1
|
Fang CF, Li Y, Yang C, Fang H, Li C. Bioinformatics analysis of intrinsic drivers of immune dysregulation in multiple myeloma to elucidate immune phenotypes and discover prognostic gene signatures. Sci Rep 2025; 15:15662. [PMID: 40325058 PMCID: PMC12053621 DOI: 10.1038/s41598-025-00074-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 04/24/2025] [Indexed: 05/07/2025] Open
Abstract
Multiple myeloma (MM) progression is driven by immune dysregulation within the tumor microenvironment (TME). However, myeloma-intrinsic mechanisms underlying immune dysfunction remain poorly defined, and current immunotherapies show limited efficacy. Using RNA-seq data from 859 MM patients (MMRF-CoMMpass), we integrated xCELL, CIBERSORT, and ESTIMATE algorithms to deconvolute immune-stromal dynamics. Consensus clustering identified immune subtypes, followed by differential gene analysis and LASSO-Cox regression to construct a prognostic model validated in an independent cohort (GSE19784, N = 328). Immune Subtype Classification: Two subgroups emerged: Multiple myeloma-associated immune-related cluster 1 (N = 482): Immune-dysfunctional TME with Th2 cell enrichment, preadipocyte accumulation, and CXCL family suppression, linked to poor survival (P < 0.001). Multiple myeloma-associated immune-related cluster 2 (N = 377): Immune-active TME with cytotoxic CD8 + T/NK cell infiltration and favorable outcomes. Prognostic Gene Signature: Ten immune-related genes (UBE2T, E2F2, EXO1, SH2D2A, DRP2, WNT9A, SHROOM3, TMC8, CDCA7, and GPR132) predicted survival (The One-year AUC = 0.682 and The Over 5-years AUC = 0.714). We define a myeloma-intrinsic immune classification system and a 10-gene prognostic index, offering a framework for risk-stratified immunotherapy. Integration with flow cytometry could optimize precision treatment in MM.
Collapse
Affiliation(s)
- Chuan-Feng Fang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, 150001, People's Republic of China.
| | - Yan Li
- Department of Anesthesia, The Fourth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, 150001, People's Republic of China
| | - Chun Yang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, 150001, People's Republic of China
| | - Hua Fang
- Department of Medical Oncology, Fuxing Hospital of the Capital Medical University, Xicheng District, Beijing, 10001, People's Republic of China
| | - Chen Li
- Department of Bioengineering, the Hebei Agriculture University, Baoding City, Hebei Provence, 071001, People's Republic of China
| |
Collapse
|
2
|
Kamrani S, Naseramini R, Khani P, Razavi ZS, Afkhami H, Atashzar MR, Nasri F, Alavimanesh S, Saeidi F, Ronaghi H. Mesenchymal stromal cells in bone marrow niche of patients with multiple myeloma: a double-edged sword. Cancer Cell Int 2025; 25:117. [PMID: 40140850 PMCID: PMC11948648 DOI: 10.1186/s12935-025-03741-x] [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: 10/24/2024] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy defined by the abnormal proliferation and accumulation of plasma cells (PC) within the bone marrow (BM). While multiple myeloma impacts the bone, it is not classified as a primary bone cancer. The bone marrow microenvironment significantly influences the progression of myeloma and its treatment response. Mesenchymal stromal cells (MSCs) in this environment engage with myeloma cells and other bone marrow components via direct contact and the secretion of soluble factors. This review examines the established roles of MSCs in multiple facets of MM pathology, encompassing their pro-inflammatory functions, contributions to tumor epigenetics, effects on immune checkpoint inhibitors (ICIs), influence on reprogramming, chemotherapy resistance, and senescence. This review investigates the role of MSCs in the development and progression of MM.
Collapse
Affiliation(s)
- Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Naseramini
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Mohammad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hossein Ronaghi
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
3
|
Wang H, Zhang J, Ren H, Chen L, Ren J, Liu C, Wu H, Zhou L. Lipid metabolism in multiple myeloma: pathogenesis, therapeutic opportunities, and future directions. Front Oncol 2025; 15:1531928. [PMID: 40110197 PMCID: PMC11919907 DOI: 10.3389/fonc.2025.1531928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a complex hematological malignancy characterized by the clonal expansion of plasma cells in the bone marrow. Emerging studies have emphasized the importance of lipid metabolism, which is closely associated with the survival, proliferation, and drug resistance of tumor cells. The hypoxic environment in the bone marrow (BM) contributes to metabolic reprogramming in MM cells, including alterations in metabolite levels, changes in metabolic enzyme activity, and metabolic shifts. Cancer cells possess the ability to adapt their metabolism in order to fulfill their continuously increasing energy demands. In this review, we will discuss the alterations in lipid metabolism during the development of MM, and their reciprocal interactions with the tumor microenvironment.
Collapse
Affiliation(s)
- Huiquan Wang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiafeng Zhang
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hefei Ren
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lei Chen
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jigang Ren
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chang Liu
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Hongkun Wu
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lin Zhou
- Department of Laboratory Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Ghamlouch H, Gagler DC, Blaney P, Boyle EM, Wang Y, Avigan J, Choi J, Landgren O, Tsirigos A, Maura F, Morgan GJ, Davies FE. A proinflammatory response and polarized differentiation of stromal elements characterizes the murine myeloma bone marrow niche. Exp Hematol Oncol 2025; 14:22. [PMID: 40011943 DOI: 10.1186/s40164-025-00606-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND The bone marrow (BM) niche contains non-hematopoietic elements including mesenchymal stromal cells (MSC) and bone marrow endothelial cells (BMEC) which provide mechanical support, and control hematopoietic cell growth and differentiation. Although it is known that multiple myeloma (MM) cells interact closely with the BM microenvironment, little is known about the impact of MM on non-hematopoietic niche-forming cells. METHODS To address the role of the niche in MM pathogenesis, we utilized the 5TGM1 murine model. During the asymptomatic precursor stage of the model, we isolated the rare non-hematopoietic cells and performed single cell RNA sequencing. Using in-silico methods we characterized the individual cellular components of the niche, their relative abundance and differentiation state before and after exposure to MM cells as well as their intercellular interactions. RESULTS MM engraftment increased the abundance of MSC-lineage cells, BMECs and enhanced endothelial to mesenchymal transition. An inflammatory and oxidative stress signal was identified together with polarization of MSC differentiation away from osteocyte formation towards adipocytes which provide growth factors that are known to support MM expansion. BMEC differentiation was polarized towards sinusoidal endothelial cells with a pro-angiogenic/pro-inflammatory phenotype. CONCLUSIONS MM cells impact the BM niche by generating a pro-inflammatory microenvironment with MSC differentiation being changed to generate cell subsets that favor MM growth and survival. In order to induce remission and improve long-term outcome for MM patients these inflammatory and oxidative stress signals need to be reduced and normal niche differentiation trajectories restored.
Collapse
Affiliation(s)
- Hussein Ghamlouch
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA.
| | - Dylan C Gagler
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
- Applied Bioinformatics Laboratories (ABL), NYU Langone Medical Center, New York, NY, USA
| | - Patrick Blaney
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
- Applied Bioinformatics Laboratories (ABL), NYU Langone Medical Center, New York, NY, USA
| | - Eileen M Boyle
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
- Cancer Institute, University College London, London, UK
- Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Yubao Wang
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
| | - Jason Avigan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
| | - Jinyoung Choi
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories (ABL), NYU Langone Medical Center, New York, NY, USA
| | - Francesco Maura
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Gareth J Morgan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA
| | - Faith E Davies
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Health, 522 First Avenue, MSB4, New York, NY, 10016, USA.
| |
Collapse
|
5
|
Xu Q, Hou W, Zhao B, Fan P, Wang S, Wang L, Gao J. Mesenchymal stem cells lineage and their role in disease development. Mol Med 2024; 30:207. [PMID: 39523306 PMCID: PMC11552129 DOI: 10.1186/s10020-024-00967-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are widely dispersed in vivo and are isolated from several tissues, including bone marrow, heart, body fluids, skin, and perinatal tissues. Bone marrow MSCs have a multidirectional differentiation potential, which can be induced to differentiate the medium in a specific direction or by adding specific regulatory factors. MSCs repair damaged tissues through lineage differentiation, and the ex vivo transplantation of bone marrow MSCs can heal injured sites. MSCs have different propensities for lineage differentiation and pathological evolution for different diseases, which are crucial in disease progression. In this study, we describe various lineage analysis methods to explore lineage ontology in vitro and in vivo, elucidate the impact of MSC lineage differentiation on diseases, advance our understanding of the role of MSC differentiation in physiological and pathological states, and explore new targets and ideas associated with disease diagnosis and treatment.
Collapse
Affiliation(s)
- Qi Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wenrun Hou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Baorui Zhao
- Stem cell Translational laboratory, Shanxi Technological Innovation Center for Clinical Diagnosis and Treatment of Immune and Rheumatic Diseases, Shanxi Bethune Hospital, Tongji Shanxi Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Peixin Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Sheng Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Lei Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
6
|
Zhang H, Sun F, Jiang S, Yang F, Dong X, Liu G, Wang M, Li Y, Su M, Wen Z, Yu C, Fan C, Li X, Zhang Z, Yang L, Li B. METTL protein family: focusing on the occurrence, progression and treatment of cancer. Biomark Res 2024; 12:105. [PMID: 39289775 PMCID: PMC11409517 DOI: 10.1186/s40364-024-00652-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
Methyltransferase-like protein is a ubiquitous enzyme-like protein in the human body, with binding domains for nucleic acids, proteins and other small molecules, and plays an important role in a variety of biological behaviours in normal organisms and diseases, characterised by the presence of a methyltransferase-like structural domain and a structurally conserved SAM-binding domain formed by the seven-stranded β-fold structure in the center of the protein. With the deepening of research, the METTL protein family has been found to be abnormally expressed in a variety of tumor diseases, and the clarification of its relationship with tumor diseases can be used as a molecular therapeutic target and has an important role in the prognosis of tumors. In this paper, we review the structure, biological process, immunotherapy, drug-targeted therapy, and markers of the METTL protein family to provide new ideas for the diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Huhu Zhang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Fulin Sun
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Shuyao Jiang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Fanghao Yang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Xiaolei Dong
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Guoxiang Liu
- Department of Clinical Laboratory, Weifang People's Hospital, 151, Guangwen Streer, Weifang, 261041, China
| | - Mengjun Wang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ya Li
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Mohan Su
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Ziyuan Wen
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Chunjuan Yu
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Chenkai Fan
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Xiaoxia Li
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Zhe Zhang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Lina Yang
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Bing Li
- Department of Genetics and Cell Biology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
- Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
7
|
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
|
8
|
Feng D, Wang Z, Cao S, Xu H, Li S. Identification of lipid metabolism-related gene signature in the bone marrow microenvironment of multiple myelomas through deep analysis of transcriptomic data. Clin Exp Med 2024; 24:136. [PMID: 38916672 PMCID: PMC11199273 DOI: 10.1007/s10238-024-01398-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
Dysregulated lipid metabolism in the bone marrow microenvironment (BMM) plays a vital role in multiple myeloma (MM) development, progression, and drug resistance. However, the exact mechanism by which lipid metabolism impacts the BMM, promotes tumorigenesis, and triggers drug resistance remains to be fully elucidated.By analyzing the bulk sequencing and single-cell sequencing data of MM patients, we identified lipid metabolism-related genes differential expression significantly associated with MM prognosis, referred to as LMRPgenes. Using a cohort of ten machine learning algorithms and 117 combinations, LMRPgenes predictive models were constructed. Further exploration of the effects of the model risk score (RS) on the survival status, immune status of patients with BMM, and response to immunotherapy was conducted. The study also facilitated the identification of personalized therapeutic strategies targeting specified risk categories within patient cohorts.Analysis of the scRNA-seq data revealed increased lipid metabolism-related gene enrichment scores (LMESs) in erythroblasts and progenitor, malignant, and Tprolif cells but decreased LMESs in lymphocytes. LMESs were also strongly correlated with most of the 50 hallmark pathways within these cell populations. An elevated malignant cell ratio and reduced lymphocytes were observed in the high LMES group. Moreover, the LMRPgenes predictive model, consisting of 14 genes, showed great predictive power. The risk score emerged as an independent indicator of poor outcomes. Inverse relationships between the RS and immune status were noted, and a high RS was associated with impaired immunotherapy responses. Drug sensitivity assays indicated the effectiveness of bortezomib, buparlisib, dinaciclib, staurosporine, rapamycin, and MST-312 in the high-RS group, suggesting their potential for treating patients with high-RS values and poor response to immunotherapy. Ultimately, upon verification via qRT-PCR, we observed a significant upregulation of ACBD6 in NDMM group compared to the control group.Our research enhances the knowledge base regarding the association between lipid metabolism-related genes (LMRGs) and the BMM in MM patients, offering substantive insights into the mechanistic effects of the BMM mediated by LMRGs.
Collapse
Affiliation(s)
- Dan Feng
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China
| | - Zhen Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China
| | - Shengji Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China
| | - Hui Xu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150000, Heilongjiang, China
| | - Shijun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China.
- College of Laboratory Medicine, Dalian Medical University, Liaoning, Dalian, 116044, China.
| |
Collapse
|
9
|
Kumar V, Stewart JH. Obesity, bone marrow adiposity, and leukemia: Time to act. Obes Rev 2024; 25:e13674. [PMID: 38092420 DOI: 10.1111/obr.13674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/07/2023] [Accepted: 11/13/2023] [Indexed: 02/28/2024]
Abstract
Obesity has taken the face of a pandemic with less direct concern among the general population and scientific community. However, obesity is considered a low-grade systemic inflammation that impacts multiple organs. Chronic inflammation is also associated with different solid and blood cancers. In addition, emerging evidence demonstrates that individuals with obesity are at higher risk of developing blood cancers and have poorer clinical outcomes than individuals in a normal weight range. The bone marrow is critical for hematopoiesis, lymphopoiesis, and myelopoiesis. Therefore, it is vital to understand the mechanisms by which obesity-associated changes in BM adiposity impact leukemia development. BM adipocytes are critical to maintain homeostasis via different means, including immune regulation. However, obesity increases BM adiposity and creates a pro-inflammatory environment to upregulate clonal hematopoiesis and a leukemia-supportive environment. Obesity further alters lymphopoiesis and myelopoiesis via different mechanisms, which dysregulate myeloid and lymphoid immune cell functions mentioned in the text under different sequentially discussed sections. The altered immune cell function during obesity alters hematological malignancies and leukemia susceptibility. Therefore, obesity-induced altered BM adiposity, immune cell generation, and function impact an individual's predisposition and severity of leukemia, which should be considered a critical factor in leukemia patients.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - John H Stewart
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Allegra A, Murdaca G, Mirabile G, Gangemi S. Protective Effects of High-Density Lipoprotein on Cancer Risk: Focus on Multiple Myeloma. Biomedicines 2024; 12:514. [PMID: 38540127 PMCID: PMC10967848 DOI: 10.3390/biomedicines12030514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 04/03/2025] Open
Abstract
Lipid metabolism is intrinsically linked to tumorigenesis. And one of the most important characteristics of cancer is the modification of lipid metabolism and its correlation with oncogenic signaling pathways within the tumors. Because lipids function as signaling molecules, membrane structures, and energy sources, lipids are essential to the development of cancer. Above all, the proper immune response of tumor cells depends on the control of lipid metabolism. Changes in metabolism can modify systems that regulate carcinogenesis, such as inflammation, oxidative stress, and angiogenesis. The dependence of various malignancies on lipid metabolism varies. This review delves into the modifications to lipid metabolism that take place in cancer, specifically focusing on multiple myeloma. The review illustrates how changes in different lipid pathways impact the growth, survival, and drug-responsiveness of multiple myeloma cells, in addition to their interactions with other cells within the tumor microenvironment. The phenotype of malignant plasma cells can be affected by lipid vulnerabilities, and these findings offer a new avenue for understanding this process. Additionally, they identify novel druggable pathways that have a major bearing on multiple myeloma care.
Collapse
Affiliation(s)
- Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (A.A.); (G.M.)
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 16132 Genova, Italy
- Allergology and Clinical Immunology Unit, San Bartolomeo Hospital, 19038 Sarzana, Italy
| | - Giuseppe Mirabile
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (A.A.); (G.M.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|
11
|
Wei X, Zhang Y, Wang Z, He Y, Ju S, Fu J. Bone marrow adipocytes is a new player in supporting myeloma cells proliferation and survival in myeloma microenvironment. Transl Oncol 2024; 40:101856. [PMID: 38134840 PMCID: PMC10776777 DOI: 10.1016/j.tranon.2023.101856] [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: 10/11/2023] [Revised: 11/09/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Multiple myeloma (MM) is a lethal B cell neoplasm characterized by clonal expansion of malignant plasma cells in the bone marrow and remains incurable due to disease relapse and drug resistance. Bone marrow adipocytes (BMAs) are emerging as playing active functions that can support myeloma cell growth and survival. The aim of this study is to investigate myeloma-mesenchymal stem cells (MSCs) interaction and the impact of such interactions on the pathogenesis of MM using in vitro co-culture assay. Here we provide evidence that MM cell up-regulated MSCs to express PPAR-γ and pushes MSCs differentiation toward adipocytes at the expense of osteoblasts in co-culture manner. The increased BMAs can effectively enhance MM cell to proliferation, migration, and chemoresistance via cell-cell contact and/or cytokines release regulated by PPAR-γ signal pathway. This effect was partially reversed in medium containing PPAR-γ antagonist G3335 and indicated that G3335 distorts the maturation of MSC-derived adipocytes and cytokines release by adipocytes through inhibition of PPAR-γ, a key transcriptional factor for the activation of adipogenesis, or cell to cell contact, or both. In meantime, we observed higher expression of adipocyte differentiation associated genes DLK1, DGAT1, FABP4, and FASN both in MSCs and MSC derived adipocytes, but the osteoblast differentiation-associated gene ALP was down regulated in MSCs. These finding mean that direct consequence of MM/MSC interaction that play a role in MM pathogenesis. Consistent with those in vitro results, our primary clinical observation also showed that bone marrow samples from MM patients had significantly higher bone adiposity in comparison with controls and the number of adipocytes decreased in those who were response to anti-MM therapy. Our finding suggested that BMAs may have an important contribution to MM progression, particularly in drugs resistant of MM cells, and plays an important contribution in MM bone disease and treatment failure, but more clinical studies are needed to confirm its role.
Collapse
Affiliation(s)
- Xiaoqian Wei
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Yangmin Zhang
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Ziyan Wang
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Yuanning He
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Songguang Ju
- Institute of Biotechnology, Soochow University, Suzhou 215007, PR China
| | - Jinxiang Fu
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China.
| |
Collapse
|
12
|
Torcasio R, Gallo Cantafio ME, Ikeda RK, Ganino L, Viglietto G, Amodio N. Lipid metabolic vulnerabilities of multiple myeloma. Clin Exp Med 2023; 23:3373-3390. [PMID: 37639069 PMCID: PMC10618328 DOI: 10.1007/s10238-023-01174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy worldwide, characterized by abnormal proliferation of malignant plasma cells within a tumor-permissive bone marrow microenvironment. Metabolic dysfunctions are emerging as key determinants in the pathobiology of MM. In this review, we highlight the metabolic features of MM, showing how alterations in various lipid pathways, mainly involving fatty acids, cholesterol and sphingolipids, affect the growth, survival and drug responsiveness of MM cells, as well as their cross-talk with other cellular components of the tumor microenvironment. These findings will provide a new path to understanding the mechanisms underlying how lipid vulnerabilities may arise and affect the phenotype of malignant plasma cells, highlighting novel druggable pathways with a significant impact on the management of MM.
Collapse
Affiliation(s)
- Roberta Torcasio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
- Department of Biology, Ecology and Heart Sciences, University of Calabria, Arcavacata Di Rende, Cosenza, Italy
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Raissa Kaori Ikeda
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
- Centro Universitário São Camilo, São Paulo, Brazil
| | - Ludovica Ganino
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Viale Europa, Campus Germaneto, 88100, Catanzaro, Italy.
| |
Collapse
|
13
|
Gudgeon N, Giles H, Bishop EL, Fulton-Ward T, Escribano-Gonzalez C, Munford H, James-Bott A, Foster K, Karim F, Jayawardana D, Mahmood A, Cribbs AP, Tennant DA, Basu S, Pratt G, Dimeloe S. Uptake of long-chain fatty acids from the bone marrow suppresses CD8+ T-cell metabolism and function in multiple myeloma. Blood Adv 2023; 7:6035-6047. [PMID: 37276076 PMCID: PMC10582277 DOI: 10.1182/bloodadvances.2023009890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/25/2023] [Accepted: 05/19/2023] [Indexed: 06/07/2023] Open
Abstract
T cells demonstrate impaired function in multiple myeloma (MM) but suppressive mechanisms in the bone marrow microenvironment remain poorly defined. We observe that bone marrow CD8+ T-cell function is decreased in MM compared with controls, and is also consistently lower within bone marrow samples than in matched peripheral blood samples. These changes are accompanied by decreased mitochondrial mass and markedly elevated long-chain fatty acid uptake. In vitro modeling confirmed that uptake of bone marrow lipids suppresses CD8+ T function, which is impaired in autologous bone marrow plasma but rescued by lipid removal. Analysis of single-cell RNA-sequencing data identified expression of fatty acid transport protein 1 (FATP1) in bone marrow CD8+ T cells in MM, and FATP1 blockade also rescued CD8+ T-cell function, thereby identifying this as a novel target to augment T-cell activity in MM. Finally, analysis of samples from cohorts of patients who had received treatment identified that CD8+ T-cell metabolic dysfunction resolves in patients with MM who are responsive to treatment but not in patients with relapsed MM, and is associated with substantial T-cell functional restoration.
Collapse
Affiliation(s)
- Nancy Gudgeon
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Hannah Giles
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Trust, Birmingham, United Kingdom
| | - Emma L. Bishop
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Taylor Fulton-Ward
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Cristina Escribano-Gonzalez
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Haydn Munford
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Anna James-Bott
- Nuffield Department of Orthopaedics, Botnar Research Centre, Rheumatology and Musculoskeletal Sciences, National Institute of Health Research Oxford Biomedical Research Unit, University of Oxford, Oxford, United Kingdom
| | - Kane Foster
- Research Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Farheen Karim
- Clinical Haematology Unit, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom
| | - Dedunu Jayawardana
- Clinical Haematology Unit, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom
| | - Ansar Mahmood
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Trust, Birmingham, United Kingdom
| | - Adam P. Cribbs
- Nuffield Department of Orthopaedics, Botnar Research Centre, Rheumatology and Musculoskeletal Sciences, National Institute of Health Research Oxford Biomedical Research Unit, University of Oxford, Oxford, United Kingdom
| | - Daniel A. Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Supratik Basu
- Clinical Haematology Unit, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom
| | - Guy Pratt
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Clinical Haematology, University Hospitals Birmingham NHS Trust, Birmingham, United Kingdom
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
14
|
Choa R, Panaroni C, Bhatia R, Raje N. It is worth the weight: obesity and the transition from monoclonal gammopathy of undetermined significance to multiple myeloma. Blood Adv 2023; 7:5510-5523. [PMID: 37493975 PMCID: PMC10515310 DOI: 10.1182/bloodadvances.2023010822] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
The overweight/obesity epidemic is a serious public health concern that affects >40% of adults globally and increases the risk of numerous chronic diseases, such as type 2 diabetes, heart disease, and various cancers. Multiple myeloma (MM) is a lymphohematopoietic cancer caused by the uncontrolled clonal expansion of plasma cells. Recent studies have shown that obesity is a risk factor not only for MM but also monoclonal gammopathy of undetermined significance (MGUS), a precursor disease state of MM. Furthermore, obesity may promote the transition from MGUS to MM. Thus, in this review, we summarize the epidemiological evidence regarding the role of obesity in MM and MGUS, discuss the biologic mechanisms that drive these disease processes, and detail the obesity-targeted pharmacologic and lifestyle interventions that may reduce the risk of progression from MGUS to MM.
Collapse
Affiliation(s)
- Ruth Choa
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Cristina Panaroni
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Roma Bhatia
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Noopur Raje
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
15
|
Ochiai M, Fierstein S, XsSali F, DeVito N, Purkey LR, May R, Correa-Medina A, Kelley M, Page TD, DeCicco-Skinner K. Unlocking Drug Resistance in Multiple Myeloma: Adipocytes as Modulators of Treatment Response. Cancers (Basel) 2023; 15:4347. [PMID: 37686623 PMCID: PMC10486466 DOI: 10.3390/cancers15174347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological malignancy characterized by the clonal proliferation of malignant plasma cells. Despite the development of a diverse array of targeted drug therapies over the last decade, patients often relapse and develop refractory disease due to multidrug resistance. Obesity is a growing public health threat and a risk factor for multiple myeloma, although the mechanisms by which obesity contributes to MM growth and progression have not been fully elucidated. In the present study, we evaluated whether crosstalk between adipocytes and MM cells promoted drug resistance and whether this was amplified by obesity. Human adipose-derived stem cells (ASCs) from nineteen normal (BMI = 20-25 kg/m2), overweight (25-30 kg/m2), or obese (30-35 kg/m2) patients undergoing elective liposuction were utilized. Cells were differentiated into adipocytes, co-cultured with RPMI 8226 or U266B1 multiple myeloma cell lines, and treated with standard MM therapies, including bortezomib or a triple combination of bortezomib, dexamethasone, and lenalidomide. We found that adipocytes from overweight and obese individuals increased cell adhesion-mediated drug resistance (CAM-DR) survival signals in MM cells, and P-glycoprotein (P-gp) and multidrug resistance-associated protein (MRP) drug transporter expression. Further, co-culture enhanced in vitro angiogenesis, MMP-2 activity, and protected MM cells from drug-induced decreases in viability. In summary, we provide an underlying mechanism by which obesity can impair the drug response to MM and allow for recurrence and/or disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kathleen DeCicco-Skinner
- Department of Biology, American University, 4400 Massachusetts Ave, NW, Washington, DC 20016, USA
| |
Collapse
|
16
|
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
|
17
|
Wieder R. Awakening of Dormant Breast Cancer Cells in the Bone Marrow. Cancers (Basel) 2023; 15:cancers15113021. [PMID: 37296983 DOI: 10.3390/cancers15113021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Up to 40% of patients with breast cancer (BC) have metastatic cells in the bone marrow (BM) at the initial diagnosis of localized disease. Despite definitive systemic adjuvant therapy, these cells survive in the BM microenvironment, enter a dormant state and recur stochastically for more than 20 years. Once they begin to proliferate, recurrent macrometastases are not curable, and patients generally succumb to their disease. Many potential mechanisms for initiating recurrence have been proposed, but no definitive predictive data have been generated. This manuscript reviews the proposed mechanisms that maintain BC cell dormancy in the BM microenvironment and discusses the data supporting specific mechanisms for recurrence. It addresses the well-described mechanisms of secretory senescence, inflammation, aging, adipogenic BM conversion, autophagy, systemic effects of trauma and surgery, sympathetic signaling, transient angiogenic bursts, hypercoagulable states, osteoclast activation, and epigenetic modifications of dormant cells. This review addresses proposed approaches for either eliminating micrometastases or maintaining a dormant state.
Collapse
Affiliation(s)
- Robert Wieder
- Rutgers New Jersey Medical School and the Cancer Institute of New Jersey, 185 South Orange Avenue, MSB F671, Newark, NJ 07103, USA
| |
Collapse
|
18
|
Austin MJ, Kalampalika F, Cawthorn WP, Patel B. Turning the spotlight on bone marrow adipocytes in haematological malignancy and non-malignant conditions. Br J Haematol 2023; 201:605-619. [PMID: 37067783 PMCID: PMC10952811 DOI: 10.1111/bjh.18748] [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: 12/20/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 04/18/2023]
Abstract
Whilst bone marrow adipocytes (BMAd) have long been appreciated by clinical haemato-pathologists, it is only relatively recently, in the face of emerging data, that the adipocytic niche has come under the watchful eye of biologists. There is now mounting evidence to suggest that BMAds are not just a simple structural entity of bone marrow microenvironments but a bona fide driver of physio- and pathophysiological processes relevant to multiple aspects of health and disease. Whilst the truly multifaceted nature of BMAds has only just begun to emerge, paradigms have shifted already for normal, malignant and non-malignant haemopoiesis incorporating a view of adipocyte regulation. Major efforts are ongoing, to delineate the routes by which BMAds participate in health and disease with a final aim of achieving clinical tractability. This review summarises the emerging role of BMAds across the spectrum of normal and pathological haematological conditions with a particular focus on its impact on cancer therapy.
Collapse
Affiliation(s)
- Michael J. Austin
- Barts Cancer Institute, Centre for Haemato‐OncologyQueen Mary University of LondonLondonUK
| | - Foteini Kalampalika
- Barts Cancer Institute, Centre for Haemato‐OncologyQueen Mary University of LondonLondonUK
| | - William P. Cawthorn
- BHF/University Centre for Cardiovascular Science, Edinburgh BioquarterUniversity of EdinburghEdinburghUK
| | - Bela Patel
- Barts Cancer Institute, Centre for Haemato‐OncologyQueen Mary University of LondonLondonUK
| |
Collapse
|
19
|
Matamala Montoya M, van Slobbe GJJ, Chang JC, Zaal EA, Berkers CR. Metabolic changes underlying drug resistance in the multiple myeloma tumor microenvironment. Front Oncol 2023; 13:1155621. [PMID: 37091139 PMCID: PMC10117897 DOI: 10.3389/fonc.2023.1155621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells in the bone marrow (BM). MM remains an incurable disease, with the majority of patients experiencing multiple relapses from different drugs. The MM tumor microenvironment (TME) and in particular bone-marrow stromal cells (BMSCs) play a crucial role in the development of drug resistance. Metabolic reprogramming is emerging as a hallmark of cancer that can potentially be exploited for cancer treatment. Recent studies show that metabolism is further adjusted in MM cells during the development of drug resistance. However, little is known about the role of BMSCs in inducing metabolic changes that are associated with drug resistance. In this Perspective, we summarize current knowledge concerning the metabolic reprogramming of MM, with a focus on those changes associated with drug resistance to the proteasome inhibitor Bortezomib (BTZ). In addition, we present proof-of-concept fluxomics (glucose isotope-tracing) and Seahorse data to show that co-culture of MM cells with BMSCs skews the metabolic phenotype of MM cells towards a drug-resistant phenotype, with increased oxidative phosphorylation (OXPHOS), serine synthesis pathway (SSP), TCA cycle and glutathione (GSH) synthesis. Given the crucial role of BMSCs in conveying drug resistance, insights into the metabolic interaction between MM and BMSCs may ultimately aid in the identification of novel metabolic targets that can be exploited for therapy.
Collapse
Affiliation(s)
- María Matamala Montoya
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Gijs J. J. van Slobbe
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jung-Chin Chang
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Esther A. Zaal
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Celia R. Berkers, ; Esther A. Zaal,
| | - Celia R. Berkers
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Celia R. Berkers, ; Esther A. Zaal,
| |
Collapse
|
20
|
Tentolouris A, Ntanasis-Stathopoulos I, Terpos E. Obesity and multiple myeloma: emerging mechanisms and perspectives. Semin Cancer Biol 2023; 92:45-60. [PMID: 37030643 DOI: 10.1016/j.semcancer.2023.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/08/2023]
Abstract
Obesity is a global pandemic that has been associated with the development of breast, endometrial, large intestine, renal, esophageal, and pancreatic cancer. Obesity is also involved in the development of cardiovascular disease and type 2 diabetes mellitus. Recently, an increase in the incidence of obesity-related cancers has been reported. Multiple myeloma (MM) is the second most common hematological malignancy, after lymphoma. The aim of this review is to examine the epidemiological data on obesity and MM, assess the effect of obesity on MM outcomes, evaluate the possible mechanisms through which obesity might increase the incidence of MM and provide the effects of obesity management on MM. Current evidence indicates that obesity may have an impact on the progression of monoclonal gammopathy of undetermined significance (MGUS) to MM and increase the prevalence of MM. However, data regarding the effect of obesity on MGUS incidence are controversial; further studies are needed to examine whether obesity affects the development of MGUS or the progression of MGUS to MM. In addition, obesity affects MM outcomes. Increased BMI is associated with decreased survival in patients with MM, while data regarding the effect of obesity on newly diagnosed MM subjects and autologous stem cell transplantation are limited. Interestingly, the obesity paradox may also apply to patients with relapsed/refractory MM who are overweight or obese, because they may have a survival advantage. The pathophysiological pathways linking obesity to MM are very complicated and include bone marrow adipose tissue; adipokines, such as adiponectin, leptin, resistin, and visfatin; inflammatory cytokines and growth factors, such as TNF-α and IL-6; hormones including insulin and the insulin-like growth factor system as well as sex hormones. In terms of the effect of pharmacological management of obesity, orlistat has been shown to alter the proliferation of MM cells, whereas no data exist on glucagon-like peptide-1 receptor agonists, naltrexone/bupropion, or phentermine/topiramate. Bariatric surgery may be associated with a reduction in the incidence of MM, however, further studies are needed.
Collapse
|
21
|
Wieder R. Fibroblasts as Turned Agents in Cancer Progression. Cancers (Basel) 2023; 15:2014. [PMID: 37046676 PMCID: PMC10093070 DOI: 10.3390/cancers15072014] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Differentiated epithelial cells reside in the homeostatic microenvironment of the native organ stroma. The stroma supports their normal function, their G0 differentiated state, and their expansion/contraction through the various stages of the life cycle and physiologic functions of the host. When malignant transformation begins, the microenvironment tries to suppress and eliminate the transformed cells, while cancer cells, in turn, try to resist these suppressive efforts. The tumor microenvironment encompasses a large variety of cell types recruited by the tumor to perform different functions, among which fibroblasts are the most abundant. The dynamics of the mutual relationship change as the sides undertake an epic battle for control of the other. In the process, the cancer "wounds" the microenvironment through a variety of mechanisms and attracts distant mesenchymal stem cells to change their function from one attempting to suppress the cancer, to one that supports its growth, survival, and metastasis. Analogous reciprocal interactions occur as well between disseminated cancer cells and the metastatic microenvironment, where the microenvironment attempts to eliminate cancer cells or suppress their proliferation. However, the altered microenvironmental cells acquire novel characteristics that support malignant progression. Investigations have attempted to use these traits as targets of novel therapeutic approaches.
Collapse
Affiliation(s)
- Robert Wieder
- Rutgers New Jersey Medical School and the Cancer Institute of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
22
|
Bone Marrow Adipose Tissue: Regulation of Osteoblastic Niche, Hematopoiesis and Hematological Malignancies. Stem Cell Rev Rep 2023:10.1007/s12015-023-10531-3. [PMID: 36930385 DOI: 10.1007/s12015-023-10531-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Abstract
Bone marrow adipose tissue (BMAT) creates a specific microniche within multifunctional bone marrow (BM) ecosystem which imposes changes in surrounding cells and at systemic level. Moreover, BMAT contributes to spatial and temporal separation and metabolic compartmentalization of BM, thus regulating BM homeostasis and diseases. Recent findings have identified novel progenitor subsets of bone marrow adipocytes (BMAd)s recruited during the BM adipogenesis within different skeletal and hematopoietic stem cell niches. Potential of certain mesenchymal BM cells to differentiate into both osteogenic and adipogenic lineages, contributes to the complex interplay of BMAT with endosteal (osteoblastic) niche compartments as an important cellular player in bone tissue homeostasis. Targeting and ablation of BMAT cells at certain states might be an optional and promising strategy for improvement of bone health. Additionally, recent findings demonstrated spatial distribution of BMAds related to hematopoietic cells and pointed out important functional roles in the vital processes such as long-term hematopoiesis. BM adipogenesis appears to be an emergency phenomenon that follows the production of hematopoietic stem and progenitor cell niche factors, thus regulating physiological, stressed, and malignant hematopoiesis. Lipolytic and secretory activity of BMAds can influence survival and proliferation of hematopoietic cells at different maturation stages. Due to their different lipid status, constitutive and regulated BMAds are important determinants of normal and malignant hematopoietic cells. Further elucidation of cellular and molecular players involved in BMAT expansion and crosstalk with malignant cells is of paramount importance for conceiving the new therapies for improvement of BM health.
Collapse
|
23
|
Zhang H, Liesveld JL, Calvi LM, Lipe BC, Xing L, Becker MW, Schwarz EM, Yeh SCA. The roles of bone remodeling in normal hematopoiesis and age-related hematological malignancies. Bone Res 2023; 11:15. [PMID: 36918531 PMCID: PMC10014945 DOI: 10.1038/s41413-023-00249-w] [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: 08/01/2022] [Revised: 12/24/2022] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Prior research establishing that bone interacts in coordination with the bone marrow microenvironment (BMME) to regulate hematopoietic homeostasis was largely based on analyses of individual bone-associated cell populations. Recent advances in intravital imaging has suggested that the expansion of hematopoietic stem cells (HSCs) and acute myeloid leukemia cells is restricted to bone marrow microdomains during a distinct stage of bone remodeling. These findings indicate that dynamic bone remodeling likely imposes additional heterogeneity within the BMME to yield differential clonal responses. A holistic understanding of the role of bone remodeling in regulating the stem cell niche and how these interactions are altered in age-related hematological malignancies will be critical to the development of novel interventions. To advance this understanding, herein, we provide a synopsis of the cellular and molecular constituents that participate in bone turnover and their known connections to the hematopoietic compartment. Specifically, we elaborate on the coupling between bone remodeling and the BMME in homeostasis and age-related hematological malignancies and after treatment with bone-targeting approaches. We then discuss unresolved questions and ambiguities that remain in the field.
Collapse
Affiliation(s)
- Hengwei Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Jane L Liesveld
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura M Calvi
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Endocrinology/Metabolism, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea C Lipe
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy/Immunology/Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Shu-Chi A Yeh
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
24
|
Marques-Mourlet C, Di Iorio R, Fairfield H, Reagan MR. Obesity and myeloma: Clinical and mechanistic contributions to disease progression. Front Endocrinol (Lausanne) 2023; 14:1118691. [PMID: 36909335 PMCID: PMC9996186 DOI: 10.3389/fendo.2023.1118691] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Obesity and obesogenic behaviors are positively associated with both monoclonal gammopathy of unknown significance (MGUS) and multiple myeloma (MM). As the only known modifiable risk factor, this association has emerged as a new potential target for MM prevention, but little is known about the mechanistic relationship of body weight with MM progression. Here we summarize epidemiological correlations between weight, body composition, and the various stages of myeloma disease progression and treatments, as well as the current understanding of the molecular contributions of obesity-induced changes in myeloma cell phenotype and signaling. Finally, we outline groundwork for the future characterization of the relationship between body weight patterns, the bone marrow microenvironment, and MM pathogenesis in animal models, which have the potential to impact our understanding of disease pathogenesis and inform MM prevention messages.
Collapse
Affiliation(s)
- Constance Marques-Mourlet
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Strasbourg, Pharmacology Department, Strasbourg, France
| | - Reagan Di Iorio
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of New England, College of Osteopathic Medicine, Biddeford, ME, United States
| | - Heather Fairfield
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University, School of Medicine, Boston, MA, United States
| | - Michaela R. Reagan
- MaineHealth Institute for Research, Center for Molecular Medicine, Scarborough, ME, United States
- University of Maine, Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University, School of Medicine, Boston, MA, United States
| |
Collapse
|
25
|
Fairfield H, Condruti R, Farrell M, Di Iorio R, Gartner CA, Vary C, Reagan MR. Development and characterization of three cell culture systems to investigate the relationship between primary bone marrow adipocytes and myeloma cells. Front Oncol 2023; 12:912834. [PMID: 36713534 PMCID: PMC9874147 DOI: 10.3389/fonc.2022.912834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 11/21/2022] [Indexed: 01/12/2023] Open
Abstract
The unique properties of the bone marrow (BM) allow for migration and proliferation of multiple myeloma (MM) cells while also providing the perfect environment for development of quiescent, drug-resistant MM cell clones. BM adipocytes (BMAds) have recently been identified as important contributors to systemic adipokine levels, bone strength, hematopoiesis, and progression of metastatic and primary BM cancers, such as MM. Recent studies in myeloma suggest that BMAds can be reprogrammed by tumor cells to contribute to myeloma-induced bone disease, and, reciprocally, BMAds support MM cells in vitro. Importantly, most data investigating BMAds have been generated using adipocytes generated by differentiating BM-derived mesenchymal stromal cells (BMSCs) into adipocytes in vitro using adipogenic media, due to the extreme technical challenges associated with isolating and culturing primary adipocytes. However, if studies could be performed with primary adipocytes, then they likely will recapitulate in vivo biology better than BMSC-derived adipocytes, as the differentiation process is artificial and differs from in vivo differentiation, and progenitor cell(s) of the primary BMAd (pBMAds) may not be the same as the BMSCs precursors used for adipogenic differentiation in vitro. Therefore, we developed and refined three methods for culturing pBMAds: two-dimensional (2D) coverslips, 2D transwells, and three-dimensional (3D) silk scaffolds, all of which can be cultured alone or with MM cells to investigate bidirectional tumor-host signaling. To develop an in vitro model with a tissue-like structure to mimic the BM microenvironment, we developed the first 3D, tissue engineered model utilizing pBMAds derived from human BM. We found that pBMAds, which are extremely fragile, can be isolated and stably cultured in 2D for 10 days and in 3D for up to 4 week in vitro. To investigate the relationship between pBMAds and myeloma, MM cells can be added to investigate physical relationships through confocal imaging and soluble signaling molecules via mass spectrometry. In summary, we developed three in vitro cell culture systems to study pBMAds and myeloma cells, which could be adapted to investigate many diseases and biological processes involving the BM, including other bone-homing tumor types.
Collapse
Affiliation(s)
- Heather Fairfield
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | | | - Mariah Farrell
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | - Reagan Di Iorio
- MaineHealth Institute for Research, Scarborough, ME, United States,University of New England, Biddeford, ME, United States
| | - Carlos A. Gartner
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | - Calvin Vary
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States
| | - Michaela R. Reagan
- MaineHealth Institute for Research, Scarborough, ME, United States,University of Maine Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Tufts University School of Medicine, Boston, MA, United States,*Correspondence: Michaela R. Reagan,
| |
Collapse
|
26
|
Bertuglia G, Cani L, Larocca A, Gay F, D'Agostino M. Normalization of the Immunological Microenvironment and Sustained Minimal Residual Disease Negativity: Do We Need Both for Long-Term Control of Multiple Myeloma? Int J Mol Sci 2022; 23:15879. [PMID: 36555520 PMCID: PMC9781462 DOI: 10.3390/ijms232415879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Over the past two decades, the treatment landscape for multiple myeloma (MM) has progressed significantly, with the introduction of several new drug classes that have greatly improved patient outcomes. At present, it is well known how the bone marrow (BM) microenvironment (ME) exerts an immunosuppressive action leading to an exhaustion of the immune system cells and promoting the proliferation and sustenance of tumor plasma cells. Therefore, having drugs that can reconstitute a healthy BM ME can improve results in MM patients. Recent findings clearly demonstrated that achieving minimal residual disease (MRD) negativity and sustaining MRD negativity over time play a pivotal prognostic role. However, despite the achievement of MRD negativity, patients may still relapse. The understanding of immunologic changes in the BM ME during treatment, complemented by a deeper knowledge of plasma cell genomics and biology, will be critical to develop future therapies to sustain MRD negativity over time and possibly achieve an operational cure. In this review, we focus on the components of the BM ME and their role in MM, on the prognostic significance of MRD negativity and, finally, on the relative contribution of tumor plasma cell biology and BM ME to long-term disease control.
Collapse
Affiliation(s)
- Giuseppe Bertuglia
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Lorenzo Cani
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Alessandra Larocca
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Francesca Gay
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| | - Mattia D'Agostino
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| |
Collapse
|
27
|
Lourenço D, Lopes R, Pestana C, Queirós AC, João C, Carneiro EA. Patient-Derived Multiple Myeloma 3D Models for Personalized Medicine-Are We There Yet? Int J Mol Sci 2022; 23:12888. [PMID: 36361677 PMCID: PMC9657251 DOI: 10.3390/ijms232112888] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 12/03/2023] Open
Abstract
Despite the wide variety of existing therapies, multiple myeloma (MM) remains a disease with dismal prognosis. Choosing the right treatment for each patient remains one of the major challenges. A new approach being explored is the use of ex vivo models for personalized medicine. Two-dimensional culture or animal models often fail to predict clinical outcomes. Three-dimensional ex vivo models using patients' bone marrow (BM) cells may better reproduce the complexity and heterogeneity of the BM microenvironment. Here, we review the strengths and limitations of currently existing patient-derived ex vivo three-dimensional MM models. We analyze their biochemical and biophysical properties, molecular and cellular characteristics, as well as their potential for drug testing and identification of disease biomarkers. Furthermore, we discuss the remaining challenges and give some insight on how to achieve a more biomimetic and accurate MM BM model. Overall, there is still a need for standardized culture methods and refined readout techniques. Including both myeloma and other cells of the BM microenvironment in a simple and reproducible three-dimensional scaffold is the key to faithfully mapping and examining the relationship between these players in MM. This will allow a patient-personalized profile, providing a powerful tool for clinical and research applications.
Collapse
Affiliation(s)
- Diana Lourenço
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Lopes
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Carolina Pestana
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Centre of Statistics and Its Applications, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Ana C. Queirós
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Cristina João
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Myeloma Lymphoma Research Group—Champalimaud Experimental Clinical Research Programme of Champalimaud Foundation, 1400-038 Lisbon, Portugal
| |
Collapse
|
28
|
Roman-Trufero M, Auner HW, Edwards CM. Multiple myeloma metabolism - a treasure trove of therapeutic targets? Front Immunol 2022; 13:897862. [PMID: 36072593 PMCID: PMC9441940 DOI: 10.3389/fimmu.2022.897862] [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: 03/16/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma is an incurable cancer of plasma cells that is predominantly located in the bone marrow. Multiple myeloma cells are characterized by distinctive biological features that are intricately linked to their core function, the assembly and secretion of large amounts of antibodies, and their diverse interactions with the bone marrow microenvironment. Here, we provide a concise and introductory discussion of major metabolic hallmarks of plasma cells and myeloma cells, their roles in myeloma development and progression, and how they could be exploited for therapeutic purposes. We review the role of glucose consumption and catabolism, assess the dependency on glutamine to support key metabolic processes, and consider metabolic adaptations in drug-resistant myeloma cells. Finally, we examine the complex metabolic effects of proteasome inhibitors on myeloma cells and the extracellular matrix, and we explore the complex relationship between myeloma cells and bone marrow adipocytes.
Collapse
Affiliation(s)
- Monica Roman-Trufero
- Department of Immunology and Inflammation, Cancer Cell Protein Metabolism, The Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Imperial College London, London, United Kingdom
| | - Holger W. Auner
- Department of Immunology and Inflammation, Cancer Cell Protein Metabolism, The Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Imperial College London, London, United Kingdom
| | - Claire M. Edwards
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
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
|
30
|
Venglar O, Bago JR, Motais B, Hajek R, Jelinek T. Natural Killer Cells in the Malignant Niche of Multiple Myeloma. Front Immunol 2022; 12:816499. [PMID: 35087536 PMCID: PMC8787055 DOI: 10.3389/fimmu.2021.816499] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells represent a subset of CD3- CD7+ CD56+/dim lymphocytes with cytotoxic and suppressor activity against virus-infected cells and cancer cells. The overall potential of NK cells has brought them to the spotlight of targeted immunotherapy in solid and hematological malignancies, including multiple myeloma (MM). Nonetheless, NK cells are subjected to a variety of cancer defense mechanisms, leading to impaired maturation, chemotaxis, target recognition, and killing. This review aims to summarize the available and most current knowledge about cancer-related impairment of NK cell function occurring in MM.
Collapse
Affiliation(s)
- Ondrej Venglar
- Faculty of Science, University of Ostrava, Ostrava, Czechia.,Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Julio Rodriguez Bago
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Benjamin Motais
- Faculty of Science, University of Ostrava, Ostrava, Czechia.,Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Roman Hajek
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Tomas Jelinek
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| |
Collapse
|
31
|
Panaroni C, Fulzele K, Mori T, Siu KT, Onyewadume C, Maebius A, Raje N. Multiple myeloma cells induce lipolysis in adipocytes and uptake fatty acids through fatty acid transporter proteins. Blood 2022; 139:876-888. [PMID: 34662370 PMCID: PMC8832479 DOI: 10.1182/blood.2021013832] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
Adipocytes occupy 70% of the cellular volume within the bone marrow (BM) wherein multiple myeloma (MM) originates and resides. However, the nature of the interaction between MM cells and adipocytes remains unclear. Cancer-associated adipocytes support tumor cells through various mechanisms, including metabolic reprogramming of cancer cells. We hypothesized that metabolic interactions mediate the dependence of MM cells on BM adipocytes. Here we show that BM aspirates from precursor states of MM, including monoclonal gammopathy of undetermined significance and smoldering MM, exhibit significant upregulation of adipogenic commitment compared with healthy donors. In vitro coculture assays revealed an adipocyte-induced increase in MM cell proliferation in monoclonal gammopathy of undetermined significance/smoldering MM compared with newly diagnosed MM. Using murine MM cell/BM adipocyte coculture assays, we describe MM-induced lipolysis in adipocytes via activation of the lipolysis pathway. Upregulation of fatty acid transporters 1 and 4 on MM cells mediated the uptake of secreted free fatty acids (FFAs) by adjacent MM cells. The effect of FFAs on MM cells was dose dependent and revealed increased proliferation at lower concentrations vs induction of lipotoxicity at higher concentrations. Lipotoxicity occurred via the ferroptosis pathway. Exogenous treatment with arachidonic acid, a very-long-chain FFA, in a murine plasmacytoma model displayed a reduction in tumor burden. Taken together, our data reveal a novel pathway involving MM cell-induced lipolysis in BM adipocytes and suggest prevention of FFA uptake by MM cells as a potential target for myeloma therapeutics.
Collapse
Affiliation(s)
- Cristina Panaroni
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
| | - Keertik Fulzele
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
| | - Tomoaki Mori
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
| | - Ka Tat Siu
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
- Beam Therapeutics, Cambridge, MA
| | - Chukwuamaka Onyewadume
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
| | - Allison Maebius
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
| | - Noopur Raje
- Center for Multiple Myeloma, Division of Hematology and Oncology, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA; and
| |
Collapse
|
32
|
Nicol JL, Woodrow C, Cunningham BJ, Mollee P, Weber N, Smith MD, Nicol AJ, Gordon LG, Hill MM, Skinner TL. An Individualized Exercise Intervention for People with Multiple Myeloma—Study Protocol of a Randomized Waitlist-Controlled Trial. Curr Oncol 2022; 29:901-923. [PMID: 35200576 PMCID: PMC8870457 DOI: 10.3390/curroncol29020077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
People with multiple myeloma (MM) are second only to people with lung cancer for the poorest reported health-related quality of life (HRQoL) of all cancer types. Whether exercise can improve HRQoL in MM, where bone pain and lesions are common, requires investigation. This trial aims to evaluate the efficacy of an exercise intervention compared with control on HRQoL in people with MM. Following baseline testing, people with MM (n = 60) will be randomized to an exercise (EX) or waitlist control (WT) group. EX will complete 12-weeks of supervised (24 sessions) and unsupervised (12 sessions) individualized, modular multimodal exercise training. From weeks 12–52, EX continue unsupervised training thrice weekly, with one optional supervised group-based session weekly from weeks 12–24. The WT will be asked to maintain their current activity levels for the first 12-weeks, before completing the same protocol as EX for the following 52 weeks. Primary (patient-reported HRQoL) and secondary (bone health and pain, fatigue, cardiorespiratory fitness, muscle strength, body composition, disease response, and blood biomarkers) outcomes will be assessed at baseline, 12-, 24- and 52-weeks. Adverse events, attendance, and adherence will be recorded and cost-effectiveness analysis performed. The findings will inform whether exercise should be included as part of standard myeloma care to improve the health of this unique population.
Collapse
Affiliation(s)
- Jennifer L. Nicol
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia; (L.G.G.); (M.M.H.)
- Correspondence:
| | - Carmel Woodrow
- Haematology, Division of Cancer, Princess Alexandra Hospital, Brisbane 4102, Australia; (C.W.); (P.M.)
| | - Brent J. Cunningham
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
| | - Peter Mollee
- Haematology, Division of Cancer, Princess Alexandra Hospital, Brisbane 4102, Australia; (C.W.); (P.M.)
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Nicholas Weber
- Haematology, Cancer Care Services, Royal Brisbane and Women’s Hospital, Brisbane 4006, Australia;
| | - Michelle D. Smith
- School of Health and Rehabilitation Sciences, The University of Queensland, Brisbane 4072, Australia;
| | - Andrew J. Nicol
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
- Brisbane Clinic for Lymphoma, Myeloma and Leukaemia, Greenslopes Private Hospital, Brisbane 4120, Australia
| | - Louisa G. Gordon
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia; (L.G.G.); (M.M.H.)
- Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
- Palliative Care Outcomes Centre, School of Nursing and Cancer, Queensland University of Technology, Brisbane 4059, Australia
| | - Michelle M. Hill
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia; (L.G.G.); (M.M.H.)
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane 4006, Australia
| | - Tina L. Skinner
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Australia; (B.J.C.); (A.J.N.); (T.L.S.)
| |
Collapse
|
33
|
Da Ros F, Persano L, Bizzotto D, Michieli M, Braghetta P, Mazzucato M, Bonaldo P. Emilin-2 is a component of bone marrow extracellular matrix regulating mesenchymal stem cell differentiation and hematopoietic progenitors. Stem Cell Res Ther 2022; 13:2. [PMID: 35012633 PMCID: PMC8744352 DOI: 10.1186/s13287-021-02674-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Dissection of mechanisms involved in the regulation of bone marrow microenvironment through cell–cell and cell–matrix contacts is essential for the detailed understanding of processes underlying bone marrow activities both under physiological conditions and in hematologic malignancies. Here we describe Emilin-2 as an abundant extracellular matrix component of bone marrow stroma. Methods Immunodetection of Emilin-2 was performed in bone marrow sections of mice from 30 days to 6 months of age. Emilin-2 expression was monitored in vitro in primary and mesenchymal stem cell lines under undifferentiated and adipogenic conditions. Hematopoietic stem cells and progenitors in bone marrow of 3- to 10-month-old wild-type and Emilin-2 null mice were analyzed by flow cytometry. Results Emilin-2 is deposited in bone marrow extracellular matrix in an age-dependent manner, forming a meshwork that extends from compact bone boundaries to the central trabecular regions. Emilin-2 is expressed and secreted by both primary and immortalized bone marrow mesenchymal stem cells, exerting an inhibitory action in adipogenic differentiation. In vivo Emilin-2 deficiency impairs the frequency of hematopoietic stem/progenitor cells in bone marrow during aging. Conclusion Our data provide new insights in the contribution of bone marrow extracellular matrix microenvironment in the regulation of stem cell niches and hematopoietic progenitor differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02674-2.
Collapse
Affiliation(s)
- Francesco Da Ros
- SOSd Cell Stem Unit, Department of Translational Research, National Cancer Center CRO-IRCSS, 33081, Aviano, Italy.,Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Luca Persano
- Department of Women's and Children's Health, University of Padova, 35131, Padova, Italy.,IRP - Pediatric Research Institute, 35131, Padova, Italy
| | - Dario Bizzotto
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Mariagrazia Michieli
- SOSd Cell Therapy and High Dose Chemotherapy, National Cancer Center CRO- IRCCS, 33081, Aviano, Italy
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Mario Mazzucato
- SOSd Cell Stem Unit, Department of Translational Research, National Cancer Center CRO-IRCSS, 33081, Aviano, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131, Padova, Italy. .,CRIBI Biotechnology Center, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
34
|
Wang Z, He J, Bach DH, Huang YH, Li Z, Liu H, Lin P, Yang J. Induction of m 6A methylation in adipocyte exosomal LncRNAs mediates myeloma drug resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:4. [PMID: 34980213 PMCID: PMC8722039 DOI: 10.1186/s13046-021-02209-w] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/04/2021] [Indexed: 11/10/2022]
Abstract
Background Therapeutic resistance occurs in most patients with multiple myeloma (MM). One of the key mechanisms for MM drug resistance comes from the interaction between MM cells and adipocytes that inhibits drug-induced apoptosis in MM cells; MM cells reprogram adipocytes to morph into different characterizations, including exosomes, which are important for tumor-stroma cellular communication. However, the mechanism by which exosomes mediate the cellular machinery of the vicious cycle between MM cells and adipocytes remains unclear. Methods Adipocytes were either isolated from bone marrow aspirates of healthy donors or MM patients or derived from mesenchymal stem cells. Co-culturing normal adipocytes with MM cells was used to generate MM-associated adipocytes. Exosomes were collected from the culture medium of adipocytes. Annexin V-binding and TUNEL assays were performed to assess MM cell apoptosis. Methyltransferase activity assay and dot blotting were used to access the m6A methylation activity of methyltransferase like 7A (METTL7A). RIP, MeRIP-seq, and RNA–protein pull down for assessing the interaction between long non-cording RNAs (LncRNAs) and RNA binding proteins were performed. Adipocyte-specific enhancer of zeste homolog 2 (EZH2) knockout mice and MM-xenografted mice were used for evaluating MM therapeutic response in vivo. Results Exosomes collected from MM patient adipocytes protect MM cells from chemotherapy-induced apoptosis. Two LncRNAs in particular, LOC606724 and SNHG1, are significantly upregulated in MM cells after exposure to adipocyte exosomes. The raised LncRNA levels in MM cells are positively correlated to worse outcomes in patients, indicating their clinical relevancy in MM. The functional roles of adipocyte exosomal LOC606724 or SNHG1 in inhibition of MM cell apoptosis are determined by knockdown in adipocytes or overexpression in MM cells. We discovered the interactions between LncRNAs and RNA binding proteins and identified methyltransferase like 7A (METTL7A) as an RNA methyltransferase. MM cells promote LncRNA package into adipocyte exosomes through METTL7A-mediated LncRNA m6A methylation. Exposure of adipocytes to MM cells enhances METTL7A activity in m6A methylation through EZH2-mediated protein methylation. Conclusion This study elucidates an unexplored mechanism of how adipocyte-rich microenvironment exacerbates MM therapeutic resistance and indicates a potential strategy to improve therapeutic efficacy by blocking this vicious exosome-mediated cycle. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02209-w.
Collapse
Affiliation(s)
- Zhiming Wang
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Jin He
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Duc-Hiep Bach
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Yung-Hsing Huang
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Zongwei Li
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Huan Liu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jing Yang
- Houston Methodist Cancer Center, Research Institute Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Dello Spedale Venti M, Palmisano B, Donsante S, Farinacci G, Adotti F, Coletta I, Serafini M, Corsi A, Riminucci M. Morphological and Immunophenotypical Changes of Human Bone Marrow Adipocytes in Marrow Metastasis and Myelofibrosis. Front Endocrinol (Lausanne) 2022; 13:882379. [PMID: 35757418 PMCID: PMC9215173 DOI: 10.3389/fendo.2022.882379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022] Open
Abstract
The bone marrow adipose tissue constitutes more than two-thirds of the bone marrow volume in adult life and is known to have unique metabolic and functional properties. In neoplastic disorders, bone marrow adipocytes (BMAds) contribute to create a favorable microenvironment to survival and proliferation of cancer cells. Many studies explored the molecular crosstalk between BMAds and neoplastic cells, predominantly in ex-vivo experimental systems or in animal models. However, little is known on the features of BMAds in the human neoplastic marrow. The aim of our study was to analyze the in situ changes in morphology and immunophenotype of BMAds in two different types of neoplastic marrow conditions. We selected a series of archival iliac crest and vertebral bone biopsies from patients with bone marrow metastasis (MET), patients with myeloproliferative neoplasia with grade-3 myelofibrosis (MPN-MF) and age-matched controls (CTR). We observed a significant reduction in the number of BMAds in MET and MPN-MF compared to CTR. Accordingly, in the same groups, we also detected a significant reduction in the mean cell diameter and area. Immunolocalization of different adipocyte markers showed that, compared to CTR, in both MET and MPN-MF the percentages of adiponectin- and phosphorylated hormone sensitive lipase-positive BMAds were significantly reduced and increased respectively. No statistically significant difference was found between MET and MPN-MF. Interestingly, in one MET sample, "remodeled" BMAds containing a large lipid vacuole and multiple, smaller and polarized lipid droplets were identified. In conclusion, our data show that in different types of marrow cancers, BMAds undergo significant quantitative and qualitative changes, which need to be further investigated in future studies.
Collapse
Affiliation(s)
| | - Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giorgia Farinacci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Flavia Adotti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilenia Coletta
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- *Correspondence: Mara Riminucci,
| |
Collapse
|
36
|
Khalife J, Sanchez JF, Pichiorri F. The Emerging Role of Extracellular Vesicle-Associated RNAs in the Multiple Myeloma Microenvironment. Front Oncol 2021; 11:689538. [PMID: 34235082 PMCID: PMC8255802 DOI: 10.3389/fonc.2021.689538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a cancer of terminally differentiated plasma cells (PCs) that develop at multiple sites within the bone marrow (BM). MM is treatable but rarely curable because of the frequent emergence of drug resistance and relapse. Increasing evidence indicates that the BM microenvironment plays a major role in supporting MM-PC survival and resistance to therapy. The BM microenvironment is a complex milieu containing hematopoietic cells, stromal cells, endothelial cells, immune cells, osteoclasts and osteoblasts, all contributing to the pathobiology of MM, including PC proliferation, escape from immune surveillance, angiogenesis and bone disease development. Small extracellular vesicles (EVs) are heterogenous lipid structures released by all cell types and mediate local and distal cellular communication. In MM, EVs are key mediators of the cross-talk between PCs and the surrounding microenvironment because of their ability to deliver bioactive cargo molecules such as lipids, mRNAs, non-coding regulatory RNA and proteins. Hence, MM-EVs highly contribute to establish a tumor-supportive BM niche that impacts MM pathogenesis and disease progression. In this review, we will first highlight the effects of RNA-containing, MM-derived EVs on the several cellular compartments within the BM microenvironment that play a role in the different aspects of MM pathology. We will also touch on the prospective use of MM-EV-associated non-coding RNAs as clinical biomarkers in the context of “liquid biopsy” in light of their importance as a promising tool in MM diagnosis, prognosis and prediction of drug resistance.
Collapse
Affiliation(s)
- Jihane Khalife
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA, United States.,Department of Hematologic Malignancies Translational Science, City of Hope, Duarte, CA, United States
| | - James F Sanchez
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA, United States
| | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA, United States.,Department of Hematologic Malignancies Translational Science, City of Hope, Duarte, CA, United States
| |
Collapse
|
37
|
Maiso P, Mogollón P, Ocio EM, Garayoa M. Bone Marrow Mesenchymal Stromal Cells in Multiple Myeloma: Their Role as Active Contributors to Myeloma Progression. Cancers (Basel) 2021; 13:2542. [PMID: 34067236 PMCID: PMC8196907 DOI: 10.3390/cancers13112542] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Patricia Maiso
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| |
Collapse
|
38
|
Targeting Reactive Oxygen Species Metabolism to Induce Myeloma Cell Death. Cancers (Basel) 2021; 13:cancers13102411. [PMID: 34067602 PMCID: PMC8156203 DOI: 10.3390/cancers13102411] [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: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a common hematological disease characterized by the accumulation of clonal malignant plasma cells in the bone marrow. Over the past two decades, new therapeutic strategies have significantly improved the treatment outcome and patients survival. Nevertheless, most MM patients relapse underlying the need of new therapeutic approaches. Plasma cells are prone to produce large amounts of immunoglobulins causing the production of intracellular ROS. Although adapted to high level of ROS, MM cells die when exposed to drugs increasing ROS production either directly or by inhibiting antioxidant enzymes. In this review, we discuss the efficacy of ROS-generating drugs for inducing MM cell death and counteracting acquired drug resistance specifically toward proteasome inhibitors.
Collapse
|
39
|
Lazaris V, Hatziri A, Symeonidis A, Kypreos KE. The Lipoprotein Transport System in the Pathogenesis of Multiple Myeloma: Advances and Challenges. Front Oncol 2021; 11:638288. [PMID: 33842343 PMCID: PMC8032975 DOI: 10.3389/fonc.2021.638288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/10/2021] [Indexed: 01/02/2023] Open
Abstract
Multiple myeloma (MM) is an incurable neoplastic hematologic disorder characterized by malignant plasma cells, mainly in the bone marrow. MM is associated with multiple factors, such as lipid metabolism, obesity, and age-associated disease development. Although, the precise pathogenetic mechanisms remain unknown, abnormal lipid and lipoprotein levels have been reported in patients with MM. Interestingly, patients with higher APOA1 levels, the major apolipoprotein of high density lipoprotein (HDL), have better overall survival. The limited existing studies regarding serum lipoproteins in MM are inconclusive, and often contradictory. Nevertheless, it appears that deregulation of the lipoprotein transport system may facilitate the development of the disease. Here, we provide a critical review of the literature on the role of lipids and lipoproteins in MM pathophysiology. We also propose novel mechanisms, linking the development and progression of MM to the metabolism of blood lipoproteins. We anticipate that proteomic and lipidomic analyses of serum lipoproteins along with analyses of their functionality may improve our understanding and shed light on novel mechanistic aspects of MM pathophysiology.
Collapse
Affiliation(s)
- Vasileios Lazaris
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece.,Hematology Clinic, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece
| | - Aikaterini Hatziri
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece
| | - Argiris Symeonidis
- Hematology Clinic, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece
| | - Kyriakos E Kypreos
- Pharmacology Laboratory, Department of Medicine, School of Health Sciences, University of Patras, Patras, Greece.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
40
|
Fairfield H, Costa S, Falank C, Farrell M, Murphy CS, D’Amico A, Driscoll H, Reagan MR. Multiple Myeloma Cells Alter Adipogenesis, Increase Senescence-Related and Inflammatory Gene Transcript Expression, and Alter Metabolism in Preadipocytes. Front Oncol 2021; 10:584683. [PMID: 33680918 PMCID: PMC7930573 DOI: 10.3389/fonc.2020.584683] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/23/2020] [Indexed: 12/27/2022] Open
Abstract
Within the bone marrow microenvironment, mesenchymal stromal cells (MSCs) are an essential precursor to bone marrow adipocytes and osteoblasts. The balance between this progenitor pool and mature cells (adipocytes and osteoblasts) is often skewed by disease and aging. In multiple myeloma (MM), a cancer of the plasma cell that predominantly grows within the bone marrow, as well as other cancers, MSCs, preadipocytes, and adipocytes have been shown to directly support tumor cell survival and proliferation. Increasing evidence supports the idea that MM-associated MSCs are distinct from healthy MSCs, and their gene expression profiles may be predictive of myeloma patient outcomes. Here we directly investigate how MM cells affect the differentiation capacity and gene expression profiles of preadipocytes and bone marrow MSCs. Our studies reveal that MM.1S cells cause a marked decrease in lipid accumulation in differentiating 3T3-L1 cells. Also, MM.1S cells or MM.1S-conditioned media altered gene expression profiles of both 3T3-L1 and mouse bone marrow MSCs. 3T3-L1 cells exposed to MM.1S cells before adipogenic differentiation displayed gene expression changes leading to significantly altered pathways involved in steroid biosynthesis, the cell cycle, and metabolism (oxidative phosphorylation and glycolysis) after adipogenesis. MM.1S cells induced a marked increase in 3T3-L1 expression of MM-supportive genes including Il-6 and Cxcl12 (SDF1), which was confirmed in mouse MSCs by qRT-PCR, suggesting a forward-feedback mechanism. In vitro experiments revealed that indirect MM exposure prior to differentiation drives a senescent-like phenotype in differentiating MSCs, and this trend was confirmed in MM-associated MSCs compared to MSCs from normal donors. In direct co-culture, human mesenchymal stem cells (hMSCs) exposed to MM.1S, RPMI-8226, and OPM-2 prior to and during differentiation, exhibited different levels of lipid accumulation as well as secreted cytokines. Combined, our results suggest that MM cells can inhibit adipogenic differentiation while stimulating expression of the senescence associated secretory phenotype (SASP) and other pro-myeloma molecules. This study provides insight into a novel way in which MM cells manipulate their microenvironment by altering the expression of supportive cytokines and skewing the cellular diversity of the marrow.
Collapse
Affiliation(s)
- Heather Fairfield
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Carolyne Falank
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Mariah Farrell
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Biology Department, University of Southern Maine, Portland, ME, United States
| | - Connor S. Murphy
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Anastasia D’Amico
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Biology Department, University of Southern Maine, Portland, ME, United States
| | - Heather Driscoll
- Biology Department, Norwich University, Northfield, VT, United States
| | - Michaela R. Reagan
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, United States,School of Medicine, Tufts University, Boston, MA, United States,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States,Biology Department, University of Southern Maine, Portland, ME, United States,*Correspondence: Michaela R. Reagan,
| |
Collapse
|
41
|
Georgakopoulou R, Andrikopoulou A, Sergentanis TN, Fiste O, Zagouri F, Gavriatopoulou M, Psaltopoulou T, Kastritis E, Terpos E, Dimopoulos MA. Overweight/Obesity and Monoclonal Gammopathy of Undetermined Significance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:361-367. [PMID: 33582053 DOI: 10.1016/j.clml.2021.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Obesity and high body mass index (BMI) are associated with increased incidence of multiple myeloma (MM). MM usually evolves from a precursor asymptomatic disease, namely monoclonal gammopathy of undetermined significance (MGUS). MGUS progresses to MM at a 1% annual rate; however, risk factors predisposing to MGUS are not completely understood. We conducted a systematic review to assess the relationship between obesity and high BMI with MGUS prevalence and progression to MM. To our knowledge, this is the first systematic review evaluating the role of obesity in MGUS. PATIENTS AND METHODS We searched the Medline database and ClinicalTrials.gov for studies investigating BMI and obesity association with MGUS incidence and progression. The algorithm consisted of a predefined combination of the words "obesity," "obese," "body mass index," "overweight," "diet," "nutrition," "food," "dietary," "adiponectin," "monoclonal gammopathy," and "MGUS". RESULTS Overall, 12 articles were retrieved, including 11 eligible articles and 1 clinical trial. More than 57,068 patients were evaluated in this systematic review. Discrepancies between the identified studies were noted. Multiple studies support the notion that obesity or high BMI are positively linked to MGUS prevalence and transition to MM. In contrast, other studies revealed no such association. Visceral adipose tissue metabolic activity and decreased adiponectin concentrations were identified as biomarkers of MGUS progression to MM. CONCLUSION Obesity and increased BMI seem to be implicated both in MGUS development and progression to MM. Further studies should be designed to confirm this hypothesis.
Collapse
Affiliation(s)
- Rebecca Georgakopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Plasma Cell Dyscrasias Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Angeliki Andrikopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros N Sergentanis
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Oraianthi Fiste
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Plasma Cell Dyscrasias Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodora Psaltopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Plasma Cell Dyscrasias Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Plasma Cell Dyscrasias Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Plasma Cell Dyscrasias Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Plasma Cell Dyscrasias Unit, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
42
|
Reagan MR, Fairfield H, Rosen CJ. Bone Marrow Adipocytes: A Link between Obesity and Bone Cancer. Cancers (Basel) 2021; 13:364. [PMID: 33498240 PMCID: PMC7863952 DOI: 10.3390/cancers13030364] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/24/2020] [Accepted: 01/15/2021] [Indexed: 12/30/2022] Open
Abstract
Cancers that grow in the bone marrow are for most patients scary, painful, and incurable. These cancers are especially hard to treat due to the supportive microenvironment provided by the bone marrow niche in which they reside. New therapies designed to target tumor cells have extended the life expectancy for these patients, but better therapies are needed and new ideas for how to target these cancers are crucial. This need has led researchers to interrogate whether bone marrow adipocytes (BMAds), which increase in number and size during aging and in obesity, contribute to cancer initiation or progression within the bone marrow. Across the globe, the consensus in the field is a unified "yes". However, how to target these adipocytes or the factors they produce and how BMAds interact with different tumor cells are open research questions. Herein, we review this research field, with the goal of accelerating research in the network of laboratories working in this area and attracting bright scientists with new perspectives and ideas to the field in order to bring about better therapies for patients with bone cancers.
Collapse
Affiliation(s)
- Michaela R. Reagan
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, ME 04074, USA; (H.F.); (C.J.R.)
- School of Medicine, Tufts University, Boston, MA 02111, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - Heather Fairfield
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, ME 04074, USA; (H.F.); (C.J.R.)
- School of Medicine, Tufts University, Boston, MA 02111, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - Clifford J. Rosen
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, ME 04074, USA; (H.F.); (C.J.R.)
- School of Medicine, Tufts University, Boston, MA 02111, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| |
Collapse
|
43
|
Zeissig MN, Zannettino ACW, Vandyke K. Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma. Cancers (Basel) 2020; 12:cancers12123643. [PMID: 33291672 PMCID: PMC7761917 DOI: 10.3390/cancers12123643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Like in solid cancers, the process of dissemination is a critical feature of disease progression in the blood cancer multiple myeloma. At diagnosis, myeloma patients have cancer that has spread throughout the bone marrow, with patients with more disseminatory myeloma having worse outcomes for their disease. In this review, we discuss the current understanding of the mechanisms that underpin the dissemination process in multiple myeloma. Furthermore, we discuss the potential for the use of therapies that target the dissemination process as a novel means of improving outcomes for multiple myeloma patients. Abstract Multiple myeloma (MM) is a plasma cell (PC) malignancy characterised by the presence of MM PCs at multiple sites throughout the bone marrow. Increased numbers of peripheral blood MM PCs are associated with rapid disease progression, shorter time to relapse and are a feature of advanced disease. In this review, the current understanding of the process of MM PC dissemination and the extrinsic and intrinsic factors potentially driving it are addressed through analysis of patient-derived MM PCs and MM cell lines as well as mouse models of homing and dissemination. In addition, we discuss how patient cytogenetic subgroups that present with highly disseminated disease, such as t(4;14), t(14;16) and t(14;20), suggest that intrinsic properties of MM PC influence their ability to disseminate. Finally, we discuss the possibility of using therapeutic targeting of tumour dissemination to slow disease progression and prevent overt relapse.
Collapse
Affiliation(s)
- Mara N. Zeissig
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| | - Andrew C. W. Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- Central Adelaide Local Health Network, Adelaide 5000, Australia
- Centre for Cancer Biology, University of South Australia, Adelaide 5000, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- Correspondence: ; Tel.: +61-8-8128-4694
| |
Collapse
|
44
|
Fairfield H, Dudakovic A, Khatib CM, Farrell M, Costa S, Falank C, Hinge M, Murphy CS, DeMambro V, Pettitt JA, Lary CW, Driscoll HE, McDonald MM, Kassem M, Rosen C, Andersen TL, van Wijnen AJ, Jafari A, Reagan MR. Myeloma-Modified Adipocytes Exhibit Metabolic Dysfunction and a Senescence-Associated Secretory Phenotype. Cancer Res 2020; 81:634-647. [PMID: 33218968 PMCID: PMC7854508 DOI: 10.1158/0008-5472.can-20-1088] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/05/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
Bone marrow adipocytes (BMAd) have recently been implicated in accelerating bone metastatic cancers, such as acute myelogenous leukemia and breast cancer. Importantly, bone marrow adipose tissue (BMAT) expands with aging and obesity, two key risk factors in multiple myeloma disease prevalence, suggesting that BMAds may influence and be influenced by myeloma cells in the marrow. Here, we provide evidence that reciprocal interactions and cross-regulation of myeloma cells and BMAds play a role in multiple myeloma pathogenesis and treatment response. Bone marrow biopsies from patients with multiple myeloma revealed significant loss of BMAT with myeloma cell infiltration of the marrow, whereas BMAT was restored after treatment for multiple myeloma. Myeloma cells reduced BMAT in different preclinical murine models of multiple myeloma and in vitro using myeloma cell-adipocyte cocultures. In addition, multiple myeloma cells altered adipocyte gene expression and cytokine secretory profiles, which were also associated with bioenergetic changes and induction of a senescent-like phenotype. In vivo, senescence markers were also increased in the bone marrow of tumor-burdened mice. BMAds, in turn, provided resistance to dexamethasone-induced cell-cycle arrest and apoptosis, illuminating a new possible driver of myeloma cell evolution in a drug-resistant clone. Our findings reveal that bidirectional interactions between BMAds and myeloma cells have significant implications for the pathogenesis and treatment of multiple myeloma. Targeting senescence in the BMAd or other bone marrow cells may represent a novel therapeutic approach for treatment of multiple myeloma. SIGNIFICANCE: This study changes the foundational understanding of how cancer cells hijack the bone marrow microenvironment and demonstrates that tumor cells induce senescence and metabolic changes in adipocytes, potentially driving new therapeutic directions.
Collapse
Affiliation(s)
- Heather Fairfield
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Amel Dudakovic
- Departments of Orthopedic Surgery and Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Casper M Khatib
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Mariah Farrell
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Samantha Costa
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Maja Hinge
- Division of Haematology, Department of Internal Medicine, Vejle Hospital, Vejle, Denmark
| | - Connor S Murphy
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Victoria DeMambro
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Jessica A Pettitt
- The Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | | | - Michelle M McDonald
- The Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.,Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Clifford Rosen
- Maine Medical Center Research Institute, Scarborough, Maine.,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| | - Thomas L Andersen
- Clinical Cell Biology, Department of Regional Health Research, Vejle/Lillebaelt Hospital, University of Southern Denmark, Vejle, Denmark.,Clinical Cell Biology, Department of Pathology, Odense University Hospital - Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery and Biochemistry & Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.
| | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, Maine. .,Tufts University School of Medicine, Boston, Massachusetts.,University of Maine Graduate School of Biomedical Science and Engineering, Orono, Maine
| |
Collapse
|
45
|
Luo G, Tang M, Zhao Q, Lu L, Xie Y, Li Y, Liu C, Tian L, Chen X, Yu X. Bone marrow adipocytes enhance osteolytic bone destruction by activating 1q21.3(S100A7/8/9-IL6R)-TLR4 pathway in lung cancer. J Cancer Res Clin Oncol 2020; 146:2241-2253. [PMID: 32494918 DOI: 10.1007/s00432-020-03277-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/29/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Bone metastasis is the result of complex crosstalk between tumor cells and bone marrow cells. Bone marrow adipocytes (BMAs) are the most abundant cell type in adult bone marrow. Therefore, we explore the effects of BMAs on bone metastasis in lung cancer. METHODS RNA-seq was used to compare the mRNA expression level of bone metastatic SBC5 cells and non-bone metastatic SBC3 cells. Rosiglitazone-induced marrow adiposity and intra-femoral injection of SBC5 cells were used to demonstrate the relationship between BMAs and SBC5 cells in vivo. Co-culture system, gene co-expression, gene ontology (GO) enrichment analysis and protein-protein interaction (PPI) network were used to explore the potential mechanism. RESULTS BMAs specially enhance the invasion of bone metastatic SBC5 instead of non-bone metastatic SBC3 in vitro. SBC5 instead of SBC3 promoted osteoblast and osteoclast differentiation as well as de-differentiation of mature BMAs. Rosiglitazone-induced marrow adiposity significantly enhanced osteolytic lesion induced by SBC5 in vivo. RNA-seq revealed that compared with SBC3, S100A9 and S100A8 genes were the most prominent genes up-regulated in SBC5 cells. High expression of S100A8/9 in SBC5 could be responsible for the crosstalk between lung cancer cells and BMAs. More importantly, interleukin 6 receptor (IL6R), which is adjacent to S100A8/A9 in 1q21.3, was significantly up-regulated by BMAs in vitro. S100A8/A9 (1 μg/ml) could obviously enhance the osteoblastic differentiation and inhibit adipogenic differentiation, whereas TLR4 inhibitor TAK242 (10 μmol/l) significantly attenuated this effect. CONCLUSIONS Our study suggested that bone marrow adipocyte may communicate with lung cancer cells via 1q21.3 (S100A8/A9-IL6R)-TLR4 pathway to promote osteolytic bone destruction. 1q21.3 (S100A8/A9-IL6R) is a potential target for the treatment of lung cancer bone metastasis.
Collapse
Affiliation(s)
- Guojing Luo
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Mengjia Tang
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zhao
- Department of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Lingyun Lu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Xie
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yujue Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chang Liu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Li Tian
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Chen
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
46
|
Tedesco S, Ciciliot S, Menegazzo L, D'Anna M, Scattolini V, Cappellari R, Cignarella A, Avogaro A, Albiero M, Fadini GP. Pharmacologic PPAR-γ Activation Reprograms Bone Marrow Macrophages and Partially Rescues HSPC Mobilization in Human and Murine Diabetes. Diabetes 2020; 69:1562-1572. [PMID: 32345753 DOI: 10.2337/db19-0640] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 04/22/2020] [Indexed: 11/13/2022]
Abstract
Mobilization of hematopoietic stem/progenitor cells (HSPC) from the bone marrow (BM) is impaired in diabetes. Excess oncostatin M (OSM) produced by M1 macrophages in the diabetic BM signals through p66Shc to induce Cxcl12 in stromal cells and retain HSPC. BM adipocytes are another source of CXCL12 that blunts mobilization. We tested a strategy of pharmacologic macrophage reprogramming to rescue HSPC mobilization. In vitro, PPAR-γ activation with pioglitazone switched macrophages from M1 to M2, reduced Osm expression, and prevented transcellular induction of Cxcl12 In diabetic mice, pioglitazone treatment downregulated Osm, p66Shc, and Cxcl12 in the hematopoietic BM, restored the effects of granulocyte-colony stimulation factor (G-CSF), and partially rescued HSPC mobilization, but it increased BM adipocytes. Osm deletion recapitulated the effects of pioglitazone on adipogenesis, which was p66Shc independent, and double knockout of Osm and p66Shc completely rescued HSPC mobilization. In the absence of OSM, BM adipocytes produced less CXCL12, being arguably devoid of HSPC-retaining activity, whereas pioglitazone failed to downregulate Cxcl12 in BM adipocytes. In patients with diabetes on pioglitazone therapy, HSPC mobilization after G-CSF was partially rescued. In summary, pioglitazone reprogrammed BM macrophages and suppressed OSM signaling, but sustained Cxcl12 expression by BM adipocytes could limit full recovery of HSPC mobilization.
Collapse
Affiliation(s)
- Serena Tedesco
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | | | | | - Marianna D'Anna
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Valentina Scattolini
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Roberta Cappellari
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | | | - Angelo Avogaro
- Department of Medicine, University of Padova, Padova, Italy
| | - Mattia Albiero
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
47
|
Liu Z, Liu H, He J, Lin P, Tong Q, Yang J. Myeloma cells shift osteoblastogenesis to adipogenesis by inhibiting the ubiquitin ligase MURF1 in mesenchymal stem cells. Sci Signal 2020; 13:13/633/eaay8203. [PMID: 32457115 DOI: 10.1126/scisignal.aay8203] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The suppression of bone formation is a hallmark of multiple myeloma. Myeloma cells inhibit osteoblastogenesis from mesenchymal stem cells (MSCs), which can also differentiate into adipocytes. We investigated myeloma-MSC interactions and the effects of such interactions on the differentiation of MSCs into adipocytes or osteoblasts using single-cell RNA sequencing, in vitro coculture, and subcutaneous injection of MSCs and myeloma cells into mice. Our results revealed that the α4 integrin subunit on myeloma cells stimulated vascular cell adhesion molecule-1 (VCAM1) on MSCs, leading to the activation of protein kinase C β1 (PKCβ1) signaling and repression of the muscle ring-finger protein-1 (MURF1)-mediated ubiquitylation of peroxisome proliferator-activated receptor γ2 (PPARγ2). Stabilized PPARγ2 proteins enhanced adipogenesis and consequently reduced osteoblastogenesis from MSCs, thus suppressing bone formation in vitro and in vivo. These findings reveal that suppressed bone formation is a direct consequence of myeloma-MSC contact that promotes the differentiation of MSCs into adipocytes at the expense of osteoblasts. Thus, this study provides a potential strategy for treating bone resorption in patients with myeloma by counteracting tumor-MSC interactions.
Collapse
Affiliation(s)
- Zhiqiang Liu
- Department of Lymphoma and Myeloma, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. .,Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, School of Basic Medical Science, Tianjin Medical University, Tianjin, China.,Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Huan Liu
- Department of Lymphoma and Myeloma, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for Hematologic Malignancy, Research Institute Houston Methodist Hospital, Houston, TX 77030, USA
| | - Jin He
- Department of Lymphoma and Myeloma, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Center for Hematologic Malignancy, Research Institute Houston Methodist Hospital, Houston, TX 77030, USA
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jing Yang
- Department of Lymphoma and Myeloma, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. .,Center for Hematologic Malignancy, Research Institute Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
48
|
Spurny M, Jiang Y, Sowah SA, Schübel R, Nonnenmacher T, Bertheau R, Kirsten R, Johnson T, Hillengass J, Schlett CL, von Stackelberg O, Ulrich CM, Kaaks R, Kauczor HU, Kühn T, Nattenmüller J. Changes in Bone Marrow Fat upon Dietary-Induced Weight Loss. Nutrients 2020; 12:nu12051509. [PMID: 32455947 PMCID: PMC7284630 DOI: 10.3390/nu12051509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/09/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Bone marrow fat is implicated in metabolism, bone health and haematological diseases. Thus, this study aims to analyse the impact of moderate weight loss on bone marrow fat content (BMFC) in obese, healthy individuals. Methods: Data of the HELENA-Trial (Healthy nutrition and energy restriction as cancer prevention strategies: a randomized controlled intervention trial), a randomized controlled trial (RCT) among 137 non-smoking, overweight or obese participants, were analysed to quantify the Magnetic Resonance Imaging (MRI)-derived BMFC at baseline, after a 12-week dietary intervention phase, and after a 50-week follow-up. The study cohort was classified into quartiles based on changes in body weight between baseline and week 12. Changes in BMFC in respect of weight loss were analysed by linear mixed models. Spearman’s coefficients were used to assess correlations between anthropometric parameters, blood biochemical markers, blood cells and BMFC. Results: Relative changes in BMFC from baseline to week 12 were 0.0 ± 0.2%, −3.2 ± 0.1%, −6.1 ± 0.2% and −11.5 ± 0.6% for Q1 to Q4. Across all four quartiles and for the two-group comparison, Q1 versus Q4, there was a significant difference (p < 0.05) for changes in BMFC. BMFC was not associated with blood cell counts and showed only weaker correlations (<0.3) with metabolic biomarkers. Conclusion: Weight loss is associated with a decrease of BMFC. However, BMFC showed no stronger associations with inflammatory and metabolic biomarkers.
Collapse
Affiliation(s)
- Manuela Spurny
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
| | - Yixin Jiang
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
| | - Solomon A. Sowah
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; (S.A.S.); (R.K.); (T.J.); (R.K.); (T.K.)
| | - Ruth Schübel
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; (S.A.S.); (R.K.); (T.J.); (R.K.); (T.K.)
| | - Tobias Nonnenmacher
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
| | - Robert Bertheau
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
| | - Romy Kirsten
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; (S.A.S.); (R.K.); (T.J.); (R.K.); (T.K.)
| | - Theron Johnson
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; (S.A.S.); (R.K.); (T.J.); (R.K.); (T.K.)
| | - Jens Hillengass
- Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, New York 14263, USA;
| | - Christopher L. Schlett
- Department of Diagnostic and Interventional Radiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, D-79106 Freiburg, Germany;
| | - Oyunbileg von Stackelberg
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
| | - Cornelia M. Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, 2000 Circle of Hope, Salt Lake City, UT 84112-5550, USA;
| | - Rudolf Kaaks
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; (S.A.S.); (R.K.); (T.J.); (R.K.); (T.K.)
| | - Hans-Ulrich Kauczor
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
| | - Tilman Kühn
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany; (S.A.S.); (R.K.); (T.J.); (R.K.); (T.K.)
| | - Johanna Nattenmüller
- Heidelberg University Hospital, Diagnostic and Interventional Radiology, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany; (M.S.); (Y.J.); (R.S.); (T.N.); (R.B.); (O.v.S.); (H.-U.K.)
- Correspondence: ; Tel.: +49-6221-5636462
| |
Collapse
|
49
|
Morris EV, Suchacki KJ, Hocking J, Cartwright R, Sowman A, Gamez B, Lea R, Drake MT, Cawthorn WP, Edwards CM. Myeloma Cells Down-Regulate Adiponectin in Bone Marrow Adipocytes Via TNF-Alpha. J Bone Miner Res 2020; 35:942-955. [PMID: 31886918 PMCID: PMC9328417 DOI: 10.1002/jbmr.3951] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 12/09/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023]
Abstract
Multiple myeloma is caused by abnormal plasma cells that accumulate in the bone marrow and interact with resident cells of the bone microenvironment to drive disease progression and development of an osteolytic bone disease. Bone marrow adipocytes (BMAds) are emerging as having important endocrine functions that can support myeloma cell growth and survival. However, how BMAds respond to infiltrating tumor cells remains poorly understood. Using the C57BL/KaLwRij murine model of myeloma, bone marrow adiposity was found to be increased in early stage myeloma with BMAds localizing along the tumor-bone interface at later stages of disease. Myeloma cells were found to uptake BMAd-derived lipids in vitro and in vivo, although lipid uptake was not associated with the ability of BMAds to promote myeloma cell growth and survival. However, BMAd-derived factors were found to increase myeloma cell migration, viability, and the evasion of apoptosis. BMAds are a major source of adiponectin, which is known to be myeloma-suppressive. Myeloma cells were found to downregulate adiponectin specifically in a model of BMAds but not in white adipocytes. The ability of myeloma cells to downregulate adiponectin was dependent at least in part on TNF-α. Collectively our data support the link between increased bone marrow adiposity and myeloma progression. By demonstrating how TNF-α downregulates BMAd-derived adiponectin, we reveal a new mechanism by which myeloma cells alter the bone microenvironment to support disease progression. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Emma V Morris
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,NIHR Oxford BRC, Oxford, UK.,Oxford Centre for Translational Myeloma Research, Oxford, UK
| | - Karla J Suchacki
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Joseph Hocking
- NIHR Oxford BRC, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Oxford, UK
| | - Rachel Cartwright
- NIHR Oxford BRC, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Oxford, UK
| | - Aneka Sowman
- NIHR Oxford BRC, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Oxford, UK
| | - Beatriz Gamez
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,NIHR Oxford BRC, Oxford, UK.,Oxford Centre for Translational Myeloma Research, Oxford, UK
| | - Ryan Lea
- NIHR Oxford BRC, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Oxford, UK
| | - Matthew T Drake
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - William P Cawthorn
- University/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Claire M Edwards
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,NIHR Oxford BRC, Oxford, UK.,Oxford Centre for Translational Myeloma Research, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Oxford, UK
| |
Collapse
|
50
|
Abstract
PURPOSE OF THE REVIEW The purpose of this review is to describe the in vitro and in vivo methods that researchers use to model and investigate bone marrow adipocytes (BMAds). RECENT FINDINGS The bone marrow (BM) niche is one of the most interesting and dynamic tissues of the human body. Relatively little is understood about BMAds, perhaps in part because these cells do not easily survive flow cytometry and histology processing and hence have been overlooked. Recently, researchers have developed in vitro and in vivo models to study normal function and dysfunction in the BM niche. Using these models, scientists and clinicians have noticed that BMAds, which form bone marrow adipose tissue (BMAT), are able to respond to numerous signals and stimuli, and communicate with local cells and distant tissues in the body. This review provides an overview of how BMAds are modeled and studied in vitro and in vivo.
Collapse
Affiliation(s)
- Michaela R Reagan
- Center for Molecular Medicine and Center for Translational Research, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA.
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, USA.
- School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA, USA.
| |
Collapse
|