944] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Obesity alters metabolic microenvironment and is thus associated with several tumours. The aim of the present study was to investigate the role, molecular mechanism of action, and potential clinical value of lipid metabolism-related long non-coding RNA (lncRNA) SLC25A21-AS1 in oesophageal squamous cell carcinoma (ESCC). METHODS A high-fat diets (HFDs)-induced obesity nude mouse model was established, and targeted metabolomics analysis was used to identify critical medium-long chain fatty acids influencing the growth of ESCC cells. Transcriptomic analysis of public dataset GSE53625 confirmed that lncRNA SLC25A21-AS1 was a lipid metabolism-related lncRNA. The biological function of lncRNA SLC25A21-AS1 in ESCC was investigated both in vivo and in vitro. Chromatin immunoprecipitation(ChIP)assay, RNA-pull down, mass spectrometry, co-IP, and RNA IP(RIP) were performed to explore the molecular mechanism. Finally, an ESCC cDNA microarray was used to determine the clinical prognostic value of SLC25A21-AS1 by RT-qPCR. RESULTS Palmitic acid (PA) is an important fatty acid component of HFD and had an inhibitory effect on ESCC cell lines. LncRNA SLC25A21-AS1 expression was downregulated by PA and associated with the proliferation and migration of ESCC cells in vitro and in vivo. Mechanistically, SLC25A21-AS1 interacted with nucleophosmin-1 (NPM1) protein to promote the downstream gene transcription of the c-Myc in the nucleus. In the cytoplasm, SLC25A21-AS1 maintained the stability of SLC25A21 mRNA and reduced the intracellular NAD+ /NADH ratio by influencing tryptophan catabolism. Finally, we demonstrated that high expression of SLC25A21-AS1 promoted resistance to cisplatin-induced apoptosis and was correlated with poor tumour grade and overall survival. CONCLUSIONS HFD/PA has an inhibitory effect on ESCC cells and SLC25A21-AS1 expression. SLC25A21-AS1 promotes the proliferation and migration of ESCC cells by regulating the NPM1/c-Myc axis and SLC25A21 expression. In addition, lncRNA SLC25A21-AS1 may serve as a favourable prognostic biomarker and a potential therapeutic target for ESCC.
Collapse
Affiliation(s)
- Yu Liu
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chunxiang Li
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lingling Fang
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Liyu Wang
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hengchang Liu
- Department of Colorectal SurgeryNational Cancer Center/Natbibional Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - He Tian
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yujia zheng
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tao Fan
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jie He
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
42
|
Gonzalez Suarez N, Fernandez-Marrero Y, Torabidastgerdooei S, Annabi B. EGCG Prevents the Onset of an Inflammatory and Cancer-Associated Adipocyte-like Phenotype in Adipose-Derived Mesenchymal Stem/Stromal Cells in Response to the Triple-Negative Breast Cancer Secretome. Nutrients 2022; 14:1099. [PMID: 35268073 PMCID: PMC8912398 DOI: 10.3390/nu14051099] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) cells secretome induces a pro-inflammatory microenvironment within the adipose tissue, which hosts both mature adipocytes and adipose-derived mesenchymal stem/stromal cells (ADMSC). The subsequent acquisition of a cancer-associated adipocyte (CAA)-like phenotype is, however, unknown in ADMSC. While epidemiological studies suggest that consuming a polyphenol-rich diet reduces the incidence of some obesity-related cancers, the chemopreventive impact of green tea-derived epigallocatechin-3-gallate (EGCG) against the cues that trigger the CAA phenotype remain undocumented in ADMSC. METHODS Human ADMSC were exposed to human TNBC-derived MDA-MB-231 conditioned media (TNBC cells secretome) supplemented or not with EGCG. Differential gene expression was assessed through RNA-Seq analysis and confirmed by RT-qPCR. Protein expression levels and the activation status of signal transduction pathways mediators were determined by Western blotting. ADMSC chemotaxis was assessed by a real-time cell migration assay. RESULTS The TNBC cells secretome induced in ADMSC the expression of the CAA cytokines CCL2, CCL5, IL-1β, and IL-6, and of immunomodulators COX2, HIF-1α, VEGFα, and PD-L1. The epithelial-to-mesenchymal biomarker Snail was found to control the CAA phenotype. EGCG inhibited the induction of CAA genes and the activation status of Smad2 and NF-κB. The induced chemotactic response was also inhibited by EGCG. CONCLUSION The induction of an inflammatory and CAA-like phenotype in ADMSC can be triggered by the TNBC cells secretome, while still efficiently prevented by diet-derived polyphenols.
Collapse
Affiliation(s)
- Narjara Gonzalez Suarez
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (N.G.S.); (S.T.)
| | - Yuniel Fernandez-Marrero
- Biological Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Science Centre, Toronto, ON M4N 3M5, Canada;
| | - Sima Torabidastgerdooei
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (N.G.S.); (S.T.)
| | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H3C 3P8, Canada; (N.G.S.); (S.T.)
| |
Collapse
|
43
|
Du D, Liu C, Qin M, Zhang X, Xi T, Yuan S, Hao H, Xiong J. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B 2022; 12:558-580. [PMID: 35256934 PMCID: PMC8897153 DOI: 10.1016/j.apsb.2021.09.019] [Citation(s) in RCA: 317] [Impact Index Per Article: 105.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive human cancer with increasing incidence worldwide. Multiple efforts have been made to explore pharmaceutical therapies to treat HCC, such as targeted tyrosine kinase inhibitors, immune based therapies and combination of chemotherapy. However, limitations exist in current strategies including chemoresistance for instance. Tumor initiation and progression is driven by reprogramming of metabolism, in particular during HCC development. Recently, metabolic associated fatty liver disease (MAFLD), a reappraisal of new nomenclature for non-alcoholic fatty liver disease (NAFLD), indicates growing appreciation of metabolism in the pathogenesis of liver disease, including HCC, thereby suggesting new strategies by targeting abnormal metabolism for HCC treatment. In this review, we introduce directions by highlighting the metabolic targets in glucose, fatty acid, amino acid and glutamine metabolism, which are suitable for HCC pharmaceutical intervention. We also summarize and discuss current pharmaceutical agents and studies targeting deregulated metabolism during HCC treatment. Furthermore, opportunities and challenges in the discovery and development of HCC therapy targeting metabolism are discussed.
Collapse
Key Words
- 1,3-BPG, 1,3-bisphosphoglycerate
- 2-DG, 2-deoxy-d-glucose
- 3-BrPA, 3-bromopyruvic acid
- ACC, acetyl-CoA carboxylase
- ACLY, adenosine triphosphate (ATP) citrate lyase
- ACS, acyl-CoA synthease
- AKT, protein kinase B
- AML, acute myeloblastic leukemia
- AMPK, adenosine mono-phosphate-activated protein kinase
- ASS1, argininosuccinate synthase 1
- ATGL, adipose triacylglycerol lipase
- CANA, canagliflozin
- CPT, carnitine palmitoyl-transferase
- CYP4, cytochrome P450s (CYPs) 4 family
- Cancer therapy
- DNL, de novo lipogenesis
- EMT, epithelial-to-mesenchymal transition
- ER, endoplasmic reticulum
- ERK, extracellular-signal regulated kinase
- FABP1, fatty acid binding protein 1
- FASN, fatty acid synthase
- FBP1, fructose-1,6-bisphosphatase 1
- FFA, free fatty acid
- Fatty acid β-oxidation
- G6PD, glucose-6-phosphate dehydrogenase
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GLS1, renal-type glutaminase
- GLS2, liver-type glutaminase
- GLUT1, glucose transporter 1
- GOT1, glutamate oxaloacetate transaminase 1
- Glutamine metabolism
- Glycolysis
- HCC, hepatocellular carcinoma
- HIF-1α, hypoxia-inducible factor-1 alpha
- HK, hexokinase
- HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase
- HSCs, hepatic stellate cells
- Hepatocellular carcinoma
- IDH2, isocitrate dehydrogenase 2
- LCAD, long-chain acyl-CoA dehydrogenase
- LDH, lactate dehydrogenase
- LPL, lipid lipase
- LXR, liver X receptor
- MAFLD, metabolic associated fatty liver disease
- MAGL, monoacyglycerol lipase
- MCAD, medium-chain acyl-CoA dehydrogenase
- MEs, malic enzymes
- MMP9, matrix metallopeptidase 9
- Metabolic dysregulation
- NADPH, nicotinamide adenine nucleotide phosphate
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- OTC, ornithine transcarbamylase
- PCK1, phosphoenolpyruvate carboxykinase 1
- PFK1, phosphofructokinase 1
- PGAM1, phosphoglycerate mutase 1
- PGK1, phosphoglycerate kinase 1
- PI3K, phosphoinositide 3-kinase
- PKM2, pyruvate kinase M2
- PPARα, peroxisome proliferator-activated receptor alpha
- PPP, pentose phosphate pathway
- Pentose phosphate pathway
- ROS, reactive oxygen species
- SCD1, stearoyl-CoA-desaturase 1
- SGLT2, sodium-glucose cotransporter 2
- SLC1A5/ASCT2, solute carrier family 1 member 5/alanine serine cysteine preferring transporter 2
- SLC7A5/LAT1, solute carrier family 7 member 5/L-type amino acid transporter 1
- SREBP1, sterol regulatory element-binding protein 1
- TAGs, triacylglycerols
- TCA cycle, tricarboxylic acid cycle
- TKIs, tyrosine kinase inhibitors
- TKT, transketolase
- Tricarboxylic acid cycle
- VEGFR, vascular endothelial growth factor receptor
- WD-fed MC4R-KO, Western diet (WD)-fed melanocortin 4 receptor-deficient (MC4R-KO)
- WNT, wingless-type MMTV integration site family
- mIDH, mutant IDH
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Danyu Du
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Chan Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyao Qin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Zhang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Xi
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| |
Collapse
|
44
|
Iozzo M, Sgrignani G, Comito G, Chiarugi P, Giannoni E. Endocannabinoid System and Tumour Microenvironment: New Intertwined Connections for Anticancer Approaches. Cells 2021; 10:cells10123396. [PMID: 34943903 PMCID: PMC8699381 DOI: 10.3390/cells10123396] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 01/01/2023] Open
Abstract
The tumour microenvironment (TME) is now recognised as a hallmark of cancer, since tumour:stroma crosstalk supports the key steps of tumour growth and progression. The dynamic co-evolution of the tumour and stromal compartments may alter the surrounding microenvironment, including the composition in metabolites and signalling mediators. A growing number of evidence reports the involvement of the endocannabinoid system (ECS) in cancer. ECS is composed by a complex network of ligands, receptors, and enzymes, which act in synergy and contribute to several physiological but also pathological processes. Several in vitro and in vivo evidence show that ECS deregulation in cancer cells affects proliferation, migration, invasion, apoptosis, and metastatic potential. Although it is still an evolving research, recent experimental evidence also suggests that ECS can modulate the functional behaviour of several components of the TME, above all the immune cells, endothelial cells and stromal components. However, the role of ECS in the tumour:stroma interplay remains unclear and research in this area is particularly intriguing. This review aims to shed light on the latest relevant findings of the tumour response to ECS modulation, encouraging a more in-depth analysis in this field. Novel discoveries could be promising for novel anti-tumour approaches, targeting the microenvironmental components and the supportive tumour:stroma crosstalk, thereby hindering tumour development.
Collapse
|
45
|
Hoy AJ, Nagarajan SR, Butler LM. Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat Rev Cancer 2021; 21:753-766. [PMID: 34417571 DOI: 10.1038/s41568-021-00388-4] [Citation(s) in RCA: 207] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
Fatty acid metabolism is known to support tumorigenesis and disease progression as well as treatment resistance through enhanced lipid synthesis, storage and catabolism. More recently, the role of membrane fatty acid composition, for example, ratios of saturated, monounsaturated and polyunsaturated fatty acids, in promoting cell survival while limiting lipotoxicity and ferroptosis has been increasingly appreciated. Alongside these insights, it has become clear that tumour cells exhibit plasticity with respect to fatty acid metabolism, responding to extratumoural and systemic metabolic signals, such as obesity and cancer therapeutics, to promote the development of aggressive, treatment-resistant disease. Here, we describe cellular fatty acid metabolic changes that are connected to therapy resistance and contextualize obesity-associated changes in host fatty acid metabolism that likely influence the local tumour microenvironment to further modify cancer cell behaviour while simultaneously creating potential new vulnerabilities.
Collapse
Affiliation(s)
- Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | - Shilpa R Nagarajan
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
46
|
MCF-7 Drug Resistant Cell Lines Switch Their Lipid Metabolism to Triple Negative Breast Cancer Signature. Cancers (Basel) 2021; 13:cancers13235871. [PMID: 34884983 PMCID: PMC8657222 DOI: 10.3390/cancers13235871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity and adipose tissue have been closely related to a poor cancer prognosis, especially in prostate and breast cancer patients. The ability of transferring lipids from the adipose tissue to the tumor cells is actively linked to tumor progression. However, different types of breast tumor seem to use these lipids in different ways and metabolize them in different pathways. In this study we have tracked by mass spectrometry how palmitic acid from the adipocytes is released to media being later incorporated in different breast cancer cell lines (MDA-MB-231, SKBR3, BT474, MCF-7 and its resistant MCF-7 EPIR and MCF-7 TAXR). We have observed that different lines metabolize the palmitic acid in a different way and use their carbons in the synthesis of different new lipid families. Furthermore, we have observed that the lipid synthesis pattern varied according to the cell line. Surprisingly, the metabolic pattern of the resistant cells was more related to the TNBC cell line compared to their sensitive cell line MCF-7. These results allow us to determine a specific lipid pattern in different cell lines that later might be used in breast cancer diagnosis and to find a better treatment according to the cancer molecular type.
Collapse
|
47
|
Kostecka LG, Pienta KJ, Amend SR. Lipid droplet evolution gives insight into polyaneuploid cancer cell lipid droplet functions. Med Oncol 2021; 38:133. [PMID: 34581907 PMCID: PMC8478749 DOI: 10.1007/s12032-021-01584-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/12/2021] [Indexed: 12/16/2022]
Abstract
Lipid droplets (LDs) are found throughout all phyla across the tree of life. Originating as pure energy stores in the most basic organisms, LDs have evolved to fill various roles as regulators of lipid metabolism, signaling, and trafficking. LDs have been noted in cancer cells and have shown to increase tumor aggressiveness and chemotherapy resistance. A certain transitory state of cancer cell, the polyaneuploid cancer cell (PACC), appears to have higher LD levels than the cancer cell from which they are derived. PACCs are postulated to be the mediators of metastasis and resistance in many different cancers. Utilizing the evolutionarily conserved roles of LDs to protect from cellular lipotoxicity allows PACCs to survive otherwise lethal stressors. By better understanding how LDs have evolved throughout different phyla we will identify opportunities to target LDs in PACCs to increase therapeutic efficiency in cancer cells.
Collapse
Affiliation(s)
- Laurie G Kostecka
- The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, MD, 21287, USA. .,Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, MD, 21287, USA.,Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Sarah R Amend
- The Brady Urological Institute, Johns Hopkins School of Medicine, 600 N. Wolfe St., Marburg Building Room 113, Baltimore, MD, 21287, USA.,Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
48
|
Skeletal Muscle-Adipose Tissue-Tumor Axis: Molecular Mechanisms Linking Exercise Training in Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094469. [PMID: 33922898 PMCID: PMC8123194 DOI: 10.3390/ijms22094469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Increased visceral adiposity may influence the development of prostate cancer (PCa) aggressive tumors and cancer mortality. White adipose tissue (WAT), usually referred to as periprostatic adipose tissue (PPAT), surrounds the prostatic gland and has emerged as a potential mediator of the tumor microenvironment. Exercise training (ET) induces several adaptations in both skeletal muscle and WAT. Some of these effects are mediated by ET-induced synthesis and secretion of several proteins, known as myo- and adipokines. Together, myokines and adipokines may act in an endocrine-like manner to favor communication between skeletal muscle and WAT, as they may work together to improve whole-body metabolic health. This crosstalk may constitute a potential mechanism by which ET exerts its beneficial role in the prevention and treatment of PCa-related disorders; however, this has not yet been explored. Therefore, we reviewed the current evidence on the effects of skeletal muscle–WAT–tumor crosstalk in PCa, and the potential mediators of this process to provide a better understanding of underlying ET-related mechanisms in cancer.
Collapse
|
49
|
Cancer-Associated Adipocytes in Breast Cancer: Causes and Consequences. Int J Mol Sci 2021; 22:ijms22073775. [PMID: 33917351 PMCID: PMC8038661 DOI: 10.3390/ijms22073775] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer progression is highly dependent on the heterotypic interaction between tumor cells and stromal cells of the tumor microenvironment. Cancer-associated adipocytes (CAAs) are emerging as breast cancer cell partners favoring proliferation, invasion, and metastasis. This article discussed the intersection between extracellular signals and the transcriptional cascade that regulates adipocyte differentiation in order to appreciate the molecular pathways that have been described to drive adipocyte dedifferentiation. Moreover, recent studies on the mechanisms through which CAAs affect the progression of breast cancer were reviewed, including adipokine regulation, metabolic reprogramming, extracellular matrix remodeling, and immune cell modulation. An in-depth understanding of the complex vicious cycle between CAAs and breast cancer cells is crucial for designing novel strategies for new therapeutic interventions.
Collapse
|
50
|
The Chemokine Receptor CCR3 Is Potentially Involved in the Homing of Prostate Cancer Cells to Bone: Implication of Bone-Marrow Adipocytes. Int J Mol Sci 2021; 22:ijms22041994. [PMID: 33671469 PMCID: PMC7922974 DOI: 10.3390/ijms22041994] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/26/2022] Open
Abstract
Bone metastasis remains the most frequent and the deadliest complication of prostate cancer (PCa). Mechanisms leading to the homing of tumor cells to bone remain poorly characterized. Role of chemokines in providing navigational cues to migrating cancer cells bearing specific receptors is well established. Bone is an adipocyte-rich organ since 50 to 70% of the adult bone marrow (BM) volume comprise bone marrow adipocytes (BM-Ads), which are likely to produce chemokines within the bone microenvironment. Using in vitro migration assays, we demonstrated that soluble factors released by human primary BM-Ads are able to support the directed migration of PCa cells in a CCR3-dependent manner. In addition, we showed that CCL7, a chemokine previously involved in the CCR3-dependent migration of PCa cells outside of the prostate gland, is released by human BM-Ads. These effects are amplified by obesity and ageing, two clinical conditions known to promote aggressive and metastatic PCa. In human tumors, we found an enrichment of CCR3 in bone metastasis vs. primary tumors at mRNA levels using Oncomine microarray database. In addition, immunohistochemistry experiments demonstrated overexpression of CCR3 in bone versus visceral metastases. These results underline the potential importance of BM-Ads in the bone metastatic process and imply a CCR3/CCL7 axis whose pharmacological interest needs to be evaluated.
Collapse
|