1
|
Solsona‐Vilarrasa E, Vousden KH. Obesity, white adipose tissue and cancer. FEBS J 2025; 292:2189-2207. [PMID: 39496581 PMCID: PMC12062788 DOI: 10.1111/febs.17312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
2
|
Poswal J, Mandal CC. Lipid metabolism dysregulation for bone metastasis and its prevention. Expert Rev Anticancer Ther 2025:1-17. [PMID: 40219980 DOI: 10.1080/14737140.2025.2492784] [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: 02/16/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Bone metastasis often develops in advanced malignancies. Lipid metabolic dysregulation might play pivotal role in cancer progression and subsequent deterioration of bone health at metastatic condition. In-depth understanding of lipid reprogramming in metastasized cancer cells and other stromal cells including bone marrow adipocyte (BMA) is an urgent need to develop effective therapy. AREA COVERED This paper emphasizes providing an overview of multifaceted role of dysregulated lipids and BMA in cancer cells in association with bone metastasis by utilizing search terms lipid metabolism, lipid and metastasis in PubMed. This study extends to address mechanism linked with lipid metabolism and various crucial genes (e.g. CSF-1, RANKL, NFkB and NFATc1) involved in bone metastasis. This review examines therapeutic strategies targeting lipid metabolism to offer potential avenues to disrupt lipid-driven metastasis. EXPERT OPINION On metastatic condition, dysregulated lipid molecules especially in BMA and other stromal cells not only favors cancer progression but also potentiate lipid reprogramming within cancer cells. Distinct dysregulated lipid-metabolism associated genes may act as biomarker, and targeting these is challenging task for specific treatment. Curbing function of bone resorption associated genes by lipid controlling drugs (e.g. statins, omega-3 FA and metformin) may provide additional support to curtail lipid-associated bone metastasis.
Collapse
Affiliation(s)
- Jyoti Poswal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
3
|
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
|
4
|
Liu H, Liu L, Rosen CJ. Bone Marrow Adipocytes as Novel Regulators of Metabolic Homeostasis: Clinical Consequences of Bone Marrow Adiposity. Curr Obes Rep 2025; 14:9. [PMID: 39808256 DOI: 10.1007/s13679-024-00594-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW Bone marrow adipose tissue is a distinctive fat depot located within the skeleton, with the potential to influence both local and systemic metabolic processes. Although significant strides have been made in understanding bone marrow adipose tissue over the past decade, many questions remain regarding their precise lineage and functional roles. RECENT FINDINGS Recent studies have highlighted bone marrow adipose tissue's involvement in continuous cross-talk with other organs and systems, exerting both endocrine and paracrine functions that play a crucial role in metabolic homeostasis, skeletal remodeling, hematopoiesis, and the progression of bone metastases. The advancement of imaging techniques, particularly cross-sectional imaging, has profoundly expanded our understanding of the complexities beyond the traditional view of bone marrow adipose tissue as an inert depot. Notably, marrow adipocytes are anatomically and functionally distinct from brown, beige, and classic white adipocytes. Emerging evidence suggests that bone marrow adipocytes, bone marrow adipose tissue originate from the differentiation of bone marrow mesenchymal stromal cells; however, they appear to be a heterogeneous population with varying metabolic profiles, lipid compositions, secretory properties, and functional responses depending on their specific location within the bone marrow. This review provides an up-to-date synthesis of current knowledge on bone marrow adipocytes, emphasizing the relationships between bone marrow adipogenesis and factors such as aging, osteoporosis, obesity, and bone marrow tumors or metastases, thereby elucidating the mechanisms underlying musculoskeletal pathophysiology.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology &, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Linyi Liu
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA.
| |
Collapse
|
5
|
Lin W, Li Y, Qiu C, Zou B, Gong Y, Zhang X, Tian D, Sherman W, Sanchez F, Wu D, Su KJ, Xiao X, Luo Z, Tian Q, Chen Y, Shen H, Deng H. Mapping the spatial atlas of the human bone tissue integrating spatial and single-cell transcriptomics. Nucleic Acids Res 2025; 53:gkae1298. [PMID: 39817519 PMCID: PMC11736439 DOI: 10.1093/nar/gkae1298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
Bone is a multifaceted tissue requiring orchestrated interplays of diverse cells within specialized microenvironments. Although significant progress has been made in understanding cellular and molecular mechanisms of component cells of bone, revealing their spatial organization and interactions in native bone tissue microenvironment is crucial for advancing precision medicine, as they govern fundamental signaling pathways and functional dependencies among various bone cells. In this study, we present the first integrative high-resolution map of human bone and bone marrow, using spatial and single-cell transcriptomics profiling from femoral tissue. This multi-modal approach discovered a novel bone formation-specialized niche enriched with osteoblastic lineage cells and fibroblasts and unveiled critical cell-cell communications and co-localization patterns between osteoblastic lineage cells and other cells. Furthermore, we discovered a novel spatial gradient of cellular composition, gene expression and signaling pathway activities radiating from the trabecular bone. This comprehensive atlas delineates the intricate bone cellular architecture and illuminates key molecular processes and dependencies among cells that coordinate bone metabolism. In sum, our study provides an essential reference for the field of bone biology and lays the foundation for advanced mechanistic studies and precision medicine approaches in bone-related disorders.
Collapse
Affiliation(s)
- Weiqiang Lin
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Yisu Li
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6823 St. Charles Avenue, Uptown, New Orleans, LA 70118, USA
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Binghao Zou
- Department of Structural and Cellular Biology, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Yun Gong
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Xiao Zhang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Di Tian
- The Molecular Pathology Laboratory, Department of Pathology and Laboratory Medicine, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - William Sherman
- Department of Orthopaedic Surgery, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Fernando Sanchez
- Department of Orthopaedic Surgery, School of Medicine, Tulane University, 1430 Tulane Avenue, Downtown, New Orleans, LA 70112, USA
| | - Di Wu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Xinyi Xiao
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Zhe Luo
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Yiping Chen
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, 6823 St. Charles Avenue, Uptown, New Orleans, LA 70118, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| | - Hongwen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, 1440 Canal Street, Downtown, New Orleans, LA 70112, USA
| |
Collapse
|
6
|
Hindes MT, McElligott AM, Best OG, Ward MP, Selemidis S, Miles MA, Nturubika BD, Gregory PA, Anderson PH, Logan JM, Butler LM, Waugh DJ, O'Leary JJ, Hickey SM, Thurgood LA, Brooks DA. Metabolic reprogramming, malignant transformation and metastasis: Lessons from chronic lymphocytic leukaemia and prostate cancer. Cancer Lett 2025; 611:217441. [PMID: 39755364 DOI: 10.1016/j.canlet.2025.217441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer, crucial for malignant transformation and metastasis. Chronic lymphocytic leukaemia (CLL) and prostate cancer exhibit similar metabolic adaptations, particularly in glucose and lipid metabolism. Understanding this metabolic plasticity is crucial for identifying mechanisms contributing to metastasis. This review considers glucose and lipid metabolism in CLL and prostate cancer, exploring their roles in healthy and malignant states and during disease progression. In CLL, lipid metabolism supports cell survival and migration, with aggressive disease characterised by increased fatty acid oxidation and altered sphingolipids. Richter's transformation and aggressive lymphoma, however, exhibit a metabolic shift towards increased glycolysis. Similarly, prostate cell metabolism is unique, relying on citrate production in the healthy state and undergoing metabolic reprogramming during malignant transformation. Early-stage prostate cancer cells increase lipid synthesis and uptake, and decrease glycolysis, whereas metastatic cells re-adopt glucose metabolism, likely driven by interactions with the tumour microenvironment. Genetic drivers including TP53 and ATM mutations connect metabolic alterations to disease severity in these two malignancies. The bone microenvironment supports the metabolic demands of these malignancies, serving as an initiation niche for CLL and a homing site for prostate cancer metastases. By comparing these malignancies, this review underscores the importance of metabolic plasticity in cancer progression and highlights how CLL and prostate cancer may be models of circulating and solid tumours more broadly. The metabolic phenotypes throughout cancer cell transformation and metastasis, and the microenvironment in which these processes occur, present opportunities for interventions that could disrupt metastatic processes and improve patient outcomes.
Collapse
Affiliation(s)
- Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Anthony M McElligott
- Discipline of Haematology, School of Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College, Dublin, Ireland
| | - Oliver G Best
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Bukuru D Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Paul H Anderson
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, Australia; Solid Tumour Program, Precision Cancer Medicine theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - David J Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lauren A Thurgood
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
7
|
Liermann-Wooldrik KT, Kosmacek EA, Oberley-Deegan RE. Adipose Tissues Have Been Overlooked as Players in Prostate Cancer Progression. Int J Mol Sci 2024; 25:12137. [PMID: 39596205 PMCID: PMC11594286 DOI: 10.3390/ijms252212137] [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: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a common risk factor in multiple tumor types, including prostate cancer. Obesity has been associated with driving metastasis, therapeutic resistance, and increased mortality. The effect of adipose tissue on the tumor microenvironment is still poorly understood. This review aims to highlight the work conducted in the field of obesity and prostate cancer and bring attention to areas where more research is needed. In this review, we have described key differences between healthy adipose tissues and obese adipose tissues, as they relate to the tumor microenvironment, focusing on mechanisms related to metabolic changes, abnormal adipokine secretion, altered immune cell presence, and heightened oxidative stress as drivers of prostate cancer formation and progression. Interestingly, common treatment options for prostate cancer ignore the adipose tissue located near the site of the tumor. Because of this, we have outlined how excess adipose tissue potentially affects therapeutics' efficacy, such as androgen deprivation, chemotherapy, and radiation treatment, and identified possible drug targets to increase prostate cancer responsiveness to clinical treatments. Understanding how obesity affects the tumor microenvironment will pave the way for understanding why some prostate cancers become metastatic or treatment-resistant, and why patients experience recurrence.
Collapse
Affiliation(s)
| | | | - Rebecca E. Oberley-Deegan
- Department of Biochemistry and Molecular Biology, 985870 University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.T.L.-W.)
| |
Collapse
|
8
|
Linke JA, Munn LL, Jain RK. Compressive stresses in cancer: characterization and implications for tumour progression and treatment. Nat Rev Cancer 2024; 24:768-791. [PMID: 39390249 DOI: 10.1038/s41568-024-00745-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Beyond their many well-established biological aberrations, solid tumours create an abnormal physical microenvironment that fuels cancer progression and confers treatment resistance. Mechanical forces impact tumours across a range of biological sizes and timescales, from rapid events at the molecular level involved in their sensing and transmission, to slower and larger-scale events, including clonal selection, epigenetic changes, cell invasion, metastasis and immune response. Owing to challenges with studying these dynamic stimuli in biological systems, the mechanistic understanding of the effects and pathways triggered by abnormally elevated mechanical forces remains elusive, despite clear correlations with cancer pathophysiology, aggressiveness and therapeutic resistance. In this Review, we examine the emerging and diverse roles of physical forces in solid tumours and provide a comprehensive framework for understanding solid stress mechanobiology. We first review the physiological importance of mechanical forces, especially compressive stresses, and discuss their defining characteristics, biological context and relative magnitudes. We then explain how abnormal compressive stresses emerge in tumours and describe the experimental challenges in investigating these mechanically induced processes. Finally, we discuss the clinical translation of mechanotherapeutics that alleviate solid stresses and their potential to synergize with chemotherapy, radiotherapy and immunotherapies.
Collapse
Affiliation(s)
- Julia A Linke
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Dobrovinskaya O, Alamilla J, Olivas-Aguirre M. Impact of Modern Lifestyle on Circadian Health and Its Contribution to Adipogenesis and Cancer Risk. Cancers (Basel) 2024; 16:3706. [PMID: 39518143 PMCID: PMC11545514 DOI: 10.3390/cancers16213706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Recent research underscores a crucial connection between circadian rhythm disruption and cancer promotion, highlighting an urgent need for attention. OBJECTIVES Explore the molecular mechanisms by which modern lifestyle factors-such as artificial light exposure, shift work, and dietary patterns-affect cortisol/melatonin regulation and cancer risk. METHODS Employing a narrative review approach, we synthesized findings from Scopus, Google Scholar, and PubMed to analyze lifestyle impacts on circadian health, focusing on cortisol and melatonin chronobiology as molecular markers. We included studies that documented quantitative changes in these markers due to modern lifestyle habits, excluding those lacking quantitative data or presenting inconclusive results. Subsequent sections focused solely on articles that quantified the effects of circadian disruption on adipogenesis and tumor microenvironment modifications. RESULTS This review shows how modern habits lead to molecular changes in cortisol and melatonin, creating adipose microenvironments that support cancer development. These disruptions facilitate immune evasion, chemotherapy resistance, and tumor growth, highlighting the critical roles of cortisol dysregulation and melatonin imbalance. CONCLUSIONS Through the presented findings, we establish a causal link between circadian rhythm dysregulation and the promotion of certain cancer types. By elucidating this relationship, the study emphasizes the importance of addressing lifestyle factors that contribute to circadian misalignment, suggesting that targeted interventions could play a crucial role in mitigating cancer risk and improving overall health outcomes.
Collapse
Affiliation(s)
- Oxana Dobrovinskaya
- Laboratory of Immunobiology and Ionic Transport Regulation, University Center for Biomedical Research, University of Colima, Colima 28040, Mexico;
| | - Javier Alamilla
- Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT), Programa de Investigadores e Investigadoras por México, México City 03940, Mexico;
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima 28040, Mexico
| | - Miguel Olivas-Aguirre
- Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT), Programa de Investigadores e Investigadoras por México, México City 03940, Mexico;
- Laboratory of Cancer Pathophysiology, University Center for Biomedical Research, University of Colima, Colima 28040, Mexico
| |
Collapse
|
10
|
Li Y, Wang L, Wang J, Xin Y, Lyu X. Relationship between adipocytes and hematological tumors in the bone marrow microenvironment: a literature review. Transl Cancer Res 2024; 13:5691-5701. [PMID: 39525009 PMCID: PMC11543051 DOI: 10.21037/tcr-24-52] [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: 01/09/2024] [Accepted: 08/29/2024] [Indexed: 11/16/2024]
Abstract
Background and Objective The bone marrow microenvironment is closely related to normal hematopoiesis and hematologic tumors. Adipocytes are an important part of the bone marrow microenvironment, in which they can release free fatty acids (FFAs) through lipolysis and secrete adipocytokines, etc., and participate in normal hematopoiesis, which is closely related to the occurrence and treatment of hematological tumors. In this review, we aim to discuss how bone marrow adipocytes (BMAs) can influence the proliferation, apoptosis, and chemotherapy resistance of cancer cells by reprogramming lipid metabolism and the secretion of various adipocytokines. Methods Studies from 2000 to July 2024 were reviewed from PubMed, Springer Link, and the Web of Science using the keywords bone marrow microenvironment, adipocytes, lipid metabolism, adipocytokines, hematological tumor, cancer, and their combinations. Unreliable articles such as those that are old and have a low impact factor are excluded, and there is no restriction on language. Key Content and Findings Adipocytes can regulate the proliferation and differentiation of hematopoietic stem cells (HSCs) by secreting inflammatory factors and adipocytokines to maintain hematopoietic homeostasis. Adipocytes can also stimulate and accelerate the occurrence and progression of hematological tumors by secreting adipocytokines and mediating the reprogramming of lipid metabolism. Moreover, abundant adipocytes in bone marrow can protect tumor cells by physically blocking and/or secreting cytokines, leading to chemotherapy resistance. Conclusions Therefore, the targeted inhibition of related lipid metabolism pathways and adipocytokines might be a potential therapeutic target for hematological tumors, which would be helpful to inhibit tumor growth and correct chemotherapy resistance.
Collapse
Affiliation(s)
- Yuchun Li
- Central Laboratory, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Linlin Wang
- Central Laboratory, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jingyu Wang
- Central Laboratory, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yaping Xin
- Department of Endocrinology and Metabolic Diseases, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaodong Lyu
- Central Laboratory, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
11
|
Garmo LC, Herroon MK, Mecca S, Wilson A, Allen DR, Agarwal M, Kim S, Petriello MC, Podgorski I. The long-chain polyfluorinated alkyl substance perfluorohexane sulfonate (PFHxS) promotes bone marrow adipogenesis. Toxicol Appl Pharmacol 2024; 491:117047. [PMID: 39111555 DOI: 10.1016/j.taap.2024.117047] [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: 05/17/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/11/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) bioaccumulate in different organ systems, including bone. While existing research highlights the adverse impact of PFAS on bone density, a critical gap remains in understanding the specific effects on the bone marrow microenvironment, especially the bone marrow adipose tissue (BMAT). Changes in BMAT have been linked to various health consequences, such as the development of osteoporosis and the progression of metastatic tumors in bone. Studies presented herein demonstrate that exposure to a mixture of five environmentally relevant PFAS compounds promotes marrow adipogenesis in vitro and in vivo. We show that among the components of the mixture, PFHxS, an alternative to PFOS, has the highest propensity to accumulate in bone and effectively promote marrow adipogenesis. Utilizing RNAseq approaches, we identified the peroxisome proliferator-activated receptor (PPAR) signaling as a top pathway modulated by PFHxS exposure. Furthermore, we provide results suggesting the activation and involvement of PPAR-gamma (PPARγ) in PFHxS-mediated bone marrow adipogenesis, especially in combination with high-fat diet. In conclusion, our findings demonstrate the potential impact of elevated PFHxS levels, particularly in occupational settings, on bone health, and specifically bone marrow adiposity. This study contributes new insights into the health risks of PFHxS exposure, urging further research on the relationship between environmental factors, diet, and adipose tissue dynamics.
Collapse
Affiliation(s)
- Laimar C Garmo
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Mackenzie K Herroon
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Shane Mecca
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Alexis Wilson
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America
| | - David R Allen
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Manisha Agarwal
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Seongho Kim
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America
| | - Michael C Petriello
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America.
| |
Collapse
|
12
|
Xin Z, Qin L, Tang Y, Guo S, Li F, Fang Y, Li G, Yao Y, Zheng B, Zhang B, Wu D, Xiao J, Ni C, Wei Q, Zhang T. Immune mediated support of metastasis: Implication for bone invasion. Cancer Commun (Lond) 2024; 44:967-991. [PMID: 39003618 PMCID: PMC11492328 DOI: 10.1002/cac2.12584] [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: 12/28/2023] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024] Open
Abstract
Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.
Collapse
Affiliation(s)
- Zengfeng Xin
- Department of Orthopedic SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Luying Qin
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yang Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Siyu Guo
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Fangfang Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yuan Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Gege Li
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Yihan Yao
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Binbin Zheng
- Department of Respiratory MedicineNingbo Hangzhou Bay HospitalNingboZhejiangP. R. China
| | - Bicheng Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Dang Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Jie Xiao
- Department of Orthopedic SurgerySecond Affiliated Hospital (Jiande Branch)Zhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Chao Ni
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Breast SurgerySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qichun Wei
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Ting Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and InterventionNational Ministry of Education)Second Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
13
|
Trivanović D, Vujačić M, Labella R, Djordjević IO, Ćazić M, Chernak B, Jauković A. Molecular Deconvolution of Bone Marrow Adipose Tissue Interactions with Malignant Hematopoiesis: Potential for New Therapy Development. Curr Osteoporos Rep 2024; 22:367-377. [PMID: 38922359 DOI: 10.1007/s11914-024-00879-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE OF REVIEW Along with a strong impact on skeletal integrity, bone marrow adipose tissue (BMAT) is an important modulator of the adult hematopoietic system. This review will summarize the current knowledge on the causal relationship between bone marrow (BM) adipogenesis and the development and progression of hematologic malignancies. RECENT FINDINGS BM adipocytes (BMAds) support a number of processes promoting oncogenesis, including the evolution of clonal hematopoiesis, malignant cell survival, proliferation, angiogenesis, and chemoresistance. In addition, leukemic cells manipulate surrounding BMAds by promoting lipolysis and release of free fatty acids, which are then utilized by leukemic cells via β-oxidation. Therefore, limiting BM adipogenesis, blocking BMAd-derived adipokines, or lipid metabolism obstruction have been considered as potential treatment options for hematological malignancies. Leukemic stem cells rely heavily on BMAds within the structural BM microenvironment for necessary signals which foster disease progression. Further development of 3D constructs resembling BMAT at different skeletal regions are critical to better understand these relationships in geometric space and may provide essential insight into the development of hematologic malignancies within the BM niche. In turn, these mechanisms provide promising potential as novel approaches to targeting the microenvironment with new therapeutic strategies.
Collapse
Affiliation(s)
- Drenka Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia.
| | - Marko Vujačić
- Institute for Orthopedy Banjica, 11000, Belgrade, Serbia
- School of Medicine, University of Belgrade, 11000, Belgrade, Serbia
| | - Rossella Labella
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Edward P. Evans Center for Myelodysplastic Syndromes, Columbia University Medical Center, New York, NY, USA
| | - Ivana Okić Djordjević
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia
| | - Marija Ćazić
- Department of Hematology and Oncology, University Children's Hospital Tiršova, 11000, Belgrade, Serbia
| | - Brian Chernak
- Division of Hematology/Oncology, Columbia University, New York, NY, USA
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000, Belgrade, Serbia
| |
Collapse
|
14
|
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: 3] [Impact Index Per Article: 3.0] [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
|
15
|
Morse PT, Wan J, Arroum T, Herroon MK, Kalpage HA, Bazylianska V, Lee I, Heath EI, Podgorski I, Hüttemann M. Prostate Cancer-Specific Lysine 53 Acetylation of Cytochrome c Drives Metabolic Reprogramming and Protects from Apoptosis in Intact Cells. Biomolecules 2024; 14:695. [PMID: 38927098 PMCID: PMC11201891 DOI: 10.3390/biom14060695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Cytochrome c (Cytc) is important for both mitochondrial respiration and apoptosis, both of which are altered in cancer cells that switch to Warburg metabolism and manage to evade apoptosis. We earlier reported that lysine 53 (K53) of Cytc is acetylated in prostate cancer. K53 is conserved in mammals that is known to be essential for binding to cytochrome c oxidase and apoptosis protease activating factor-1 (Apaf-1). Here we report the effects of this acetylation on the main functions of cytochrome c by expressing acetylmimetic K53Q in cytochrome c double knockout cells. Other cytochrome c variants analyzed were wild-type, K53R as a control that maintains the positive charge, and K53I, which is present in some non-mammalian species. Intact cells expressing K53Q cytochrome c showed 49% decreased mitochondrial respiration and a concomitant increase in glycolytic activity (Warburg effect). Furthermore, mitochondrial membrane potential was decreased, correlating with notably reduced basal mitochondrial superoxide levels and decreased cell death upon challenge with H2O2 or staurosporine. To test for markers of cancer aggressiveness and invasiveness, cells were grown in 3D spheroid culture. K53Q cytochrome c-expressing cells showed profoundly increased protrusions compared to WT, suggesting increased invasiveness. We propose that K53 acetylation of cytochrome c is an adaptive response that mediates prostate cancer metabolic reprogramming and evasion of apoptosis, which are two hallmarks of cancer, to better promote tumor survival and metastasis.
Collapse
Affiliation(s)
- Paul T. Morse
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (P.T.M.)
| | - Junmei Wan
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (P.T.M.)
| | - Tasnim Arroum
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (P.T.M.)
| | | | - Hasini A. Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (P.T.M.)
| | - Viktoriia Bazylianska
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (P.T.M.)
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI 48201, USA
| | - Icksoo Lee
- College of Medicine, Dankook University, Cheonan-si 31116, Republic of Korea;
| | - Elisabeth I. Heath
- Karmanos Cancer Institute, Department of Oncology, Wayne State University, Detroit, MI 48201, USA
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; (P.T.M.)
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
16
|
Lecka-Czernik B, Khan MP, Letson J, Baroi S, Chougule A. Regulatory Effect of Osteocytes on Extramedullary and Bone Marrow Adipose Tissue Development and Function. Curr Osteoporos Rep 2024; 22:301-307. [PMID: 38625510 PMCID: PMC11186862 DOI: 10.1007/s11914-024-00871-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE OF REVIEW This review summarizes evidence on osteocyte support of extramedullary and bone marrow adipocyte development and discusses the role of endogenous osteocyte activities of nuclear receptors peroxisome proliferator-activated receptor gamma (PPARG) and alpha (PPARA) in this support. RECENT FINDINGS PPARG and PPARA proteins, key regulators of glucose and fatty acid metabolism, are highly expressed in osteocytes. They play significant roles in the regulation of osteocyte secretome and osteocyte bioenergetics; both activities contributing to the levels of systemic energy metabolism in part through an effect on metabolic function of extramedullary and bone marrow adipocytes. The PPARs-controlled osteocyte endocrine/paracrine activities, including sclerostin expression, directly regulate adipocyte function, while the PPARs-controlled osteocyte fuel utilization and oxidative phosphorylation contribute to the skeletal demands for glucose and fatty acids, whose availability is under the control of adipocytes. Bone is an inherent element of systemic energy metabolism with PPAR nuclear receptors regulating osteocyte-adipocyte metabolic axes.
Collapse
Affiliation(s)
- Beata Lecka-Czernik
- Department of Orthopaedic Surgery, Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, 43614, USA.
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, 43614, USA
| | - Joshua Letson
- Department of Orthopaedic Surgery, Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, 43614, USA
| | - Sudipta Baroi
- Department of Orthopaedic Surgery, Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, 43614, USA
- Harvard University, School of Dental Medicine, Boston, MA, 02115, USA
| | - Amit Chougule
- Department of Orthopaedic Surgery, Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, 43614, USA
- University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
17
|
Fontana F, Giannitti G, Marchesi S, Limonta P. The PI3K/Akt Pathway and Glucose Metabolism: A Dangerous Liaison in Cancer. Int J Biol Sci 2024; 20:3113-3125. [PMID: 38904014 PMCID: PMC11186371 DOI: 10.7150/ijbs.89942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/11/2024] [Indexed: 06/22/2024] Open
Abstract
Aberrant activation of the PI3K/Akt pathway commonly occurs in cancers and correlates with multiple aspects of malignant progression. In particular, recent evidence suggests that the PI3K/Akt signaling plays a fundamental role in promoting the so-called aerobic glycolysis or Warburg effect, by phosphorylating different nutrient transporters and metabolic enzymes, such as GLUT1, HK2, PFKB3/4 and PKM2, and by regulating various molecular networks and proteins, including mTORC1, GSK3, FOXO transcription factors, MYC and HIF-1α. This leads to a profound reprogramming of cancer metabolism, also impacting on pentose phosphate pathway, mitochondrial oxidative phosphorylation, de novo lipid synthesis and redox homeostasis and thereby allowing the fulfillment of both the catabolic and anabolic demands of tumor cells. The present review discusses the interactions between the PI3K/Akt cascade and its metabolic targets, focusing on their possible therapeutic implications.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | | | | | | |
Collapse
|
18
|
Ambrosini G, Cordani M, Zarrabi A, Alcon-Rodriguez S, Sainz RM, Velasco G, Gonzalez-Menendez P, Dando I. Transcending frontiers in prostate cancer: the role of oncometabolites on epigenetic regulation, CSCs, and tumor microenvironment to identify new therapeutic strategies. Cell Commun Signal 2024; 22:36. [PMID: 38216942 PMCID: PMC10790277 DOI: 10.1186/s12964-023-01462-0] [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: 10/05/2023] [Accepted: 12/27/2023] [Indexed: 01/14/2024] Open
Abstract
Prostate cancer, as one of the most prevalent malignancies in males, exhibits an approximate 5-year survival rate of 95% in advanced stages. A myriad of molecular events and mutations, including the accumulation of oncometabolites, underpin the genesis and progression of this cancer type. Despite growing research demonstrating the pivotal role of oncometabolites in supporting various cancers, including prostate cancer, the root causes of their accumulation, especially in the absence of enzymatic mutations, remain elusive. Consequently, identifying a tangible therapeutic target poses a formidable challenge. In this review, we aim to delve deeper into the implications of oncometabolite accumulation in prostate cancer. We center our focus on the consequential epigenetic alterations and impacts on cancer stem cells, with the ultimate goal of outlining novel therapeutic strategies.
Collapse
Affiliation(s)
- Giulia Ambrosini
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering & Natural Sciences, Istinye University, Istanbul, 34396, Turkey
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
| | - Sergio Alcon-Rodriguez
- Departamento de Morfología y Biología Celular, School of Medicine, Julián Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain
| | - Rosa M Sainz
- Departamento de Morfología y Biología Celular, School of Medicine, Julián Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, 28040, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain
| | - Pedro Gonzalez-Menendez
- Departamento de Morfología y Biología Celular, School of Medicine, Julián Claveria 6, 33006, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, 33006, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias (HUCA), 33011, Oviedo, Spain.
| | - Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy.
| |
Collapse
|
19
|
Diedrich JD, Cole CE, Pianko MJ, Colacino JA, Bernard JJ. Non-Toxicological Role of Aryl Hydrocarbon Receptor in Obesity-Associated Multiple Myeloma Cell Growth and Survival. Cancers (Basel) 2023; 15:5255. [PMID: 37958428 PMCID: PMC10649826 DOI: 10.3390/cancers15215255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/27/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Obesity is not only a risk factor for multiple myeloma (MM) incidence, but it is also associated with an increased risk of progression from myeloma precursors-monoclonal gammopathy of undetermined significance-and smoldering myeloma. Adipocytes in the bone marrow (BMAs) microenvironment have been shown to facilitate MM cell growth via secreted factors, but the nature of these secreted factors and their mechanism of action have not been fully elucidated. The elevated expression of aryl hydrocarbon receptor (AhR) is associated with a variety of different cancers, including MM; however, the role of AhR activity in obesity-associated MM cell growth and survival has not been explored. Indeed, this is of particular interest as it has been recently shown that bone marrow adipocytes are a source of endogenous AhR ligands. Using multiple in vitro models of tumor-adipocyte crosstalk to mimic the bone microenvironment, we identified a novel, non-toxicological role of the adipocyte-secreted factors in the suppression of AhR activity in MM cells. A panel of six MM cell lines were cultured in the presence of bone marrow adipocytes in (1) a direct co-culture, (2) a transwell co-culture, or (3) an adipocyte-conditioned media to interrogate the effects of the secreted factors on MM cell AhR activity. Nuclear localization and the transcriptional activity of the AhR, as measured by CYP1A1 and CYP1B1 gene induction, were suppressed by exposure to BMA-derived factors. Additionally, decreased AhR target gene expression was associated with worse clinical outcomes. The knockdown of AhR resulted in reduced CYP1B1 expression and increased cellular growth. This tumor-suppressing role of CYP1A1 and CYP1B1 was supported by patient data which demonstrated an association between reduced target gene expression and worse overall survival. These data demonstrated a novel mechanism by which bone marrow adipocytes promote MM progression.
Collapse
Affiliation(s)
- Jonathan D. Diedrich
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Craig E. Cole
- Department of Medicine, Division of Hematology/Oncology, Michigan State University, East Lansing, MI 48910, USA;
- Karmanos Cancer Institute, McLaren Greater Lansing, Lansing, MI 48910, USA
- Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Matthew J. Pianko
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Justin A. Colacino
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jamie J. Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
- Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
20
|
Chaba A, Fodil S, Lemiale V, Mariotte E, Valade S, Azoulay E, Zafrani L. Clinical Warburg effect in lymphoma patients admitted to intensive care unit. Ann Intensive Care 2023; 13:97. [PMID: 37796407 PMCID: PMC10555986 DOI: 10.1186/s13613-023-01192-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: 07/20/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND The Warburg effect, characterized by elevated lactate levels without tissue hypoxia or shock, has been described in patients with aggressive lymphoproliferative malignancies. However, the clinical characteristics and long-term outcomes in this population remain poorly understood. METHODS We retrospectively analyzed 135 patients with aggressive lymphoproliferative malignancies admitted to the ICU between January 2017 and December 2022. Patients were classified into three groups: Clinical Warburg Effect (CWE), No Warburg with High Lactate level (NW-HL), and No Warburg with Normal Lactate level (NW-NL). Clinical characteristics and outcomes were compared between the groups and factors associated with 1-year mortality and CWE were identified using multivariable analyses. RESULTS Of the 135 patients, 46 (34%) had a CWE. This group had a higher proportion of Burkitt and T cell lymphomas, greater tumor burden, and more frequent bone and cerebral involvement than the other groups. At 1 year, 72 patients (53%) died, with significantly higher mortality in the CWE and NW-HL groups (70% each) than in the NW-NL group (38%). Factors independently associated with 1-year mortality were age [HR = 1.02 CI 95% (1.00-1.04)], total SOFA score at admission [HR = 1.19 CI 95% (1.12-1.25)], and CWE [HR = 3.87 CI 95% (2.13-7.02)]. The main factors associated with the CWE were tumor lysis syndrome [OR = 2.84 CI 95% (1.14-7.42)], bone involvement of the underlying malignancy [OR = 3.58 CI 95% (1.02-12.91)], the total SOFA score at admission [OR = 0.81 CI 95% (0.69-0.91)] and hypoglycemia at admission [OR = 14.90 CI 95% (5.42-47.18)]. CONCLUSION CWE is associated with a higher tumor burden and increased 1-year mortality compared to patients without this condition. Our findings underscore the importance of recognizing patients with CWE as a high-risk cohort, as their outcomes closely resemble those of individuals with lymphoma and shock, despite not requiring advanced organ support. Clinicians should recognize the urgency of managing these patients and consider early intervention to improve their prognosis.
Collapse
Affiliation(s)
- Anis Chaba
- Medical Intensive Care Unit, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - Sofiane Fodil
- Department of Hematology, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Virginie Lemiale
- Medical Intensive Care Unit, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - Eric Mariotte
- Medical Intensive Care Unit, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - Sandrine Valade
- Medical Intensive Care Unit, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - Elie Azoulay
- Medical Intensive Care Unit, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 1 Avenue Claude Vellefaux, 75010, Paris, France
- University Paris Cité, Paris, France
| | - Lara Zafrani
- Medical Intensive Care Unit, Saint-Louis University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 1 Avenue Claude Vellefaux, 75010, Paris, France.
- University Paris Cité, Paris, France.
- INSERM, UMR 944, University Paris Cité, Paris, France.
| |
Collapse
|
21
|
Diedrich JD, Gonzalez-Pons R, Medeiros HCD, Ensink E, Liby KT, Wellberg EA, Lunt SY, Bernard JJ. Adipocyte-derived kynurenine stimulates malignant transformation of mammary epithelial cells through the aryl hydrocarbon receptor. Biochem Pharmacol 2023; 216:115763. [PMID: 37625554 PMCID: PMC10587895 DOI: 10.1016/j.bcp.2023.115763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Anti-hormone therapies are not efficacious for reducing the incidence of triple negative breast cancer (TNBC), which lacks both estrogen and progesterone receptors. While the etiology of this aggressive breast cancer subtype is unclear, visceral obesity is a strong risk factor for both pre- and post-menopausal cases. The mechanism by which excessive deposition of visceral adipose tissue (VAT) promotes the malignant transformation of hormone receptor-negative mammary epithelial cells is currently unknown. We developed a novel in vitro system of malignant transformation in which non-tumorigenic human breast epithelial cells (MCF-10A) grow in soft agar when cultured with factors released from VAT. These cells, which acquire the capacity for 3D growth, show elevated aryl hydrocarbon receptor (AhR) protein and AhR target genes, suggesting that AhR activity may drive malignant transformation by VAT. AhR is a ligand-dependent transcription factor that generates biological responses to exogenous carcinogens and to the endogenous tryptophan pathway metabolite, kynurenine. The serum kynurenine to tryptophan ratio has been shown to be elevated in patients with obesity. Herein, we demonstrate that AhR inhibitors or knockdown of AhR in MCF-10A cells prevents VAT-induced malignant transformation. Specifically, VAT-induced transformation is inhibited by Kyn-101, an inhibitor for the endogenous ligand binding site of AhR. Mass spectrometry analysis demonstrates that adipocytes metabolize tryptophan and release kynurenine, which is taken up by MCF-10A cells and activates the AhR to induce CYP1B1 and promote malignant transformation. This novel hormone receptor-independent mechanism of malignant transformation suggests targeting AhR for TNBC prevention in the context of visceral adiposity.
Collapse
Affiliation(s)
- Jonathan D Diedrich
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA
| | - Romina Gonzalez-Pons
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA
| | - Hyllana C D Medeiros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Elliot Ensink
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA
| | - Karen T Liby
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824 USA; Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824 USA
| | - Jamie J Bernard
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824 USA; Department of Medicine, Michigan State University, East Lansing, MI 48824 USA.
| |
Collapse
|
22
|
Saha A, Kolonin MG, DiGiovanni J. Obesity and prostate cancer - microenvironmental roles of adipose tissue. Nat Rev Urol 2023; 20:579-596. [PMID: 37198266 DOI: 10.1038/s41585-023-00764-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 05/19/2023]
Abstract
Obesity is known to have important roles in driving prostate cancer aggressiveness and increased mortality. Multiple mechanisms have been postulated for these clinical observations, including effects of diet and lifestyle, systemic changes in energy balance and hormonal regulation and activation of signalling by growth factors and cytokines and other components of the immune system. Over the past decade, research on obesity has shifted towards investigating the role of peri-prostatic white adipose tissue as an important source of locally produced factors that stimulate prostate cancer progression. Cells that comprise white adipose tissue, the adipocytes and their progenitor adipose stromal cells (ASCs), which proliferate to accommodate white adipose tissue expansion in obesity, have been identified as important drivers of obesity-associated cancer progression. Accumulating evidence suggests that adipocytes are a source of lipids that are used by adjacent prostate cancer cells. However, results of preclinical studies indicate that ASCs promote tumour growth by remodelling extracellular matrix and supporting neovascularization, contributing to the recruitment of immunosuppressive cells, and inducing epithelial-mesenchymal transition through paracrine signalling. Because epithelial-mesenchymal transition is associated with cancer chemotherapy resistance and metastasis, ASCs are considered to be potential targets of therapies that could be developed to suppress cancer aggressiveness in patients with obesity.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Disease, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA.
| | - John DiGiovanni
- Division of Pharmacology and Toxicology and Dell Paediatric Research Institute, The University of Texas at Austin, Austin, TX, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
23
|
Salamanna F, Contartese D, Errani C, Sartori M, Borsari V, Giavaresi G. Role of bone marrow adipocytes in bone metastasis development and progression: a systematic review. Front Endocrinol (Lausanne) 2023; 14:1207416. [PMID: 37711896 PMCID: PMC10497772 DOI: 10.3389/fendo.2023.1207416] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
Purpose Bone marrow adipocytes (BMAs) are the most plentiful cells in the bone marrow and function as an endocrine organ by producing fatty acids, cytokines, and adipokines. Consequently, BMAs can interact with tumor cells, influencing both tumor growth and the onset and progression of bone metastasis. This review aims to systematically evaluate the role of BMAs in the development and progression of bone metastasis. Methods A comprehensive search was conducted on PubMed, Web of Science, and Scopus electronic databases, following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement standards, to identify studies published from March 2013 to June 2023. Two independent reviewers assessed and screened the literature, extracted the data, and evaluated the quality of the studies. The body of evidence was evaluated and graded using the ROBINS-I tool for non-randomized studies of interventions and the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) tool for in vivo studies. The results were synthesized using descriptive methods. Results The search yielded a total of 463 studies, of which 17 studies were included in the final analysis, including 15 preclinical studies and two non-randomized clinical studies. Analysis of preclinical studies revealed that BMAs play a significant role in bone metastasis, particularly in prostate cancer followed by breast and malignant melanoma cancers. BMAs primarily influence cancer cells by inducing a glycolytic phenotype and releasing or upregulating soluble factors, chemokines, cytokines, adipokines, tumor-derived fatty acid-binding protein (FABP), and members of the nuclear receptor superfamily, such as chemokine (C-C motif) ligand 7 (CCL7), C-X-C Motif Chemokine Ligand (CXCL)1, CXCL2, interleukin (IL)-1β, IL-6, FABP4, and peroxisome proliferator-activated receptor γ (PPARγ). These factors also contribute to adipocyte lipolysis and regulate a pro-inflammatory phenotype in BMAs. However, the number of clinical studies is limited, and definitive conclusions cannot be drawn. Conclusion The preclinical studies reviewed indicate that BMAs may play a crucial role in bone metastasis in prostate, breast, and malignant melanoma cancers. Nevertheless, further preclinical and clinical studies are needed to better understand the complex role and relationship between BMAs and cancer cells in the bone microenvironment. Targeting BMAs in combination with standard treatments holds promise as a potential therapeutic strategy for bone metastasis.
Collapse
Affiliation(s)
- F. Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - D. Contartese
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - C. Errani
- 3rd Orthopaedic and Traumatologic Clinic Prevalently Oncologic, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - M. Sartori
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V. Borsari
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - G. Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
24
|
Chetta P, Sriram R, Zadra G. Lactate as Key Metabolite in Prostate Cancer Progression: What Are the Clinical Implications? Cancers (Basel) 2023; 15:3473. [PMID: 37444583 PMCID: PMC10340474 DOI: 10.3390/cancers15133473] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Advanced prostate cancer represents the fifth leading cause of cancer death in men worldwide. Although androgen-receptor signaling is the major driver of the disease, evidence is accumulating that disease progression is supported by substantial metabolic changes. Alterations in de novo lipogenesis and fatty acid catabolism are consistently reported during prostate cancer development and progression in association with androgen-receptor signaling. Therefore, the term "lipogenic phenotype" is frequently used to describe the complex metabolic rewiring that occurs in prostate cancer. However, a new scenario has emerged in which lactate may play a major role. Alterations in oncogenes/tumor suppressors, androgen signaling, hypoxic conditions, and cells in the tumor microenvironment can promote aerobic glycolysis in prostate cancer cells and the release of lactate in the tumor microenvironment, favoring immune evasion and metastasis. As prostate cancer is composed of metabolically heterogenous cells, glycolytic prostate cancer cells or cancer-associated fibroblasts can also secrete lactate and create "symbiotic" interactions with oxidative prostate cancer cells via lactate shuttling to sustain disease progression. Here, we discuss the multifaceted role of lactate in prostate cancer progression, taking into account the influence of the systemic metabolic and gut microbiota. We call special attention to the clinical opportunities of imaging lactate accumulation for patient stratification and targeting lactate metabolism.
Collapse
Affiliation(s)
- Paolo Chetta
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Giorgia Zadra
- Institute of Molecular Genetics, National Research Council (IGM-CNR), 27100 Pavia, Italy
| |
Collapse
|
25
|
Cheng F, He J, Yang J. Bone marrow microenvironment: roles and therapeutic implications in obesity-associated cancer. Trends Cancer 2023; 9:566-577. [PMID: 37087397 PMCID: PMC10329995 DOI: 10.1016/j.trecan.2023.03.007] [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: 02/01/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023]
Abstract
Obesity is increasing globally and has been closely linked to the initiation and progression of multiple human cancers. These relationships, to a large degree, are mediated through obesity-driven disruption of physiological homeostasis characterized by local and systemic endocrinologic, inflammatory, and metabolic changes. Bone marrow microenvironment (BMME), which evolves during obesity, has been implicated in multiple types of cancer. Growing evidence shows that physiological dysfunction of BMME with altered cellular composition, stromal and immune cell function, and energy metabolism, as well as inflammation and hypoxia, in the context of obesity contributes to cancer initiation and progression. Nonetheless, the mechanisms underlying the obesity-BMME-cancer axis remain elusive. In this review, we discuss the recent advances in understanding the evolution of BMME during obesity, its contributions to cancer initiation and progression, and the implications for cancer therapy.
Collapse
Affiliation(s)
- Feifei Cheng
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Jin He
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Jing Yang
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
26
|
Tirado HA, Balasundaram N, Laaouimir L, Erdem A, van Gastel N. Metabolic crosstalk between stromal and malignant cells in the bone marrow niche. Bone Rep 2023; 18:101669. [PMID: 36909665 PMCID: PMC9996235 DOI: 10.1016/j.bonr.2023.101669] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/03/2023] Open
Abstract
Bone marrow is the primary site of blood cell production in adults and serves as the source of osteoblasts and osteoclasts that maintain bone homeostasis. The medullary microenvironment is also involved in malignancy, providing a fertile soil for the growth of blood cancers or solid tumors metastasizing to bone. The cellular composition of the bone marrow is highly complex, consisting of hematopoietic stem and progenitor cells, maturing blood cells, skeletal stem cells, osteoblasts, mesenchymal stromal cells, adipocytes, endothelial cells, lymphatic endothelial cells, perivascular cells, and nerve cells. Intercellular communication at different levels is essential to ensure proper skeletal and hematopoietic tissue function, but it is altered when malignant cells colonize the bone marrow niche. While communication often involves soluble factors such as cytokines, chemokines, and growth factors, as well as their respective cell-surface receptors, cells can also communicate by exchanging metabolic information. In this review, we discuss the importance of metabolic crosstalk between different cells in the bone marrow microenvironment, particularly concerning the malignant setting.
Collapse
Affiliation(s)
- Hernán A Tirado
- Cellular Metabolism and Microenvironment Laboratory, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Nithya Balasundaram
- Cellular Metabolism and Microenvironment Laboratory, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Lotfi Laaouimir
- Cellular Metabolism and Microenvironment Laboratory, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Ayşegül Erdem
- Cellular Metabolism and Microenvironment Laboratory, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Nick van Gastel
- Cellular Metabolism and Microenvironment Laboratory, de Duve Institute, UCLouvain, Brussels, Belgium.,WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
27
|
Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15041192. [PMID: 36831534 PMCID: PMC9953833 DOI: 10.3390/cancers15041192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer deaths among men in Western countries. Mitochondria, the "powerhouse" of cells, undergo distinctive metabolic and structural dynamics in different types of cancer. PCa cells experience peculiar metabolic changes during their progression from normal epithelial cells to early-stage and, progressively, to late-stage cancer cells. Specifically, healthy cells display a truncated tricarboxylic acid (TCA) cycle and inefficient oxidative phosphorylation (OXPHOS) due to the high accumulation of zinc that impairs the activity of m-aconitase, the enzyme of the TCA cycle responsible for the oxidation of citrate. During the early phase of cancer development, intracellular zinc levels decrease leading to the reactivation of m-aconitase, TCA cycle and OXPHOS. PCa cells change their metabolic features again when progressing to the late stage of cancer. In particular, the Warburg effect was consistently shown to be the main metabolic feature of late-stage PCa cells. However, accumulating evidence sustains that both the TCA cycle and the OXPHOS pathway are still present and active in these cells. The androgen receptor axis as well as mutations in mitochondrial genes involved in metabolic rewiring were shown to play a key role in PCa cell metabolic reprogramming. Mitochondrial structural dynamics, such as biogenesis, fusion/fission and mitophagy, were also observed in PCa cells. In this review, we focus on the mitochondrial metabolic and structural dynamics occurring in PCa during tumor development and progression; their role as effective molecular targets for novel therapeutic strategies in PCa patients is also discussed.
Collapse
|
28
|
Wanjari UR, Mukherjee AG, Gopalakrishnan AV, Murali R, Dey A, Vellingiri B, Ganesan R. Role of Metabolism and Metabolic Pathways in Prostate Cancer. Metabolites 2023; 13:183. [PMID: 36837801 PMCID: PMC9962346 DOI: 10.3390/metabo13020183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/21/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Prostate cancer (PCa) is the common cause of death in men. The pathophysiological factors contributing to PCa are not well known. PCa cells gain a protective mechanism via abnormal lipid signaling and metabolism. PCa cells modify their metabolism in response to an excessive intake of nutrients to facilitate advancement. Metabolic syndrome (MetS) is inextricably linked to the carcinogenic progression of PCa, which heightens the severity of the disease. It is hypothesized that changes in the metabolism of the mitochondria contribute to the onset of PCa. The studies of particular alterations in the progress of PCa are best accomplished by examining the metabolome of prostate tissue. Due to the inconsistent findings written initially, additional epidemiological research is required to identify whether or not MetS is an aspect of PCa. There is a correlation between several risk factors and the progression of PCa, one of which is MetS. The metabolic symbiosis between PCa cells and the tumor milieu and how this type of crosstalk may aid in the development of PCa is portrayed in this work. This review focuses on in-depth analysis and evaluation of the metabolic changes that occur within PCa, and also aims to assess the effect of metabolic abnormalities on the aggressiveness status and metabolism of PCa.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Balachandar Vellingiri
- Stem Cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
29
|
Interplay between fat cells and immune cells in bone: Impact on malignant progression and therapeutic response. Pharmacol Ther 2022; 238:108274. [DOI: 10.1016/j.pharmthera.2022.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022]
|
30
|
Sethakorn N, Heninger E, Breneman MT, Recchia E, Ding AB, Jarrard DF, Hematti P, Beebe DJ, Kosoff D. Integrated analysis of the tumor microenvironment using a reconfigurable microfluidic cell culture platform. FASEB J 2022; 36:e22540. [PMID: 36083096 PMCID: PMC9476232 DOI: 10.1096/fj.202200684rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/11/2022]
Abstract
The tumor microenvironment (TME) is a complex network of non-malignant cells and stroma that perform a wide array of vital roles in tumor growth, immune evasion, metastasis, and therapeutic resistance. These highly diverse roles have been shown to be critically important to the progression of cancers and have already shown potential as therapeutic targets. Therefore, there has been a tremendous push to elucidate the pathways that underlie these roles and to develop new TME-directed therapies for cancer treatment. Unfortunately, TME-focused research has been limited by a lack of translational in vitro culture platforms that can model this highly complex niche and can support the integrated analysis of cell biology and function. In the current study, we investigate whether an independently developed reconfigurable microfluidic platform, known as Stacks, can address the critical need for translational multi-cellular tumor models and integrated analytics in TME research. We present data on multi-cellular culture of primary human cells in Stacks as well as the orthogonal analysis of cellular polarization, differentiation, migration, and cytotoxicity in this reconfigurable system. These expanded capabilities of Stacks are highly relevant to the cancer research community with the potential to enhance clinical translation of pre-clinical TME studies and to yield novel biological insight into TME crosstalk, metastasis, and responses to novel drug combinations or immune therapies.
Collapse
Affiliation(s)
- Nan Sethakorn
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Erika Heninger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Matthew T Breneman
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Emma Recchia
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Adeline B Ding
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| |
Collapse
|
31
|
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: 17] [Impact Index Per Article: 5.7] [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
|
32
|
Fontana F, Anselmi M, Carollo E, Sartori P, Procacci P, Carter D, Limonta P. Adipocyte-Derived Extracellular Vesicles Promote Prostate Cancer Cell Aggressiveness by Enabling Multiple Phenotypic and Metabolic Changes. Cells 2022; 11:cells11152388. [PMID: 35954232 PMCID: PMC9368412 DOI: 10.3390/cells11152388] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 12/12/2022] Open
Abstract
Background: In recent decades, obesity has widely emerged as an important risk factor for prostate cancer (PCa). Adipose tissue and PCa cells have been shown to orchestrate a complex interaction network to support tumor growth and evolution; nonetheless, the study of this communication has only been focused on soluble factors, although increasing evidence highlights the key role of extracellular vesicles (EVs) in the modulation of tumor progression. Methods and Results: In the present study, we found that EVs derived from 3T3-L1 adipocytes could affect PC3 and DU145 PCa cell traits, inducing increased proliferation, migration and invasion. Furthermore, conditioning of both PCa cell lines with adipocyte-released EVs resulted in lower sensitivity to docetaxel, with reduced phosphatidylserine externalization and decreased caspase 3 and PARP cleavage. In particular, these alterations were paralleled by an Akt/HIF-1α axis-related Warburg effect, characterized by enhanced glucose consumption, lactate release and ATP production. Conclusions: Collectively, these findings demonstrate that EV-mediated crosstalk exists between adipocytes and PCa, driving tumor aggressiveness.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; (M.A.); (P.L.)
- Correspondence:
| | - Martina Anselmi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; (M.A.); (P.L.)
| | - Emanuela Carollo
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (E.C.); (D.C.)
| | - Patrizia Sartori
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (P.S.); (P.P.)
| | - Patrizia Procacci
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milano, Italy; (P.S.); (P.P.)
| | - David Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (E.C.); (D.C.)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; (M.A.); (P.L.)
| |
Collapse
|
33
|
Sachdeva A, Hart CA, Kim K, Tawadros T, Oliveira P, Shanks J, Brown M, Clarke N. Non-canonical EphA2 activation underpins PTEN-mediated metastatic migration and poor clinical outcome in prostate cancer. Br J Cancer 2022; 127:1254-1262. [PMID: 35869144 PMCID: PMC9519535 DOI: 10.1038/s41416-022-01914-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/23/2022] [Accepted: 07/06/2022] [Indexed: 11/23/2022] Open
Abstract
Background The key process of mesenchymal to amoeboid transition (MAT), which enables prostate cancer (PCa) transendothelial migration and subsequent development of metastases in red bone marrow stroma, is driven by phosphorylation of EphA2S897 by pAkt, which is induced by the omega-6 polyunsaturated fatty acid arachidonic acid. Here we investigate the influence of EphA2 signalling in PCa progression and long-term survival. Methods The mechanisms underpinning metastatic biopotential of altered EphA2 signalling in relation to PTEN status were assessed in vitro using canonical (EphA2D739N) and non-canonical (EphA2S897G) PC3-M mutants, interrogation of publicly available PTEN-stratified databases and clinical validation using a PCa TMA (n = 177) with long-term follow-up data. Spatial heterogeneity of EphA2 was assessed using a radical prostatectomy cohort (n = 67). Results Non-canonical EphA2 signalling via pEphA2S897 is required for PCa transendothelial invasion of bone marrow endothelium. High expression of EphA2 or pEphA2S897 in a PTENlow background is associated with poor overall survival. Expression of EphA2, pEphA2S897 and the associated MAT marker pMLC2 are spatially regulated with the highest levels found within lesion areas within 500 µm of the prostate margin. Conclusion EphA2 MAT-related signalling confers transendothelial invasion. This is associated with a substantially worse prognosis in PTEN-deficient PCa.
Collapse
|
34
|
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
|
35
|
Pachón-Peña G, Bredella MA. Bone marrow adipose tissue in metabolic health. Trends Endocrinol Metab 2022; 33:401-408. [PMID: 35396163 PMCID: PMC9098665 DOI: 10.1016/j.tem.2022.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/25/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
Recent studies have highlighted the role of bone marrow adipose tissue (BMAT) as a regulator of skeletal homeostasis and energy metabolism. While long considered an inert filler, occupying empty spaces from bone loss and reduced hematopoiesis, BMAT is now considered a secretory and metabolic organ that responds to nutritional challenges and secretes cytokines, which indirectly impact energy and bone metabolism. The recent advances in our understanding of the function of BMAT have been enabled by novel noninvasive imaging techniques, which allow longitudinal assessment of BMAT in vivo following interventions. This review will focus on the latest advances in our understanding of BMAT and its role in metabolic health. Imaging techniques to quantify the content and composition of BMAT will be discussed.
Collapse
Affiliation(s)
| | - Miriam A Bredella
- Division of Musculoskeletal Imaging and Intervention, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
36
|
Sanchis P, Anselmino N, Lage-Vickers S, Sabater A, Lavignolle R, Labanca E, Shepherd PDA, Bizzotto J, Toro A, Mitrofanova A, Valacco MP, Navone N, Vazquez E, Cotignola J, Gueron G. Bone Progenitors Pull the Strings on the Early Metabolic Rewiring Occurring in Prostate Cancer Cells. Cancers (Basel) 2022; 14:cancers14092083. [PMID: 35565211 PMCID: PMC9104818 DOI: 10.3390/cancers14092083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 12/10/2022] Open
Abstract
Metastatic prostate cancer (PCa) cells soiling in the bone require a metabolic adaptation. Here, we identified the metabolic genes fueling the seeding of PCa in the bone niche. Using a transwell co-culture system of PCa (PC3) and bone progenitor cells (MC3T3 or Raw264.7), we assessed the transcriptome of PC3 cells modulated by soluble factors released from bone precursors. In a Principal Component Analysis using transcriptomic data from human PCa samples (GSE74685), the altered metabolic genes found in vitro were able to stratify PCa patients in two defined groups: primary PCa and bone metastasis, confirmed by an unsupervised clustering analysis. Thus, the early transcriptional metabolic profile triggered in the in vitro model has a clinical correlate in human bone metastatic samples. Further, the expression levels of five metabolic genes (VDR, PPARA, SLC16A1, GPX1 and PAPSS2) were independent risk-predictors of death in the SU2C-PCF dataset and a risk score model built using this lipid-associated signature was able to discriminate a subgroup of bone metastatic PCa patients with a 23-fold higher risk of death. This signature was validated in a PDX pre-clinical model when comparing MDA-PCa-183 growing intrafemorally vs. subcutaneously, and appears to be under the regulatory control of the Protein Kinase A (PKA) signaling pathway. Secretome analyses of conditioned media showcased fibronectin and type-1 collagen as critical bone-secreted factors that could regulate tumoral PKA. Overall, we identified a novel lipid gene signature, driving PCa aggressive metastatic disease pointing to PKA as a potential hub to halt progression.
Collapse
Affiliation(s)
- Pablo Sanchis
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (P.D.A.S.); (N.N.)
| | - Sofia Lage-Vickers
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Agustina Sabater
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
- Universidad Argentina de la Empresa (UADE), Instituto de Tecnología (INTEC), Buenos Aires C1073AAO, Argentina
| | - Rosario Lavignolle
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (P.D.A.S.); (N.N.)
| | - Peter D. A. Shepherd
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (P.D.A.S.); (N.N.)
| | - Juan Bizzotto
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Ayelen Toro
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, Rutgers School of Health Professions, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 07101, USA;
| | - Maria Pia Valacco
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Nora Navone
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (P.D.A.S.); (N.N.)
| | - Elba Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Javier Cotignola
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (P.S.); (S.L.-V.); (A.S.); (R.L.); (J.B.); (A.T.); (M.P.V.); (E.V.); (J.C.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
- Correspondence: ; Tel.: +54-9114-408-7796; Fax: +54-9114-788-5755
| |
Collapse
|
37
|
Fader Kaiser CM, Romano PS, Vanrell MC, Pocognoni CA, Jacob J, Caruso B, Delgui LR. Biogenesis and Breakdown of Lipid Droplets in Pathological Conditions. Front Cell Dev Biol 2022; 9:826248. [PMID: 35198567 PMCID: PMC8860030 DOI: 10.3389/fcell.2021.826248] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid droplets (LD) have long been considered as mere fat drops; however, LD have lately been revealed to be ubiquitous, dynamic and to be present in diverse organelles in which they have a wide range of key functions. Although incompletely understood, the biogenesis of eukaryotic LD initiates with the synthesis of neutral lipids (NL) by enzymes located in the endoplasmic reticulum (ER). The accumulation of NL leads to their segregation into nanometric nuclei which then grow into lenses between the ER leaflets as they are further filled with NL. The lipid composition and interfacial tensions of both ER and the lenses modulate their shape which, together with specific ER proteins, determine the proneness of LD to bud from the ER toward the cytoplasm. The most important function of LD is the buffering of energy. But far beyond this, LD are actively integrated into physiological processes, such as lipid metabolism, control of protein homeostasis, sequestration of toxic lipid metabolic intermediates, protection from stress, and proliferation of tumours. Besides, LD may serve as platforms for pathogen replication and defense. To accomplish these functions, from biogenesis to breakdown, eukaryotic LD have developed mechanisms to travel within the cytoplasm and to establish contact with other organelles. When nutrient deprivation occurs, LD undergo breakdown (lipolysis), which begins with the LD-associated members of the perilipins family PLIN2 and PLIN3 chaperone-mediated autophagy degradation (CMA), a specific type of autophagy that selectively degrades a subset of cytosolic proteins in lysosomes. Indeed, PLINs CMA degradation is a prerequisite for further true lipolysis, which occurs via cytosolic lipases or by lysosome luminal lipases when autophagosomes engulf portions of LD and target them to lysosomes. LD play a crucial role in several pathophysiological processes. Increased accumulation of LD in non-adipose cells is commonly observed in numerous infectious diseases caused by intracellular pathogens including viral, bacterial, and parasite infections, and is gradually recognized as a prominent characteristic in a variety of cancers. This review discusses current evidence related to the modulation of LD biogenesis and breakdown caused by intracellular pathogens and cancer.
Collapse
Affiliation(s)
- Claudio M Fader Kaiser
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Patricia S Romano
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - M Cristina Vanrell
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Cristian A Pocognoni
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Julieta Jacob
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Benjamín Caruso
- Instituto de Investigaciones Biologicas y Tecnologicas, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Laura R Delgui
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| |
Collapse
|
38
|
Beekman KM, Regenboog M, Nederveen AJ, Bravenboer N, den Heijer M, Bisschop PH, Hollak CE, Akkerman EM, Maas M. Gender- and Age-Associated Differences in Bone Marrow Adipose Tissue and Bone Marrow Fat Unsaturation Throughout the Skeleton, Quantified Using Chemical Shift Encoding-Based Water-Fat MRI. Front Endocrinol (Lausanne) 2022; 13:815835. [PMID: 35574007 PMCID: PMC9094426 DOI: 10.3389/fendo.2022.815835] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/17/2022] [Indexed: 01/17/2023] Open
Abstract
Bone marrow adipose tissue (BMAT) is a dynamic tissue which is associated with osteoporosis, bone metastasis, and primary bone tumors. The aim of this study is to determine region-specific variations and age- and gender-specific differences in BMAT and BMAT composition in healthy subjects. In this cross-sectional study, we included 40 healthy subjects (26 male: mean age 49 years, range 22-75 years; 14 female: mean age 50 years, range 29-71) and determined the bone marrow signal fat fraction and bone marrow unsaturation in the spine (C3-L5), pelvis, femora, and tibiae using chemical shift encoding-based water-fat imaging (WFI) with multiple gradient echoes (mGRE). Regions of interest covered the individual vertebral bodies, pelvis and proximal epimetaphysis, diaphysis, and distal epimetaphysis of the femur and tibia. The spinal fat fraction increased from cervical to lumbar vertebral bodies (mean fat fraction ( ± SD or (IQR): cervical spine 0.37 ± 0.1; thoracic spine 0.41 ± 0.08. lumbar spine 0.46 ± 0.01; p < 0.001). The femoral fat fraction increased from proximal to distal (proximal 0.78 ± 0.09; diaphysis 0.86 (0.15); distal 0.93 ± 0.02; p < 0.001), while within the tibia the fat fraction decreased from proximal to distal (proximal 0.92 ± 0.01; diaphysis 0.91 (0.02); distal 0.90 ± 0.01; p < 0.001). In female subjects, age was associated with fat fraction in the spine, pelvis, and proximal femur (ρ = 0.88 p < 0.001; ρ = 0.87 p < 0.001; ρ = 0.63 p = 0.02; ρ = 0.74 p = 0.002, respectively), while in male subjects age was only associated with spinal fat fraction (ρ = 0.40 p = 0.04). Fat fraction and unsaturation were negatively associated within the spine (r = -0.40 p = 0.01), while in the extremities fat fraction and unsaturation were positively associated (distal femur: r = 0.42 p = 0.01; proximal tibia: r = 0.47, p = 0.002; distal tibia: r = 0.35 p = 0.03), both independent of age and gender. In conclusion, we confirm the distinct, age- and gender-dependent, distribution of BMAT throughout the human skeleton and we show that, contradicting previous animal studies, bone marrow unsaturation in human subjects is highest within the axial skeleton compared to the appendicular skeleton. Furthermore, we show that BMAT unsaturation was negatively correlated with BMAT within the spine, while in the appendicular skeleton, BMAT and BMAT unsaturation were positively associated.
Collapse
Affiliation(s)
- Kerensa M. Beekman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
- Department of Endocrinology, Amsterdam University Medical Centers, Amsterdam Movement Sciences, Vrije Universiteit University, Amsterdam, Netherlands
| | - Martine Regenboog
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Aart J. Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Research Laboratory Bone and Calcium Metabolism, Amsterdam University Medical Centers, Amsterdam Movement Sciences, Vrije Universiteit University, Amsterdam, Netherlands
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Martin den Heijer
- Department of Endocrinology, Amsterdam University Medical Centers, Amsterdam Movement Sciences, Vrije Universiteit University, Amsterdam, Netherlands
| | - Peter H. Bisschop
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Carla E. Hollak
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Erik M. Akkerman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Mario Maas
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam Movement Sciences, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Mario Maas,
| |
Collapse
|
39
|
Li Y, Cao S, Gaculenko A, Zhan Y, Bozec A, Chen X. Distinct Metabolism of Bone Marrow Adipocytes and their Role in Bone Metastasis. Front Endocrinol (Lausanne) 2022; 13:902033. [PMID: 35800430 PMCID: PMC9253270 DOI: 10.3389/fendo.2022.902033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/06/2022] [Indexed: 12/23/2022] Open
Abstract
Bone marrow adipocytes (BMAs) represent 10% of the total fat mass of the human body and serve as an energy reservoir for the skeletal niche. They function as an endocrine organ by actively secreting fatty acids, cytokines, and adipokines. The volume of BMAs increases along with age, osteoporosis and/or obesity. With the rapid development of multi-omic analysis and the advance in in vivo imaging technology, further distinct characteristics and functions of BMAs have been revealed. There is accumulating evidence that BMAs are metabolically, biologically and functionally unique from white, brown, beige and pink adipocytes. Bone metastatic disease is an uncurable complication in cancer patients, where primary cancer cells spread from their original site into the bone marrow. Recent publications have highlighted those BMAs could also serve as a rich lipid source of fatty acids that can be utilized by the cancer cells during bone metastasis, particularly for breast, prostate, lung, ovarian and pancreatic cancer as well as melanoma. In this review, we summarize the novel progressions in BMAs metabolism, especially with multi-omic analysis and in vivo imaging technology. We also update the metabolic role of BMAs in bone metastasis, and their potential new avenues for diagnosis and therapies against metastatic cancers.
Collapse
Affiliation(s)
- Yixuan Li
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Cao
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Anastasia Gaculenko
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Xiaoxiang Chen
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaoxiang Chen,
| |
Collapse
|
40
|
Li S, Wang B, Liang W, Chen Q, Wang W, Mei J, Zhang H, Liu Q, Yuan M. Associations Between Vertebral Marrow Proton Density Fat Fraction and Risk of Prostate Cancer. Front Endocrinol (Lausanne) 2022; 13:874904. [PMID: 35498437 PMCID: PMC9047738 DOI: 10.3389/fendo.2022.874904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Bone marrow adipocytes may be responsible for cancer progression. Although marrow adipogenesis is suspected to be involved in prostate carcinogenesis, an association between marrow adiposity and prostate cancer risk has not been clearly established in vivo. This work included 115 newly diagnosed cases of histologically confirmed prostate cancer (range, 48-79 years) and 87 age-matched healthy controls. Marrow proton density fat fraction (PDFF) was measured by 3.0-T MR spectroscopy at the spine lumbar. Associations between marrow PDFF and risk of prostate cancer by stage of disease and grade sub-types were performed using multivariable polytomous logistic regression. There were no significant group differences in the vertebral marrow PDFF, despite prostate cancer patients having 6.6% higher marrow PDFF compared to the healthy controls (61.7 ± 9.8% vs. 57.9 ± 6.5%; t = 1.429, p = 0.161). After adjusting for various clinical and demographic characteristics, we found that elevated marrow PDFF was related to an increased risk of high-grade prostate cancer [odds ratios (OR) = 1.31; 95% confidence interval (CI), 1.08-1.57; p = 0.003]. Likewise, increased marrow PDFF had a significantly positive correlation with aggressive prostate cancer risk (OR = 1.54; 95% CI, 1.13-1.92; p <0.001). There were no associations between marrow PDFF and low-grade (p = 0.314) or non-aggressive (p = 0.435) prostate cancer risk. The data support the hypothesis that marrow adiposity was correlated with increased risk of aggressive prostate cancer, supporting a link between adipogenesis and prostate cancer risk.
Collapse
Affiliation(s)
- Shaojun Li
- Department of Radiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Bo Wang
- Department of Radiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Wenwen Liang
- Department of Radiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Qi Chen
- Department of Radiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Wei Wang
- Department of Radiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jiangjun Mei
- Department of Ultrasound Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - He Zhang
- Department of Urology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Qianqian Liu
- Department of Laboratory Medicine, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Mingyuan Yuan
- Department of Radiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Mingyuan Yuan,
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW For solid tumours such as breast and prostate cancer, and haematological malignancies such as myeloma, bone represents a supportive home, where the cellular crosstalk is known to underlie both tumour growth and survival, and the development of the associated bone disease. The importance of metabolic reprogramming is becoming increasingly recognised, particularly within cancer biology, enabling tumours to adapt to changing environments and pressures. This review will discuss our current understanding of metabolic requirements and adaptations within the tumour-bone microenvironment. RECENT FINDINGS The bone provides a unique metabolic microenvironment, home to highly energy-intensive processes such as bone resorption and bone formation, both of which are dysregulated in the presence of cancer. Approaches such as metabolomics demonstrate metabolic plasticity in patients with advanced disease. Metabolic crosstalk between tumour cells and surrounding stroma supports disease pathogenesis. There is increasing evidence for a key role for metabolic reprogramming within the tumour-bone microenvironment to drive disease progression. As such, understanding these metabolic adaptations should reveal new therapeutic targets and approaches.
Collapse
Affiliation(s)
- Jessica Whitburn
- Nuffield Dept. of Surgical Sciences, University of Oxford, Oxford, UK
| | - Claire M Edwards
- Nuffield Dept. of Orthopaedics, Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford, UK.
- Botnar Research Centre, Old Road, University of Oxford, Oxford, OX3 7LD, UK.
| |
Collapse
|
42
|
Altuna-Coy A, Ruiz-Plazas X, Alves-Santiago M, Segarra-Tomás J, Chacón MR. Serum Levels of the Cytokine TWEAK Are Associated with Metabolic Status in Patients with Prostate Cancer and Modulate Cancer Cell Lipid Metabolism In Vitro. Cancers (Basel) 2021; 13:cancers13184688. [PMID: 34572917 PMCID: PMC8465414 DOI: 10.3390/cancers13184688] [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: 08/20/2021] [Revised: 09/01/2021] [Accepted: 09/14/2021] [Indexed: 11/29/2022] Open
Abstract
Simple Summary TWEAK is an inflammatory cytokine related to prostate cancer (PCa) progression that exerts its effects by engaging its cognate receptor Fn14. A soluble form of TWEAK (sTWEAK) has been detected in the PCa microenvironment. Altered levels of circulating sTWEAK are associated with aberrant glucose metabolism. We show that reduced serum levels of sTWEAK are associated with the metabolic status in patients with PCa and that the treatment of PC-3 cells with sTWEAK enhances the expression of genes related to lipid, but not to glucose, metabolism. sTWEAK also increases the lipid uptake and lipid accumulation in PC-3 cells. We corroborated that the observed effects were due to TWEAK/Fn14 engagement by silencing Fn14 expression, which attenuated the aberrant gene and protein expression. Additionally, we observed that the phosphorylation of ERK1/2 and AKT (ser473) were required for TWEAK/Fn14 actions. Thus, the contribution of the sTWEAK/Fn14 axis on PCa metabolism supports its potential as a therapeutic target for PCa. Abstract Soluble TWEAK (sTWEAK) has been proposed as a prognostic biomarker of prostate cancer (PCa). We found that reduced serum levels of sTWEAK, together with higher levels of prostate-specific antigen and a higher HOMA-IR index, are independent predictors of PCa. We also showed that sTWEAK stimulus failed to alter the expression of glucose transporter genes (SLC2A4 and SLC2A1), but significantly reduced the expression of glucose metabolism-related genes (PFK, HK1 and PDK4) in PCa cells. The sTWEAK stimulation of PC-3 cells significantly increased the expression of the genes related to lipogenesis (ACACA and FASN), lipolysis (CPT1A and PNPLA2), lipid transport (FABP4 and CD36) and lipid regulation (SREBP-1 and PPARG) and increased the lipid uptake. Silencing the TWEAK receptor (Fn14) in PC-3 cells confirmed the observed lipid metabolic effects, as shown by the downregulation of ACACA, FASN, CPT1A, PNPLA2, FABP4, CD36, SREBP-1 and PPARG expression, which was paralleled by a reduction of FASN, CPT1A and FABP4 protein expression. Specific-signaling inhibitor assays show that ERK1/2 and AKT (ser473) phosphorylation can regulate lipid metabolism-related genes in PCa cells, pointing to the AKT locus as a possible target for PCa. Overall, our data support sTWEAK/Fn14 axis as a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Antonio Altuna-Coy
- Disease Biomarkers and Molecular Mechanisms Group, IISPV, Joan XXIII University Hospital, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.A.-C.); (X.R.-P.); (M.A.-S.)
| | - Xavier Ruiz-Plazas
- Disease Biomarkers and Molecular Mechanisms Group, IISPV, Joan XXIII University Hospital, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.A.-C.); (X.R.-P.); (M.A.-S.)
- Urology Unit, Joan XXIII University Hospital, 43005 Tarragona, Spain
| | - Marta Alves-Santiago
- Disease Biomarkers and Molecular Mechanisms Group, IISPV, Joan XXIII University Hospital, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.A.-C.); (X.R.-P.); (M.A.-S.)
- Urology Unit, Joan XXIII University Hospital, 43005 Tarragona, Spain
| | - José Segarra-Tomás
- Disease Biomarkers and Molecular Mechanisms Group, IISPV, Joan XXIII University Hospital, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.A.-C.); (X.R.-P.); (M.A.-S.)
- Urology Unit, Joan XXIII University Hospital, 43005 Tarragona, Spain
- Correspondence: (J.S.-T.); (M.R.C.); Tel.: +34-977295500 (ext. 3406) (J.S.-T. & M.R.C.)
| | - Matilde R. Chacón
- Disease Biomarkers and Molecular Mechanisms Group, IISPV, Joan XXIII University Hospital, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (A.A.-C.); (X.R.-P.); (M.A.-S.)
- Correspondence: (J.S.-T.); (M.R.C.); Tel.: +34-977295500 (ext. 3406) (J.S.-T. & M.R.C.)
| |
Collapse
|
43
|
Ravichandran A, Meinert C, Bas O, Hutmacher DW, Bock N. Engineering a 3D bone marrow adipose composite tissue loading model suitable for studying mechanobiological questions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112313. [PMID: 34474864 DOI: 10.1016/j.msec.2021.112313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Tissue engineering strategies are widely used to model and study the bone marrow microenvironment in healthy and pathological conditions. Yet, while bone function highly depends on mechanical stimulation, the effects of biomechanical stimuli on the bone marrow niche, specifically on bone marrow adipose tissue (BMAT) is poorly understood due to a lack of representative in vitro loading models. Here, we engineered a BMAT analog made of a GelMA (gelatin methacryloyl) hydrogel/medical-grade polycaprolactone (mPCL) scaffold composite to structurally and biologically mimic key aspects of the bone marrow microenvironment, and exploited an innovative bioreactor to study the effects of mechanical loading. Highly reproducible BMAT analogs facilitated the successful adipogenesis of human mesenchymal bone marrow stem cells. Upon long-term intermittent stimulation (1 Hz, 2 h/day, 3 days/week, 3 weeks) in the novel bioreactor, cellular proliferation and lipid accumulation were similar to unloaded controls, yet there was a significant reduction in the secretion of adipokines including leptin and adiponectin, in line with clinical evidence of reduced adipokine expression following exercise/activity. Ultimately, this innovative loading platform combined with reproducibly engineered BMAT analogs provide opportunities to study marrow physiology in greater complexity as it accounts for the dynamic mechanical microenvironment context.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia
| | - Christoph Meinert
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Metro North Hospital and Health Service, Herston 4029, QLD, Australia
| | - Onur Bas
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove 4059, QLD, Australia
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; Bone and Joint Disorders Program, School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane 4000, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia; ARC Industrial Transformation Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing, QUT, Kelvin Grove 4059, QLD, Australia.
| |
Collapse
|
44
|
Hofbauer LC, Bozec A, Rauner M, Jakob F, Perner S, Pantel K. Novel approaches to target the microenvironment of bone metastasis. Nat Rev Clin Oncol 2021; 18:488-505. [PMID: 33875860 DOI: 10.1038/s41571-021-00499-9] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Bone metastases are a frequent and severe complication of advanced-stage cancers. Breast and prostate cancers, the most common malignancies in women and men, respectively, have a particularly high propensity to metastasize to bone. Conceptually, circulating tumour cells (CTCs) in the bloodstream and disseminated tumour cells (DTCs) in the bone marrow provide a snapshot of the dissemination and colonization process en route to clinically apparent bone metastases. Many cell types that constitute the bone microenvironment, including osteoblasts, osteocytes, osteoclasts, adipocytes, endothelial cells, haematopoietic stem cells and immune cells, engage in a dialogue with tumour cells. Some of these cells modify tumour biology, while others are disrupted and out-competed by tumour cells, thus leading to distinct phases of tumour cell migration, dormancy and latency, and therapy resistance and progression to overt bone metastases. Several current bone-protective therapies act by interrupting these interactions, mainly by targeting tumour cell-osteoclast interactions. In this Review, we describe the functional roles of the bone microenvironment and its components in the initiation and propagation of skeletal metastases, outline the biology and clinical relevance of CTCs and DTCs, and discuss established and future therapeutic approaches that specifically target defined components of the bone microenvironment to prevent or treat skeletal metastases.
Collapse
Affiliation(s)
- Lorenz C Hofbauer
- University Center for Healthy Aging, Dresden University of Technology, Dresden, Germany. .,Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) partner site Dresden, Dresden, Germany.
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Martina Rauner
- University Center for Healthy Aging, Dresden University of Technology, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Franz Jakob
- Department of Orthopedic Surgery, Julius Maximilians University of Würzburg, Würzburg, Germany.,Department of Functional Materials in Medicine and Dentistry, Julius Maximilians University of Würzburg, Würzburg, Germany
| | - Sven Perner
- Institute of Pathology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
45
|
Herroon MK, Mecca S, Haimbaugh A, Garmo LC, Rajagurubandara E, Todi SV, Baker TR, Podgorski I. Adipocyte-driven unfolded protein response is a shared transcriptomic signature of metastatic prostate carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119101. [PMID: 34280426 DOI: 10.1016/j.bbamcr.2021.119101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/17/2022]
Abstract
A critical unknown in the field of skeletal metastases is how cancer cells find a way to thrive under harsh conditions, as exemplified by metastatic colonization of adipocyte-rich bone marrow by prostate carcinoma cells. To begin understanding molecular processes that enable tumor cells to survive and progress in difficult microenvironments such as bone, we performed unbiased examination of the transcriptome of two different prostate cancer cell lines in the absence or presence of bone marrow adipocytes. Our RNAseq analyses and subsequent quantitative PCR and protein-based assays reveal that upregulation of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) genes is a shared signature between metastatic prostate carcinoma cell lines of different origin. Pathway analyses and pharmacological examinations highlight the ER chaperone BIP as an upstream coordinator of this transcriptomic signature. Additional patient-based data support our overall conclusion that ER stress and UPR induction are shared, important factors in the response and adaptation of metastatic tumor cells to their micro-environment. Our studies pave the way for additional mechanistic investigations and offer new clues towards effective therapeutic interventions in metastatic disease.
Collapse
Affiliation(s)
- Mackenzie K Herroon
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Shane Mecca
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Alex Haimbaugh
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Laimar C Garmo
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Erandi Rajagurubandara
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Department of Neurology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Tracie R Baker
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America.
| |
Collapse
|
46
|
Fontaine A, Bellanger D, Guibon R, Bruyère F, Brisson L, Fromont G. Lipophagy and prostate cancer: association with disease aggressiveness and proximity to periprostatic adipose tissue. J Pathol 2021; 255:166-176. [PMID: 34219239 DOI: 10.1002/path.5754] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/15/2021] [Accepted: 06/29/2021] [Indexed: 01/09/2023]
Abstract
The prostate gland is surrounded by periprostatic adipose tissue (PPAT), which is believed to play a role in prostate cancer (PCa) progression. Cancer cells can take up lipids from the microenvironment and store them in lipid droplets (LDs). Fatty acids released from LDs are used by PCa cells as preferential metabolic fuels to provide energy and promote cancer progression. Recently, fatty acids have been associated with autophagy, a cellular recycling pathway. Lipophagy is a selective form of autophagy involved in LD degradation, the role of which in PCa progression remains unknown. Here, we explored markers of autophagy and lipophagy in human PCa tissues in correlation with factors of aggressiveness, and we evaluated the influence of PPAT adipocytes on autophagy and lipophagy. We analyzed markers of autophagy (p62, LC3), lipid droplets (PLIN and Oil Red O), androgen receptor (AR), proliferation (Ki67), and epithelial-mesenchymal transition (Zeb1) on 465 PCa samples. Co-cultures of PCa cell lines PC3 and 22RV1 with adipocytes isolated from patients' PPAT were used to analyze the influence of PPAT on autophagy and lipophagy in vitro. In human PCa tissues, we observed a correlation between markers of LD and those of autophagy, which are associated with clinical and biological factors of disease aggressiveness. In addition, PLIN staining was associated with AR expression. In locally advanced PCa, p62, LC3, and PLIN were increased in extraprostatic areas where cancer cells are in contact with PPAT. Co-culture of PCa cell lines with adipocytes decreased autophagy activity and increased LD flux in PC3 cells. These results suggest an active process of lipophagy in PCa, linked to disease aggressiveness, to the proximity of PPAT, and induced in vitro in co-culture with adipocytes. Lipophagy is therefore likely to be a crucial player in PCa progression. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Alix Fontaine
- Department of Pathology, Bretonneau Hospital, CHU - University of Tours, Tours, France
| | | | - Roseline Guibon
- Department of Pathology, Bretonneau Hospital, CHU - University of Tours, Tours, France.,Inserm UMR U1069, Tours, France
| | - Franck Bruyère
- Department of Urology, Bretonneau Hospital, CHU - University of Tours, Tours, France
| | | | - Gaelle Fromont
- Department of Pathology, Bretonneau Hospital, CHU - University of Tours, Tours, France.,Inserm UMR U1069, Tours, France
| |
Collapse
|
47
|
Pan T, Lin SC, Lee YC, Yu G, Song JH, Pan J, Titus M, Satcher RL, Panaretakis T, Logothetis C, Yu-Lee LY, Lin SH. Statins reduce castration-induced bone marrow adiposity and prostate cancer progression in bone. Oncogene 2021; 40:4592-4603. [PMID: 34127814 PMCID: PMC8384136 DOI: 10.1038/s41388-021-01874-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/14/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023]
Abstract
A fraction of patients undergoing androgen deprivation therapy (ADT) for advanced prostate cancer (PCa) will develop recurrent castrate-resistant PCa (CRPC) in bone. Strategies to prevent CRPC relapse in bone are lacking. Here we show that the cholesterol-lowering drugs statins decrease castration-induced bone marrow adiposity in the tumor microenvironment and reduce PCa progression in bone. Using primary bone marrow stromal cells (BMSC) and M2-10B4 cells, we showed that ADT increases bone marrow adiposity by enhancing BMSC-to-adipocyte transition in vitro. Knockdown of androgen receptor abrogated BMSC-to-adipocyte transition, suggesting an androgen receptor-dependent event. RNAseq analysis showed that androgens reduce the secretion of adipocyte hormones/cytokines including leptin during BMSC-to-adipocyte transition. Treatment of PCa C4-2b, C4-2B4, and PC3 cells with leptin led to an increase in cell cycle progression and nuclear Stat3. RNAseq analysis also showed that androgens inhibit cholesterol biosynthesis pathway, raising the possibility that inhibiting cholesterol biosynthesis may decrease BMSC-to-adipocyte transition. Indeed, statins decreased BMSC-to-adipocyte transition in vitro and castration-induced bone marrow adiposity in vivo. Statin pre-treatment reduced 22RV1 PCa progression in bone after ADT. Our findings with statin may provide one of the mechanisms to the clinical correlations that statin use in patients undergoing ADT seems to delay progression to "lethal" PCa.
Collapse
Affiliation(s)
- Tianhong Pan
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Song-Chang Lin
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Yu-Chen Lee
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Guoyu Yu
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Pan
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Titus
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Satcher
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
48
|
Broadfield LA, Pane AA, Talebi A, Swinnen JV, Fendt SM. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Dev Cell 2021; 56:1363-1393. [PMID: 33945792 DOI: 10.1016/j.devcel.2021.04.013] [Citation(s) in RCA: 358] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/15/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Tumors undergo metabolic transformations to sustain uncontrolled proliferation, avoid cell death, and seed in secondary organs. An increased focus on cancer lipid metabolism has unveiled a number of mechanisms that promote tumor growth and survival, many of which are independent of classical cellular bioenergetics. These mechanisms include modulation of ferroptotic-mediated cell death, support during tumor metastasis, and interactions with the cells of the tumor microenvironment. As such, targeting lipid metabolism for anti-cancer therapies is attractive, with recent work on small-molecule inhibitors identifying compounds to target lipid metabolism. Here, we discuss these topics and identify open questions.
Collapse
Affiliation(s)
- Lindsay A Broadfield
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Antonino Alejandro Pane
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ali Talebi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
49
|
Wang C, Luo D. The metabolic adaptation mechanism of metastatic organotropism. Exp Hematol Oncol 2021; 10:30. [PMID: 33926551 PMCID: PMC8082854 DOI: 10.1186/s40164-021-00223-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
Metastasis is a complex multistep cascade of cancer cell extravasation and invasion, in which metabolism plays an important role. Recently, a metabolic adaptation mechanism of cancer metastasis has been proposed as an emerging model of the interaction between cancer cells and the host microenvironment, revealing a deep and extensive relationship between cancer metabolism and cancer metastasis. However, research on how the host microenvironment affects cancer metabolism is mostly limited to the impact of the local tumour microenvironment at the primary site. There are few studies on how differences between the primary and secondary microenvironments promote metabolic changes during cancer progression or how secondary microenvironments affect cancer cell metastasis preference. Hence, we discuss how cancer cells adapt to and colonize in the metabolic microenvironments of different metastatic sites to establish a metastatic organotropism phenotype. The mechanism is expected to accelerate the research of cancer metabolism in the secondary microenvironment, and provides theoretical support for the generation of innovative therapeutic targets for clinical metastatic diseases.
Collapse
Affiliation(s)
- Chao Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
50
|
Bazylianska V, Kalpage HA, Wan J, Vaishnav A, Mahapatra G, Turner AA, Chowdhury DD, Kim K, Morse PT, Lee I, Brunzelle JS, Polin L, Subedi P, Heath EI, Podgorski I, Marcus K, Edwards BF, Hüttemann M. Lysine 53 Acetylation of Cytochrome c in Prostate Cancer: Warburg Metabolism and Evasion of Apoptosis. Cells 2021; 10:802. [PMID: 33916826 PMCID: PMC8066186 DOI: 10.3390/cells10040802] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/28/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer is the second leading cause of cancer-related death in men. Two classic cancer hallmarks are a metabolic switch from oxidative phosphorylation (OxPhos) to glycolysis, known as the Warburg effect, and resistance to cell death. Cytochrome c (Cytc) is at the intersection of both pathways, as it is essential for electron transport in mitochondrial respiration and a trigger of intrinsic apoptosis when released from the mitochondria. However, its functional role in cancer has never been studied. Our data show that Cytc is acetylated on lysine 53 in both androgen hormone-resistant and -sensitive human prostate cancer xenografts. To characterize the functional effects of K53 modification in vitro, K53 was mutated to acetylmimetic glutamine (K53Q), and to arginine (K53R) and isoleucine (K53I) as controls. Cytochrome c oxidase (COX) activity analyzed with purified Cytc variants showed reduced oxygen consumption with acetylmimetic Cytc compared to the non-acetylated Cytc (WT), supporting the Warburg effect. In contrast to WT, K53Q Cytc had significantly lower caspase-3 activity, suggesting that modification of Cytc K53 helps cancer cells evade apoptosis. Cardiolipin peroxidase activity, which is another proapoptotic function of the protein, was lower in acetylmimetic Cytc. Acetylmimetic Cytc also had a higher capacity to scavenge reactive oxygen species (ROS), another pro-survival feature. We discuss our experimental results in light of structural features of K53Q Cytc, which we crystallized at a resolution of 1.31 Å, together with molecular dynamics simulations. In conclusion, we propose that K53 acetylation of Cytc affects two hallmarks of cancer by regulating respiration and apoptosis in prostate cancer xenografts.
Collapse
Affiliation(s)
- Viktoriia Bazylianska
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| | - Hasini A. Kalpage
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
| | - Junmei Wan
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
| | - Asmita Vaishnav
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| | - Gargi Mahapatra
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| | - Alice A. Turner
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| | - Dipanwita Dutta Chowdhury
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| | - Katherine Kim
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
| | - Paul T. Morse
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
| | - Icksoo Lee
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
- College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do 31116, Korea
| | - Joseph S. Brunzelle
- Life Sciences Collaborative Access Team, Center for Synchrotron Research, Northwestern University, Argonne, IL 60439, USA;
| | - Lisa Polin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA; (L.P.); (E.I.H.)
| | - Prabal Subedi
- Medical Proteomics/Bioanalytics-Center, Ruhr-University Bochum, 44789 Bochum, Germany; (P.S.); (K.M.)
| | - Elisabeth I. Heath
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA; (L.P.); (E.I.H.)
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA;
| | - Katrin Marcus
- Medical Proteomics/Bioanalytics-Center, Ruhr-University Bochum, 44789 Bochum, Germany; (P.S.); (K.M.)
| | - Brian F.P. Edwards
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (V.B.); (H.A.K.); (J.W.); (G.M.); (A.A.T.); (K.K.); (P.T.M.); (I.L.)
- Department of Biochemistry, Microbiology, and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (A.V.); (D.D.C.); (B.F.P.E.)
| |
Collapse
|