1
|
Liu H, Liu L, Rosen CJ. Bone Marrow Adipocytes as Novel Regulators of Metabolic Homeostasis: Clinical Consequences of Bone Marrow Adiposity. Curr Obes Rep 2025; 14:9. [PMID: 39808256 DOI: 10.1007/s13679-024-00594-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW Bone marrow adipose tissue is a distinctive fat depot located within the skeleton, with the potential to influence both local and systemic metabolic processes. Although significant strides have been made in understanding bone marrow adipose tissue over the past decade, many questions remain regarding their precise lineage and functional roles. RECENT FINDINGS Recent studies have highlighted bone marrow adipose tissue's involvement in continuous cross-talk with other organs and systems, exerting both endocrine and paracrine functions that play a crucial role in metabolic homeostasis, skeletal remodeling, hematopoiesis, and the progression of bone metastases. The advancement of imaging techniques, particularly cross-sectional imaging, has profoundly expanded our understanding of the complexities beyond the traditional view of bone marrow adipose tissue as an inert depot. Notably, marrow adipocytes are anatomically and functionally distinct from brown, beige, and classic white adipocytes. Emerging evidence suggests that bone marrow adipocytes, bone marrow adipose tissue originate from the differentiation of bone marrow mesenchymal stromal cells; however, they appear to be a heterogeneous population with varying metabolic profiles, lipid compositions, secretory properties, and functional responses depending on their specific location within the bone marrow. This review provides an up-to-date synthesis of current knowledge on bone marrow adipocytes, emphasizing the relationships between bone marrow adipogenesis and factors such as aging, osteoporosis, obesity, and bone marrow tumors or metastases, thereby elucidating the mechanisms underlying musculoskeletal pathophysiology.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology &, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Linyi Liu
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA.
| |
Collapse
|
2
|
Stanisławowski M. Effect of adipose tissue on the development of multiple myeloma. Mol Biol Rep 2024; 52:74. [PMID: 39708277 DOI: 10.1007/s11033-024-10174-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Multiple myeloma (MM), also referred to as Kahler's disease, is a cancer characterized by the uncontrolled growth of abnormal plasma cells and is associated with alterations in the bone tissue microenvironment. Bone marrow adipose tissue (BMAT), which comprises approximately ten percent of total body fat, can influence the progression, survival, and drug resistance of MM cells through paracrine, hormonal, and metabolic pathways. Obesity can lead to an increase in BMAT mass, which not only disrupts bone metabolism but also reduces bone density, potentially progressing from monoclonal gammopathy of undetermined significance, a benign condition, to MM. A range of factors, including impaired fatty acid metabolism, increased production of adipokines that support myeloma, and heightened expression of oncogenic microRNAs in multiple myeloma, contribute to the progression of this incurable blood cancer. To better understand the relationship between excess adipose tissue accumulation and the risk of developing multiple myeloma, a comprehensive review of published data was conducted.
Collapse
Affiliation(s)
- Marcin Stanisławowski
- Department of Histology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland.
| |
Collapse
|
3
|
Lu Y, Zhou Z, Pan D. Genetic insights into the roles of fatty acids and gut microbiota in osteoarthritis: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40674. [PMID: 39654243 PMCID: PMC11630979 DOI: 10.1097/md.0000000000040674] [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: 09/18/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
Traditional observational studies have shown that fatty acids and gut microbiota are crucial in osteoarthritis (OA) progression, but their findings are often conflicting due to biases, confounding factors, and measurement errors. We conducted a two-sample Mendelian randomization analysis using genome-wide association study data on fatty acids from 136,016 individuals, the gut microbiota from 7738 individuals, and osteoarthritis from 314,870 individuals. Elevated levels of total (odds ratio [OR]: 0.92; 95% CI 0.84-1.00; P = .039), saturated fatty acids (OR: 0.91; 95% CI 0.84-0.99; P = .034), and linoleic acid (OR: 0.92; 95% CI 0.85-1.00; P = .040) were associated with reduced OA risk. In terms of gut microbiota, Bifidobacterium adolescentis (OR: 0.89; 95% CI 0.80-1.00; P = .048) and Escherichia (OR: 0.90; 95% CI 0.81-1.00; P = .042) demonstrated protective roles against OA. Conversely, Oscillibacter (OR: 1.16; 95% CI 1.00-1.34; P = .043), Bilophila (OR: 1.28; 95% CI 1.07-1.54; P = .007), Erysipelotrichaceae (OR: 1.08; 95% CI 1.00-1.16; P = .044), and Bilophila within the Desulfovibrionaceae family (OR: 1.19; 95% CI 1.04-1.36; P = .012) were associated with an increased risk of OA. The findings indicate that modulating dietary factors and gut microbiota can independently reduce the risk and progression of OA, potentially improving the quality of life and health management in aging populations.
Collapse
Affiliation(s)
- Yilei Lu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, China
| | - Zekun Zhou
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, China
| | - Ding Pan
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
4
|
Varzieva VG, Mesonzhnik NV, Ilgisonis IS, Belenkov YN, Kozhevnikova MV, Appolonova SA. Metabolomic biomarkers of multiple myeloma: A systematic review. Biochim Biophys Acta Rev Cancer 2024; 1879:189151. [PMID: 38986721 DOI: 10.1016/j.bbcan.2024.189151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
Multiple myeloma (MM) is an incurable malignancy of clonal plasma cells. Various diagnostic methods are used in parallel to accurately determine stage and severity of the disease. Identifying a biomarker or a panel of biomarkers could enhance the quality of medical care that patients receive by adopting a more personalized approach. Metabolomics utilizes high-throughput analytical platforms to examine the levels and quantities of biochemical compounds in biosamples. The aim of this review was to conduct a systematic literature search for potential metabolic biomarkers that may aid in the diagnosis and prognosis of MM. The review was conducted in accordance with PRISMA recommendations and was registered in PROSPERO. The systematic search was performed in PubMed, CINAHL, SciFinder, Scopus, The Cochrane Library and Google Scholar. Studies were limited to those involving people with clinically diagnosed MM and healthy controls as comparators. Articles had to be published in English and had no restrictions on publication date or sample type. The quality of articles was assessed according to QUADOMICS criteria. A total of 709 articles were collected during the literature search. Of these, 436 were excluded based on their abstract, with 26 more removed after a thorough review of the full text. Finally, 16 articles were deemed relevant and were subjected to further analysis of their data. A number of promising candidate biomarkers was discovered. Follow-up studies with large sample sizes are needed to determine their suitability for clinical applications.
Collapse
Affiliation(s)
- Valeria G Varzieva
- Department of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), Vernadskogo pr., 96, 119571 Moscow, Russia; Centre of Biopharmaceutical Analysis and Metabolomics, Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Nakhimovsky pr., 45, 117418 Moscow, Russia.
| | - Natalia V Mesonzhnik
- Centre of Biopharmaceutical Analysis and Metabolomics, Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Nakhimovsky pr., 45, 117418 Moscow, Russia.
| | - Irina S Ilgisonis
- Hospital Therapy No. 1 Department, Sechenov First Moscow State Medical University (Sechenov University), Bol'shaya Pirogovskaya st. 6/1, 119435 Moscow, Russia
| | - Yuri N Belenkov
- Hospital Therapy No. 1 Department, Sechenov First Moscow State Medical University (Sechenov University), Bol'shaya Pirogovskaya st. 6/1, 119435 Moscow, Russia
| | - Maria V Kozhevnikova
- Hospital Therapy No. 1 Department, Sechenov First Moscow State Medical University (Sechenov University), Bol'shaya Pirogovskaya st. 6/1, 119435 Moscow, Russia
| | - Svetlana A Appolonova
- Department of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), Vernadskogo pr., 96, 119571 Moscow, Russia; Centre of Biopharmaceutical Analysis and Metabolomics, Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Nakhimovsky pr., 45, 117418 Moscow, Russia
| |
Collapse
|
5
|
Bhowmick K, von Suskil M, Al-Odat OS, Elbezanti WO, Jonnalagadda SC, Budak-Alpdogan T, Pandey MK. Pathways to therapy resistance: The sheltering effect of the bone marrow microenvironment to multiple myeloma cells. Heliyon 2024; 10:e33091. [PMID: 39021902 PMCID: PMC11252793 DOI: 10.1016/j.heliyon.2024.e33091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Multiple Myeloma (MM) is a malignant expansion of plasma cells in the bone marrow (BM), resulting in a disease characterized by symptoms of end organ damage from light chain secretion, crowding of the BM, and bone lesions. Although the past two decades have been characterized by numerous novel therapies emerging, the disease remains incurable due to intrinsic or acquired drug resistance. A major player in MM's drug resistance arises from its intimate relationship with the BM microenvironment (BMME). Through stress-inducing conditions, soluble messengers, and physical adhesion to BM elements, the BMME activates numerous pathways in the myeloma cell. This not only propagates myeloma progression through survival and growth signals, but also specific mechanisms to circumvent therapeutic actions. In this review, we provide an overview of the BMME, the role of individual components in MM survival, and various therapy-specific resistance mechanisms reported in the literature.
Collapse
Affiliation(s)
- Kuntal Bhowmick
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Max von Suskil
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Weam Othman Elbezanti
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Hematology, MD Anderson Cancer Center at Cooper, Cooper University Health Care, Camden, NJ, USA
| | - Subash C. Jonnalagadda
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Rowan University, Glassboro, NJ, USA
| | - Tulin Budak-Alpdogan
- Department of Hematology, MD Anderson Cancer Center at Cooper, Cooper University Health Care, Camden, NJ, USA
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| |
Collapse
|
6
|
Zhong Y, Li Y, Sun W, Xiao M. Liposomes have a direct effect on multiple myeloma: a Mendelian randomization study. Front Oncol 2024; 14:1404744. [PMID: 38933448 PMCID: PMC11200114 DOI: 10.3389/fonc.2024.1404744] [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: 03/21/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Background Multiple myeloma (MM), a malignant disease of plasma cells originating in the bone marrow, is influenced significantly by genetic factors. Although plasma liposomes have been linked to MM, the nature of their potential causal relationship remains to be elucidated. This study aims to explore this relationship using Mendelian randomization (MR) analysis. Methods Liposome-associated genetic instrumental variables (IVs) were identified from plasma lipidomics data of 7,174 Finnish individuals within a Genome-Wide Association Study (GWAS) pooled database. A MM pooled dataset was sourced from a GWAS meta-analysis encompassing 150,797 individuals, including 598 MM patients and 218,194 controls. These IVs underwent MR analysis, adhering to strict criteria for correlation, independence, and the exclusion of confounders. The inverse variance weighted (IVW) method, MR-Egger method, weighted median (WM) method, and simple median were utilized for MR analysis assessment, alongside Cochran's Q test, MR-Egger intercept, MR-Pleiotropy Residual Sum and Outlier (MR-RESSO) method, and leave-one-out analysis for evaluating heterogeneity, multiplicity, and instrumental bias. Results The study identified 88 significant, independent single nucleotide polymorphisms (SNPs) as IVs for MR analysis, each with an F-statistic value above 10, indicating robustness against weak instrument bias. IVW analysis revealed associations between six plasma liposome components and MM risk (p < 0.05). Phosphatidylinositol (16:0_18:1) serum levels (odds ratio [OR] = 1.769, 95% confidence interval [CI]: 1.132-2.763, p = 0.012) and triacylglycerol (56:4) levels (p = 0.026, OR = 1.417, 95% CI: 1.042-1.926) were positively correlated with the risk of multiple myeloma development. Phosphatidylethanolamine (18:0_20:4) (p = 0.004, 95% CI: 0.621-0.916, OR = 0.754), phosphatidylcholine (18:2_20:4) (p = 0.004, OR = 0.680, 95% CI: 0.519-0.889), sterol ester (27:1/18:3) levels (p = 0.013, OR = 0.677, 95% CI: 0.498-0.922), and phosphatidylcholine (O-18:2_20:4) levels (OR = 0.710, 95% CI: 0.517-0.913, p = 0.033) were negatively associated with the risk of developing multiple myeloma. The Cochran's Q test did not detect statistical method heterogeneity, nor did the MR-RESSO test or the MR-Egger intercept detect horizontal pleiotropy; leave-one-out analyses confirmed the absence of bias from individual SNPs. Conclusions Our findings suggest a complex relationship between plasma liposome components and MM risk. Elevated serum levels of triacylglycerol and phosphatidylinositol are positively associated with MM risk, while certain phospholipids and sterol esters offer a protective effect. This study provides valuable insights into the clinical relevance of liposomes in the pathology of multiple myeloma.
Collapse
Affiliation(s)
- Yingbin Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhao Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weipeng Sun
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Mingfeng Xiao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
7
|
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
|
8
|
Chong PSY, Chooi JY, Lim JSL, Leow ACY, Toh SHM, Azaman I, Koh MY, Teoh PJ, Tan TZ, Chung TH, Chng WJ. Histone Methyltransferase NSD2 Activates PKCα to Drive Metabolic Reprogramming and Lenalidomide Resistance in Multiple Myeloma. Cancer Res 2023; 83:3414-3427. [PMID: 37463241 DOI: 10.1158/0008-5472.can-22-3481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/07/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023]
Abstract
Multiple myeloma cells undergo metabolic reprogramming in response to the hypoxic and nutrient-deprived bone marrow microenvironment. Primary oncogenes in recurrent translocations might be able to drive metabolic heterogeneity to survive the microenvironment that can present new vulnerabilities for therapeutic targeting. t(4;14) translocation leads to the universal overexpression of histone methyltransferase NSD2 that promotes plasma cell transformation through a global increase in H3K36me2. Here, we identified PKCα as an epigenetic target that contributes to the oncogenic potential of NSD2. RNA sequencing of t(4;14) multiple myeloma cell lines revealed a significant enrichment in the regulation of metabolic processes by PKCα, and the glycolytic gene, hexokinase 2 (HK2), was transcriptionally regulated by PKCα in a PI3K/Akt-dependent manner. Loss of PKCα displaced mitochondria-bound HK2 and reversed sensitivity to the glycolytic inhibitor 3-bromopyruvate. In addition, the perturbation of glycolytic flux led to a metabolic shift to a less energetic state and decreased ATP production. Metabolomics analysis indicated lactate as a differential metabolite associated with PKCα. As a result, PKCα conferred resistance to the immunomodulatory drugs (IMiD) lenalidomide in a cereblon-independent manner and could be phenocopied by either overexpression of HK2 or direct supplementation of lactate. Clinically, t(4;14) patients had elevated plasma lactate levels and did not benefit from lenalidomide-based regimens. Altogether, this study provides insights into the epigenetic-metabolism cross-talk in multiple myeloma and highlights the opportunity for therapeutic intervention that leverages the distinct metabolic program in t(4;14) myeloma. SIGNIFICANCE Aberrant glycolysis driven by NSD2-mediated upregulation of PKCα can be therapeutically exploited using metabolic inhibitors with lactate as a biomarker to identify high-risk patients who exhibit poor response towards IMiD-based regimens.
Collapse
Affiliation(s)
- Phyllis S Y Chong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jing-Yuan Chooi
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Julia S L Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Aaron C Y Leow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Sabrina Hui Min Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Irfan Azaman
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Mun Yee Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Phaik Ju Teoh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Tae-Hoon Chung
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Wee Joo Chng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Department of Hematology-Oncology, National University Cancer Institute of Singapore, National University Health System, Singapore
| |
Collapse
|
9
|
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: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [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
|
10
|
Metabolic Alterations in Multiple Myeloma: From Oncogenesis to Proteasome Inhibitor Resistance. Cancers (Basel) 2023; 15:cancers15061682. [PMID: 36980568 PMCID: PMC10046772 DOI: 10.3390/cancers15061682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Despite significant improvements in treatment strategies over the past couple of decades, multiple myeloma (MM) remains an incurable disease due to the development of drug resistance. Metabolic reprogramming is a key feature of cancer cells, including MM, and acts to fuel increased proliferation, create a permissive tumour microenvironment, and promote drug resistance. This review presents an overview of the key metabolic adaptations that occur in MM pathogenesis and in the development of resistance to proteasome inhibitors, the backbone of current MM therapy, and considers the potential for therapeutic targeting of key metabolic pathways to improve outcomes.
Collapse
|
11
|
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
|
12
|
Morelli E, Fulciniti M, Samur MK, Ribeiro CF, Wert-Lamas L, Henninger JE, Gullà A, Aktas-Samur A, Todoerti K, Talluri S, Park WD, Federico C, Scionti F, Amodio N, Bianchi G, Johnstone M, Liu N, Gramegna D, Maisano D, Russo NA, Lin C, Tai YT, Neri A, Chauhan D, Hideshima T, Shammas MA, Tassone P, Gryaznov S, Young RA, Anderson KC, Novina CD, Loda M, Munshi NC. A MIR17HG-derived long noncoding RNA provides an essential chromatin scaffold for protein interaction and myeloma growth. Blood 2023; 141:391-405. [PMID: 36126301 PMCID: PMC10082365 DOI: 10.1182/blood.2022016892] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/31/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) can drive tumorigenesis and are susceptible to therapeutic intervention. Here, we used a large-scale CRISPR interference viability screen to interrogate cell-growth dependency to lncRNA genes in multiple myeloma (MM) and identified a prominent role for the miR-17-92 cluster host gene (MIR17HG). We show that an MIR17HG-derived lncRNA, named lnc-17-92, is the main mediator of cell-growth dependency acting in a microRNA- and DROSHA-independent manner. Lnc-17-92 provides a chromatin scaffold for the functional interaction between c-MYC and WDR82, thus promoting the expression of ACACA, which encodes the rate-limiting enzyme of de novo lipogenesis acetyl-coA carboxylase 1. Targeting MIR17HG pre-RNA with clinically applicable antisense molecules disrupts the transcriptional and functional activities of lnc-17-92, causing potent antitumor effects both in vitro and in vivo in 3 preclinical animal models, including a clinically relevant patient-derived xenograft NSG mouse model. This study establishes a novel oncogenic function of MIR17HG and provides potent inhibitors for translation to clinical trials.
Collapse
Affiliation(s)
- Eugenio Morelli
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Mehmet K. Samur
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Caroline F. Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Leon Wert-Lamas
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA
| | - Jon E. Henninger
- Whitehead Institute of Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Annamaria Gullà
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Anil Aktas-Samur
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Katia Todoerti
- Department of Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Srikanth Talluri
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Woojun D. Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Cinzia Federico
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
| | - Francesca Scionti
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Nicola Amodio
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
| | - Giada Bianchi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Megan Johnstone
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Na Liu
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Doriana Gramegna
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Domenico Maisano
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nicola A. Russo
- Istituto di Ricerche Genetiche “G. Salvatore,” Biogem s.c.ar.l., Avellino, Italy
| | - Charles Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Yu-Tzu Tai
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Antonino Neri
- Department of Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
- Scientific Directorate, Azienda USL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Dharminder Chauhan
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Teru Hideshima
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Masood A. Shammas
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| | - Pierfrancesco Tassone
- Department of Clinical and Experimental Medicine, Magna Graecia University, Catanzaro, Italy
| | | | - Richard A. Young
- Whitehead Institute of Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA
| | - Kenneth C. Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Carl D. Novina
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Nikhil C. Munshi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
13
|
Lepore A, Kaci FN, Bubici C, Papa S. An Integrated Methodology to Quantify the Glycolytic Stress in Plasma Cell Myeloma in Response to Cytotoxic Drugs. Methods Mol Biol 2023; 2675:285-296. [PMID: 37258771 DOI: 10.1007/978-1-0716-3247-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy primarily localized within the bone marrow (BM). Myeloma plasma cells, like many other cancer cells, change their metabolism in response to internal and external stimuli. The main metabolic alterations of MM cells include deregulated glycolysis (commonly associated with enhanced uptake and utilization of glucose), lipid metabolism dysregulation, as well as deregulated mitochondrial respiration (commonly associated with the deregulated formation of reactive oxygen species). Over the past decade, the discovery of novel methodologies and the commercialization of sophisticated instrumentation and reagents have facilitated the detection of real-time changes in cellular bioenergetics. Of those, the Seahorse™ extracellular flux (XF) analyzer has been widely used to evaluate the glycolytic flux and mitochondrial respiration in many cell types. While adherent cell lines are easy to use with this technology, non-adherent suspension cells are more difficult to handle especially when their metabolic activities are being investigated in response to drug treatment. Here, we provide an integrated protocol that allows the detection of extracellular acidification rate (ECAR) of live myeloma plasma cells in response to chemotherapeutic drugs. Our optimized protocol consists of treating myeloma cells with cytotoxic drug of interest in a standard culture plate prior to the real-time analysis in the XF analyzer. Furthermore, we provide results of experiments in which the metabolic activities of myeloma cells in response to cytotoxic treatment were compared between the manufacturer's basic procedure and our optimized protocol. Our observations suggest that our integrated protocol can be used to achieve consistent, well-standardized results and thus it may have broad applications in studies focusing on the characterization of metabolic events in non-adherent suspension cells.
Collapse
Affiliation(s)
- Alessio Lepore
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Fatma Necmiye Kaci
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Concetta Bubici
- Department of Life Sciences, Center for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - Salvatore Papa
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK.
| |
Collapse
|
14
|
Oudaert I, Van der Vreken A, Maes A, De Bruyne E, De Veirman K, Vanderkerken K, Menu E. Metabolic cross-talk within the bone marrow milieu: focus on multiple myeloma. Exp Hematol Oncol 2022; 11:49. [PMID: 36050788 PMCID: PMC9438316 DOI: 10.1186/s40164-022-00303-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer cells are well-known for their capacity to adapt their metabolism to their increasing energy demands which is necessary for tumor progression. This is no different for Multiple Myeloma (MM), a hematological cancer which develops in the bone marrow (BM), whereby the malignant plasma cells accumulate and impair normal BM functions. It has become clear that the hypoxic BM environment contributes to metabolic rewiring of the MM cells, including changes in metabolite levels, increased/decreased activity of metabolic enzymes and metabolic shifts. These adaptations will lead to a pro-tumoral environment stimulating MM growth and drug resistance In this review, we discuss the identified metabolic changes in MM and the BM microenvironment and summarize how these identified changes have been targeted (by inhibitors, genetic approaches or deprivation studies) in order to block MM progression and survival.
Collapse
Affiliation(s)
- Inge Oudaert
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Arne Van der Vreken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Anke Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, 1090, Brussels, Belgium.
| |
Collapse
|
15
|
Metabolic Vulnerabilities in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14081905. [PMID: 35454812 PMCID: PMC9029117 DOI: 10.3390/cancers14081905] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) remains an incurable malignancy with eventual emergence of refractory disease. Metabolic shifts, which ensure the availability of sufficient energy to support hyperproliferation of malignant cells, are a hallmark of cancer. Deregulated metabolic pathways have implications for the tumor microenvironment, immune cell function, prognostic significance in MM and anti-myeloma drug resistance. Herein, we summarize recent findings on metabolic abnormalities in MM and clinical implications driven by metabolism that may consequently inspire novel therapeutic interventions. We highlight some future perspectives on metabolism in MM and propose potential targets that might revolutionize the field.
Collapse
|
16
|
Liu R, Gao D, Lv Y, Zhai M, He A. Importance of circulating adipocytokines in multiple myeloma: a systematic review and meta-analysis based on case-control studies. BMC Endocr Disord 2022; 22:29. [PMID: 35073877 PMCID: PMC8787905 DOI: 10.1186/s12902-022-00939-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Adipocytes and their products, adipocytokines, play important roles in the generation and development of multiple myeloma (MM). Studies have demonstrated some adipocytokines to be associated with MM, although those results are controversial. Therefore, we conducted a meta-analysis to verify the association of adipocytokines with MM. METHODS We performed a systematic retrieval of literature published prior to 26 October 2021. Standardized mean difference (SMD) with a 95% confidence interval (CI) was calculated to evaluate pooled effects. Subgroup analysis and meta-regression analysis were conducted to detect sources of heterogeneity. Sensitivity analysis was performed to evaluate the stability of the study. Publication bias was assessed by funnel plots and Egger's linear regression test. RESULTS Ten eligible studies with 1269 MM patients and 2158 controls were included. The pooled analyses indicated that circulating leptin levels of MM patients were significantly higher than control levels (SMD= 0.87, 95%CI: 0.33 to 1.41), while the circulating adiponectin levels in MM patients were significantly lower than controls with a pooled SMD of -0.49 (95%CI: -0.78 to -0.20). The difference of circulating resistin levels were not significant between MM patients and controls (SMD= -0.08, 95%CI: -0.55 to 0.39). Subgroup analysis and meta-regression analysis found that sample size, age, and sex were possible sources of heterogeneity. Sensitivity analysis demonstrated our pooled results to be stable. CONCLUSION Decreased circulating adiponectin and increased leptin levels were associated with the occurrence and development of MM. Adiponectin and leptin may be potential biomarkers and therapeutic targets for MM.
Collapse
Affiliation(s)
- Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157, 5th West Road, Xi'an, 710004, Shaanxi, China
| | - Dandan Gao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157, 5th West Road, Xi'an, 710004, Shaanxi, China
| | - Yang Lv
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157, 5th West Road, Xi'an, 710004, Shaanxi, China
| | - Meng Zhai
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157, 5th West Road, Xi'an, 710004, Shaanxi, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157, 5th West Road, Xi'an, 710004, Shaanxi, China.
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
17
|
Metabolic Disorders in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms222111430. [PMID: 34768861 PMCID: PMC8584036 DOI: 10.3390/ijms222111430] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/08/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy and is attributed to monoclonal proliferation of plasma cells in the bone marrow. Cancer cells including myeloma cells deregulate metabolic pathways to ensure proliferation, growth, survival and avoid immune surveillance, with glycolysis and glutaminolysis being the most identified procedures involved. These disorders are considered a hallmark of cancer and the alterations performed ensure that enough energy is available for rapid cell proliferation. An association between metabolic syndrome, inflammatory cytokinesand incidence of MM has been also described, while the use of metformin and statins has been identified as a positive prognostic factor for the disease course. In this review, we aim to present the metabolic disorders that occur in multiple myeloma, the potential defects on the immune system and the potential advantage of targeting the dysregulated pathways in order to enhance antitumor therapeutics.
Collapse
|
18
|
Multiple Myeloma and Hyperlipidemia - coincidence or related? J Clin Lipidol 2021. [DOI: 10.1016/j.jacl.2021.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
19
|
Xu G, Huang S, Peng J, Gao X, Li M, Yu S, Liu Z, Qie P, Wang Y, Yu S, Liu S, Wen H, Su L, Li P, Guang B, Dong R, Liu J, Yang T. Targeting lipid metabolism in multiple myeloma cells: rational development of a synergistic strategy with proteasome inhibitors. Br J Pharmacol 2021; 178:4741-4757. [PMID: 34378191 DOI: 10.1111/bph.15653] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Aberrant lipid metabolism is now recognized as a key feature of cancer cells. Our initial research on mass spectrometry-based analysis of lipids in a multiple myeloma (MM) cell line showed a significant accumulation of lipids in MM cells after proteasome inhibition. This finding prompted us to hypothesize that MM cell survival depends on the maximal utilization of abnormally accumulated lipids. Therefore, we explored whether lipid metabolism-modulating agents would synergize with proteasome inhibitors (PIs). EXPERIMENTAL APPROACH The abnormal massive lipid accumulation in MM cells was detected using mass spectrometry. Cell viability and cell apoptosis were detected to assess the synergistic effect of lipid regulators and PIs. Otherwise, a novel stable derivative (FCE) of fenofibrate (FEN) was synthesized and used to treat MM cells in vitro and in vivo along with ixazomib. ChIP-seq, western blotting and RT-qPCR were performed to explore the potential mechanism underlying the increase in lipid levels in MM cells after proteasome inhibition. KEY RESULTS The accumulation of lipids in MM cells was induced by proteasome inhibition. Lipid-lowering drugs and MG-132 exerted a synergistic effect to kill MM cells. FCE showed significant synergistic activity in vitro and in vivo with ixazomib. The abnormal lipid accumulation in MM cells that was enhanced by proteasome inhibitors might be due to the elevated SREBP1/2 expression induced by ATF4. CONCLUSIONS AND IMPLICATIONS In summary, the results provide a proof of principle and rationale for the further clinical evaluation of the combination of lipid-modulating drugs with proteasome inhibitors.
Collapse
Affiliation(s)
- Gaojie Xu
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Sheng Huang
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Jian Peng
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Xiaofang Gao
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Minhui Li
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Sisi Yu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Zuofeng Liu
- Department of Hematology, The Third People's Hospital of Chengdu, Chengdu, Sichuan Province, China
| | - Pengfan Qie
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Yu Wang
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Siqi Yu
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Siyuan Liu
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Hu Wen
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Lijuan Su
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Ping Li
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Bin Guang
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China.,Chengdu Biobel Biotechnology Co., Ltd., Chengdu, Sichuan Province, China
| | - Renhan Dong
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China.,Chengdu Biobel Biotechnology Co., Ltd., Chengdu, Sichuan Province, China
| | - Jin Liu
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Tai Yang
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan Province, China
| |
Collapse
|
20
|
Giannattasio S, Dri M, Merra G, Caparello G, Rampello T, Di Renzo L. Effects of Fatty Acids on Hematological Neoplasms: A Mini Review. Nutr Cancer 2021; 74:1538-1548. [PMID: 34355630 DOI: 10.1080/01635581.2021.1960389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Hematological neoplasias are the fourth cause of death in the world. All of them are responsible of bad quality of life, due to heavy therapies administration and a lot of side effects correlated to. It arises a new concept of "multitherapy", in which fatty acids availment is used to contrast and reduce toxic effects and ameliorate chemotherapeutic agents asset. In Vitro studies have confirmed that fatty acids, in particular ω-3 eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are able to help canonical therapies to contrast cancer cell expansion and proliferation. In clinical trials it is also almost clear that fatty acids are useful to build new personalized therapies for a better condition of life. In this review we have summarized most recent studies on cancer cell lines and clinical trials on patients with fatty acids supplementation in diet therapies. We have found that fatty acids could be useful to contrast side effects during chemotherapeutic drugs therapies; they are also able to block cancer cell metabolic pathways for proliferation and contrast adverse effects, even when they are used in combination with traditional therapies or innovative, like monoclonal antibodies or CAR-T therapy. These aspects are crucial for better health condition of patients.
Collapse
Affiliation(s)
- Silvia Giannattasio
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Maria Dri
- Doctoral School of Applied Medical and Surgical Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Merra
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Giovanna Caparello
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Tiziana Rampello
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Rome, Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
21
|
The fatty acid elongase ELOVL6 regulates bortezomib resistance in multiple myeloma. Blood Adv 2021; 5:1933-1946. [PMID: 33821992 DOI: 10.1182/bloodadvances.2020002578] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 02/10/2021] [Indexed: 12/24/2022] Open
Abstract
Resistance to the proteasome inhibitor bortezomib (BTZ) represents a major obstacle in the treatment of multiple myeloma (MM). The contribution of lipid metabolism in the resistance of MM cells to BTZ is mostly unknown. Here we report that levels of fatty acid elongase 6 (ELOVL6) were lower in MM cells from BTZ-nonresponsive vs BTZ-responsive patients and in cultured MM cells selected for BTZ resistance compared with parental counterparts. Accordingly, depletion of ELOVL6 in parental MM cells suppressed BTZ-induced endoplasmic reticulum (ER) stress and cytotoxicity, whereas restoration of ELOVL6 levels in BTZ-resistant MM cells sensitized them to BTZ in tissue culture settings and, as xenografts, in a plasmacytoma mouse model. Furthermore, for the first time, we identified changes in the BTZ-induced lipidome between parental and BTZ-resistant MM cell lines underlying a functional difference in their response to BTZ. We demonstrated that restoration of ELOVL6 levels in BTZ-resistant MM cells resensitized them to BTZ largely via upregulation of ELOVL6-dependent ceramide species, which was a prerequisite for BTZ-induced ER stress and cell death in these cells. Our data characterize ELOVL6 as a major clinically relevant regulator of MM cell resistance to BTZ, which can emerge from the impaired ability of these cells to alter ceramide composition in response to BTZ.
Collapse
|
22
|
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
|
23
|
Search for multiple myeloma risk factors using Mendelian randomization. Blood Adv 2021; 4:2172-2179. [PMID: 32433745 DOI: 10.1182/bloodadvances.2020001502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/10/2020] [Indexed: 01/01/2023] Open
Abstract
The etiology of multiple myeloma (MM) is poorly understood. Summary data from genome-wide association studies (GWASs) of multiple phenotypes can be exploited in a Mendelian randomization (MR) phenome-wide association study (PheWAS) to search for factors influencing MM risk. We performed an MR-PheWAS analyzing 249 phenotypes, proxied by 10 225 genetic variants, and summary genetic data from a GWAS of 7717 MM cases and 29 304 controls. Odds ratios (ORs) per 1 standard deviation increase in each phenotype were estimated under an inverse variance weighted random effects model. A Bonferroni-corrected threshold of P = 2 × 10-4 was considered significant, whereas P < .05 was considered suggestive of an association. Although no significant associations with MM risk were observed among the 249 phenotypes, 28 phenotypes showed evidence suggestive of association, including increased levels of serum vitamin B6 and blood carnitine (P = 1.1 × 10-3) with greater MM risk and ω-3 fatty acids (P = 5.4 × 10-4) with reduced MM risk. A suggestive association between increased telomere length and reduced MM risk was also noted; however, this association was primarily driven by the previously identified risk variant rs10936599 at 3q26 (TERC). Although not statistically significant, increased body mass index was associated with increased risk (OR, 1.10; 95% confidence interval, 0.99-1.22), supporting findings from a previous meta-analysis of prospective observational studies. Our study did not provide evidence supporting any modifiable factors examined as having a major influence on MM risk; however, it provides insight into factors for which the evidence has previously been mixed.
Collapse
|
24
|
Cancer immunoediting and immune dysregulation in multiple myeloma. Blood 2021; 136:2731-2740. [PMID: 32645135 DOI: 10.1182/blood.2020006540] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/27/2020] [Indexed: 12/15/2022] Open
Abstract
Avoiding immune destruction is a hallmark of cancer. Over the past few years, significant advances have been made in understanding immune dysfunction and immunosuppression in multiple myeloma (MM), and various immunotherapeutic approaches have delivered improved clinical responses. However, it is still challenging to completely eliminate malignant plasma cells (PCs) and achieve complete cure. The interplay between the immune system and malignant PCs is implicated throughout all stages of PC dyscrasias, including asymptomatic states called monoclonal gammopathy of undetermined significance and smoldering myeloma. Although the immune system effectively eliminates malignant PCs, or at least induces functional dormancy at early stages, malignant PCs eventually evade immune elimination, leading to progression to active MM, in which dysfunctional effector lymphocytes, tumor-educated immunosuppressive cells, and soluble mediators coordinately act as a barrier for antimyeloma immunity. An in-depth understanding of this dynamic process, called cancer immunoediting, will provide important insights into the immunopathology of PC dyscrasias and MM immunotherapy. Moreover, a growing body of evidence suggests that, together with nonhematopoietic stromal cells, bone marrow (BM) immune cells with unique functions support the survival of normal and malignant PCs in the BM niche, highlighting the diverse roles of immune cells beyond antimyeloma immunity. Together, the immune system critically acts as a rheostat that fine-tunes the balance between dormancy and disease progression in PC dyscrasias.
Collapse
|
25
|
Shimizu M, Tachikawa S, Saitoh N, Nakazono K, Yu-Jung L, Suga M, Ohnuma K. Thalidomide affects limb formation and multiple myeloma related genes in human induced pluripotent stem cells and their mesoderm differentiation. Biochem Biophys Rep 2021; 26:100978. [PMID: 33763605 PMCID: PMC7973312 DOI: 10.1016/j.bbrep.2021.100978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022] Open
Abstract
Although thalidomide is highly teratogenic, it has been prescribed for treating multiple myeloma and Hansen's disease. However, its mechanism of action is not fully understood. Here, we employed a reverse transcription quantitative PCR array to measure the expression of 84 genes in human induced pluripotent stem cells (hiPSCs) and their mesodermal differentiation. Thalidomide altered the expression of undifferentiated marker genes in both cell types. Thalidomide affected more genes in the mesoderm than in the hiPSCs. Ectoderm genes were upregulated but mesendoderm genes were downregulated by thalidomide during mesoderm induction, suggesting that thalidomide altered mesoderm differentiation. We found that FABP7 (fatty acid binding protein 7) was dramatically downregulated in the hiPSCs. FABP is related to retinoic acid, which is important signaling for limb formation. Moreover, thalidomide altered the expression of the genes involved in TGF-β signaling, limb formation, and multiple myeloma, which are related to thalidomide-induced malformations and medication. In summary, iPSCs can serve as useful tools to elucidate the mechanisms underlying thalidomide malformations in vitro. Thalidomide downregulated FABP7, a fatty acid binding protein (FABP) cording gene. FABP is related to retinoic acid, which is important signaling for limb formation. Thalidomide treatment affected the expression of limb formation related genes. Thalidomide treatment affected 5 genes related to multiple myeloma. Thalidomide upregulated ectoderm but downregulated mesendoderm markers in mesoderm.
Collapse
Affiliation(s)
- Maho Shimizu
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Saoko Tachikawa
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Nagatsuki Saitoh
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Kohei Nakazono
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | - Liu Yu-Jung
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka, Ibaraki, 567-0085, Japan
| | - Mika Suga
- Laboratory of Stem Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Osaka, Ibaraki, 567-0085, Japan
| | - Kiyoshi Ohnuma
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan.,Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Shiozawa Y. The Roles of Bone Marrow-Resident Cells as a Microenvironment for Bone Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1226:57-72. [PMID: 32030676 DOI: 10.1007/978-3-030-36214-0_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It has been appreciated that the cross talk between bone metastatic cancer cells and bone marrow microenvironment influence one another to worsen bone metastatic disease progression. Bone marrow contains various cell types, including (1) cells of mesenchymal origin (e.g., osteoblasts, osteocytes, and adipocytes), (2) cells of hematopoietic origin (e.g., osteoclast and immune cells), and (3) others (e.g., endothelial cells and nerves). The recent studies have enabled us to discover many important cancer-derived factors responsible for the development of bone metastasis. However, many critical questions regarding the roles of bone microenvironment in bone metastatic progression remain elusive. To answer these questions, a deeper understanding of the cross talk between bone metastatic cancer and bone marrow microenvironment is clearly warranted.
Collapse
Affiliation(s)
- Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
29
|
Johnson AA, Stolzing A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell 2019; 18:e13048. [PMID: 31560163 PMCID: PMC6826135 DOI: 10.1111/acel.13048] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/11/2019] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
An emerging body of data suggests that lipid metabolism has an important role to play in the aging process. Indeed, a plethora of dietary, pharmacological, genetic, and surgical lipid‐related interventions extend lifespan in nematodes, fruit flies, mice, and rats. For example, the impairment of genes involved in ceramide and sphingolipid synthesis extends lifespan in both worms and flies. The overexpression of fatty acid amide hydrolase or lysosomal lipase prolongs life in Caenorhabditis elegans, while the overexpression of diacylglycerol lipase enhances longevity in both C. elegans and Drosophila melanogaster. The surgical removal of adipose tissue extends lifespan in rats, and increased expression of apolipoprotein D enhances survival in both flies and mice. Mouse lifespan can be additionally extended by the genetic deletion of diacylglycerol acyltransferase 1, treatment with the steroid 17‐α‐estradiol, or a ketogenic diet. Moreover, deletion of the phospholipase A2 receptor improves various healthspan parameters in a progeria mouse model. Genome‐wide association studies have found several lipid‐related variants to be associated with human aging. For example, the epsilon 2 and epsilon 4 alleles of apolipoprotein E are associated with extreme longevity and late‐onset neurodegenerative disease, respectively. In humans, blood triglyceride levels tend to increase, while blood lysophosphatidylcholine levels tend to decrease with age. Specific sphingolipid and phospholipid blood profiles have also been shown to change with age and are associated with exceptional human longevity. These data suggest that lipid‐related interventions may improve human healthspan and that blood lipids likely represent a rich source of human aging biomarkers.
Collapse
|
30
|
Pojero F, Poma P, Spanò V, Montalbano A, Barraja P, Notarbartolo M. Targeting multiple myeloma with natural polyphenols. Eur J Med Chem 2019; 180:465-485. [DOI: 10.1016/j.ejmech.2019.07.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/16/2022]
|