1
|
Wu Y, Yao X, Shi X, Xu Z, Ren J, Shi M, Li M, Liu J, Du X. Myeloma extracellular vesicle-derived RAGE increases inflammatory responses and myotube atrophy in multiple myeloma through activation of the TLR4/NF-κB p65 pathway. Apoptosis 2024; 29:849-864. [PMID: 38117373 DOI: 10.1007/s10495-023-01920-7] [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] [Accepted: 11/06/2023] [Indexed: 12/21/2023]
Abstract
Sarcopenia manifests as muscle atrophy and loss that is complicated with malignancy. This study explored the mechanism of extracellular vesicles (EVs) in multiple myeloma (MM) with sarcopenia. SP2/0 conditioned medium (CM) was collected to isolate SP2/0-EVs. C2C12 cells were incubated with SP2/0 CM or SP2/0-EVs. ROS, TNF-α, IL-6, MuRF1 and MyHC levels were detected by DCF-DA fluorescent probe, ELISA, and Western blot. GW4869 was used to inhibit EV secretion in SP2/0 to confirm its effect on muscle atrophy. Serum was collected from MM patients with or without sarcopenia to detect RAGE mRNA expression. SP2/0 cells were transfected with RAGE siRNA and C2C12 cells were treated with the isolated si-RAGE-EVs or/and TLR4 agonist. SP2/0 tumor-bearing mouse model was established. Healthy mice and SP2/0-tumor bearing mice were treated with SP2/0-EVs or si-RAGE-EVs. SP2/0 CM or SP2/0-EVs stimulated ROS, inflammatory responses, and myotube atrophy in C2C12 cells. GW4869 blocked EV secretion and the effects of SP2/0 CM. RAGE mRNA expression in serum EVs was increased in MM&Sarcopenia patients and RAGE knockdown in SP2/0-EVs partially nullified SP2/0-EVs' effects. SP2/0-EVs activated the TLR4/NF-κB p65 pathway by translocating RAGE. SP2/0-EVs-derived RAGE elevated ROS production, inflammation, and myotube atrophy in C2C12 cells and caused muscle loss in SP2/0 tumor-bearing mice by activating the TLR4/NF-κB p65 pathway. SP2/0-EVs partially recapitulated muscle loss in healthy mice. SP2/0-EVs-derived RAGE increased ROS production, inflammation, and myotube atrophy in MM through TLR4/NF-κB p65 pathway activation.
Collapse
Affiliation(s)
- Yue Wu
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Xingchen Yao
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Xiangjun Shi
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Ziyu Xu
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Jie Ren
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Ming Shi
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Meng Li
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Junpeng Liu
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China
| | - Xinru Du
- Department of Orthopedics, Beijing Chao-Yang Hospital, Beijing, China.
- Beijing Chao-Yang Hospital, No.8 Gongti South Rd, Chaoyang District, 100020, Beijing, China.
| |
Collapse
|
2
|
Krishnan SR, Bebawy M. Circulating biosignatures in multiple myeloma and their role in multidrug resistance. Mol Cancer 2023; 22:79. [PMID: 37120508 PMCID: PMC10148481 DOI: 10.1186/s12943-022-01683-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 11/14/2022] [Indexed: 05/01/2023] Open
Abstract
A major obstacle to chemotherapeutic success in cancer treatment is the development of drug resistance. This occurs when a tumour fails to reduce in size after treatment or when there is clinical relapse after an initial positive response to treatment. A unique and serious type of resistance is multidrug resistance (MDR). MDR causes the simultaneous cross resistance to unrelated drugs used in chemotherapy. MDR can be acquired through genetic alterations following drug exposure, or as discovered by us, through alternative pathways mediated by the transfer of functional MDR proteins and nucleic acids by extracellular vesicles (M Bebawy V Combes E Lee R Jaiswal J Gong A Bonhoure GE Grau, 23 9 1643 1649, 2009).Multiple myeloma is an incurable cancer of bone marrow plasma cells. Treatment involves high dose combination chemotherapy and patient response is unpredictable and variable due to the presence of multisite clonal tumour infiltrates. This clonal heterogeneity can contribute to the development of MDR. There is currently no approved clinical test for the minimally invasive testing of MDR in myeloma.Extracellular vesicles comprise a group of heterogeneous cell-derived membranous structures which include; exosomes, microparticles (microvesicles), migrasomes and apoptotic bodies. Extracellular vesicles serve an important role in cellular communication through the intercellular transfer of cellular protein, nucleic acid and lipid cargo. Of these, microparticles (MPs) originate from the cell plasma membrane and vary in size from 0.1-1um. We have previously shown that MPs confer MDR through the transfer of resistance proteins and nucleic acids. A test for the early detection of MDR would benefit clinical decision making, improve survival and support rational drug use. This review focuses on microparticles as novel clinical biomarkers for the detection of MDR in Myeloma and discusses their role in the therapeutic management of the disease.
Collapse
|
3
|
Bergantim R, Peixoto da Silva S, Polónia B, Barbosa MAG, Albergaria A, Lima J, Caires HR, Guimarães JE, Vasconcelos MH. Detection of Measurable Residual Disease Biomarkers in Extracellular Vesicles from Liquid Biopsies of Multiple Myeloma Patients-A Proof of Concept. Int J Mol Sci 2022; 23:13686. [PMID: 36430163 PMCID: PMC9690807 DOI: 10.3390/ijms232213686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Monitoring measurable residual disease (MRD) is crucial to assess treatment response in Multiple Myeloma (MM). Detection of MRD in peripheral blood (PB) by exploring Extracellular Vesicles (EVs), and their cargo, would allow frequent and minimally invasive monitoring of MM. This work aims to detect biomarkers of MRD in EVs isolated from MM patient samples at diagnosis and remission and compare the MRD-associated content between BM and PB EVs. EVs were isolated by size-exclusion chromatography, concentrated by ultrafiltration, and characterized according to their size and concentration, morphology, protein concentration, and the presence of EV-associated protein markers. EVs from healthy blood donors were used as controls. It was possible to isolate EVs from PB and BM carrying MM markers. Diagnostic samples had different levels of MM markers between PB and BM paired samples, but no differences between PB and BM were found at remission. EVs concentration was lower in the PB of healthy controls than of patients, and MM markers were mostly not detected in EVs from controls. This study pinpoints the potential of PB EVs from MM remission patients as a source of MM biomarkers and as a non-invasive approach for monitoring MRD.
Collapse
Affiliation(s)
- Rui Bergantim
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital Center of São João, 4200-319 Porto, Portugal
- Clinical Hematology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Sara Peixoto da Silva
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Bárbara Polónia
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Mélanie A. G. Barbosa
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - André Albergaria
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Research Innovation Unit, Translational Research & Industry Partnerships Office, i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Jorge Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Research Innovation Unit, Translational Research & Industry Partnerships Office, i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Hugo R. Caires
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital Center of São João, 4200-319 Porto, Portugal
- Clinical Hematology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário IUCSESPU, 4585-116 Gandra-Paredes, Portugal
| | - M. Helena Vasconcelos
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
4
|
Brennan K, Iversen KF, Blanco-Fernández A, Lund T, Plesner T, Mc Gee MM. Extracellular Vesicles Isolated from Plasma of Multiple Myeloma Patients Treated with Daratumumab Express CD38, PD-L1, and the Complement Inhibitory Proteins CD55 and CD59. Cells 2022; 11:3365. [PMID: 36359760 PMCID: PMC9658084 DOI: 10.3390/cells11213365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 09/26/2023] Open
Abstract
Daratumumab (DARA) has improved the outcome of treatment of multiple myeloma (MM). DARA acts via complement-dependent and -independent mechanisms. Resistance to DARA may result from upregulation of the complement inhibitory proteins CD55 and CD59, downregulation of the DARA target CD38 on myeloma cells or altered expression of the checkpoint inhibitor ligand programmed death ligand-1 (PD-L1) or other mechanisms. In this study, EVs were isolated from peripheral blood (PB) and bone marrow (BM) from multiple myeloma (MM) patients treated with DARA and PB of healthy controls. EV size and number and the expression of CD38, CD55, CD59 and PD-L1 as well as the EV markers CD9, CD63, CD81, CD147 were determined by flow cytometry. Results reveal that all patient EV samples express CD38, PD-L1, CD55 and CD59. The level of CD55 and CD59 are elevated on MM PB EVs compared with healthy controls, and the level of PD-L1 on MM PB EVs is higher in patients responding to treatment with DARA. CD147, a marker of various aspects of malignant behaviour of cancer cells and a potential target for therapy, was significantly elevated on MM EVs compared with healthy controls. Furthermore, mass spectrometry data suggests that MM PB EVs bind DARA. This study reveals a MM PB and BM EV protein signature that may have diagnostic and prognostic value.
Collapse
Affiliation(s)
- Kieran Brennan
- School of Biomolecular & Biomedical Science, University College Dublin (UCD), Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin 4, Ireland
| | - Katrine F. Iversen
- Institute of Regional Health Science, University of Southern Denmark, 7100 Vejle, Denmark
- Department of Internal Medicine, Section of Hematology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Alfonso Blanco-Fernández
- Flow Cytometry Core Technology, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin 4, Ireland
| | - Thomas Lund
- Department of Hematology, Odense University Hospital, 5000 Odense, Denmark
| | - Torben Plesner
- Institute of Regional Health Science, University of Southern Denmark, 7100 Vejle, Denmark
- Department of Internal Medicine, Section of Hematology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Margaret M. Mc Gee
- School of Biomolecular & Biomedical Science, University College Dublin (UCD), Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin 4, Ireland
| |
Collapse
|
5
|
Goodarzi A, Valikhani M, Amiri F, Safari A. The mechanisms of mutual relationship between malignant hematologic cells and mesenchymal stem cells: Does it contradict the nursing role of mesenchymal stem cells? Cell Commun Signal 2022; 20:21. [PMID: 35236376 PMCID: PMC8889655 DOI: 10.1186/s12964-022-00822-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known as the issue in biology because of some unpredictable characteristics in the different microenvironments especially in their bone marrow niche. MSCs are used in the regenerative medicine because of their unique potentials for trans-differentiation, immunomodulation, and paracrine capacity. But, their pathogenic and pro-survival effects in tumors/cancers including hematologic malignancies are indisputable. MSCs and/or their derivatives might be involved in tumor growth, metastasis and drug resistance in the leukemias. One of important relationship is MSCs and hematologic malignancy-derived cells which affects markedly the outcome of disease. The communication between these two cells may be contact-dependent and/or contact-independent. In this review, we studied the crosstalk between MSCs and malignant hematologic cells which results the final feedback either the progression or suppression of blood cell malignancy. Video abstract.
Collapse
Affiliation(s)
- Alireza Goodarzi
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Shahid Fahmideh Blvd., The Opposite Side of Mardom Park, Hamadan, 6517838741, Iran
| | - Mohsen Valikhani
- Hematology Department, School of Allied Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amiri
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Shahid Fahmideh Blvd., The Opposite Side of Mardom Park, Hamadan, 6517838741, Iran.
| | - Armita Safari
- Student Research Committee, Hamadan University of Medical Science, Hamadan, Iran
| |
Collapse
|
6
|
Saltarella I, Lamanuzzi A, Desantis V, Di Marzo L, Melaccio A, Curci P, Annese T, Nico B, Solimando AG, Bartoli G, Tolomeo D, Storlazzi CT, Mariggiò MA, Ria R, Musto P, Vacca A, Frassanito MA. Myeloma cells regulate
miRNA
transfer from fibroblast‐derived exosomes by expression of
lncRNAs. J Pathol 2021; 256:402-413. [DOI: 10.1002/path.5852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
- Department of Biomedical Sciences and Human Oncology Pharmacology Section, University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Lucia Di Marzo
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Assunta Melaccio
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Paola Curci
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico Bari Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School Bari Italy
| | - Beatrice Nico
- Department of Basic Medical Sciences Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School Bari Italy
| | - Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
- IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari Italy
| | - Giulia Bartoli
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Doron Tolomeo
- Department of Biology University of Bari "Aldo Moro", Via E. Orabona no. 4, 70125 Bari Italy
| | | | - Maria Addolorata Mariggiò
- Department of Biomedical Sciences and Human Oncology Unit of General Pathology, University of Bari "Aldo Moro", 70124 Bari Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico Bari Italy
- Department of Emergency and Organ Transplantation "Aldo Moro", University School of Medicine Bari Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology Unit of Internal Medicine "Guido Baccelli", University of Bari Medical School, Piazza Giulio Cesare 11, 70124 Bari Italy
| | - Maria Antonia Frassanito
- Department of Biomedical Sciences and Human Oncology Unit of General Pathology, University of Bari "Aldo Moro", 70124 Bari Italy
| |
Collapse
|
7
|
tRNA Derivatives in Multiple Myeloma: Investigation of the Potential Value of a tRNA-Derived Molecular Signature. Biomedicines 2021; 9:biomedicines9121811. [PMID: 34944627 PMCID: PMC8698603 DOI: 10.3390/biomedicines9121811] [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: 10/01/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 01/11/2023] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy arising from the clonal proliferation of malignant plasma cells. tRNA-derived RNA fragments (tRFs) constitute a class of small non-coding RNAs, deriving from specific enzymatic cleavage of tRNAs. To the best of our knowledge, this is one of few studies to uncover the potential clinical significance of tRFs in MM. Total RNA was extracted from CD138+ plasma cells of MM and smoldering MM patients, and in vitro polyadenylated. First-strand cDNA synthesis was performed, priming from an oligo-dT-adaptor sequence. Next, real-time quantitative PCR (qPCR) assays were developed for the quantification of six tRFs. Biostatistical analysis was performed to assess the results and in silico analysis was conducted to predict the function of one of the tRFs. Our results showed that elevated levels of five out of six tRFs are indicators of favorable prognosis in MM, predicting prolonged overall survival (OS), while two of them constitute potential molecular biomarkers of favorable prognosis in terms of disease progression. Moreover, three tRFs could be used as surrogate prognostic biomarkers along with the R-ISS staging system to predict OS. In conclusion, tRFs show molecular biomarker utility in MM, while their mechanisms of function merit further investigation.
Collapse
|
8
|
Reale A, Khong T, Mithraprabhu S, Spencer A. Translational Potential of RNA Derived From Extracellular Vesicles in Multiple Myeloma. Front Oncol 2021; 11:718502. [PMID: 34513695 PMCID: PMC8429596 DOI: 10.3389/fonc.2021.718502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
The cross-talk between tumour cells and stromal cells is a hallmark of multiple myeloma (MM), a blood cancer that still remains incurable despite increased knowledge of its biology and advances in its treatment. Extracellular vesicles (EVs) derived from both tumour and stromal cells have been shown to play an important role in mediating this cross-talk ultimately favouring MM progression and drug resistance. Furthermore, EVs and their content including RNA (EV-RNA) have been successfully isolated from blood and are being explored as liquid biomarkers in MM with the potential to improve diagnosis and monitoring modalities with a minimally-invasive and repeatable analysis, i.e. liquid biopsy. In this review, we describe both the role of EV-RNA in defining the biological features of MM and their potential translational relevance as liquid biomarkers, therapeutic targets and delivery systems. We also discuss the limitations and technical challenges related to the isolation and characterization of EVs and provide a perspective on the future of MM-derived EV-RNA in translational research.
Collapse
Affiliation(s)
- Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia
| | - Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University/Alfred Health, Melbourne, VIC, Australia.,Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, and Department of Clinical Haematology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
9
|
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
|
10
|
Laurenzana I, Trino S, Lamorte D, Girasole M, Dinarelli S, De Stradis A, Grieco V, Maietti M, Traficante A, Statuto T, Villani O, Musto P, Sgambato A, De Luca L, Caivano A. Analysis of Amount, Size, Protein Phenotype and Molecular Content of Circulating Extracellular Vesicles Identifies New Biomarkers in Multiple Myeloma. Int J Nanomedicine 2021; 16:3141-3160. [PMID: 33994784 PMCID: PMC8114829 DOI: 10.2147/ijn.s303391] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/11/2021] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Extracellular vesicles (EVs) are naturally secreted cellular lipid bilayer particles, which carry a selected molecular content. Owing to their systemic availability and their role in tumor pathogenesis, circulating EVs (cEVs) can be a valuable source of new biomarkers useful for tumor diagnosis, prognostication and monitoring. However, a precise approach for isolation and characterization of cEVs as tumor biomarkers, exportable in a clinical setting, has not been conclusively established. METHODS We developed a novel and laboratory-made procedure based on a bench centrifuge step which allows the isolation of serum cEVs suitable for subsequent characterization of their size, amount and phenotype by nanoparticle tracking analysis, microscopy and flow cytometry, and for nucleic acid assessment by digital PCR. RESULTS Applied to blood from healthy subjects (HSs) and tumor patients, our approach permitted from a small volume of serum (i) the isolation of a great amount of EVs enriched in small vesicles free from protein contaminants; (ii) a suitable and specific cell origin identification of EVs, and (iii) nucleic acid content assessment. In clonal plasma cell malignancy, like multiple myeloma (MM), our approach allowed us to identify specific MM EVs, and to characterize their size, concentration and microRNA content allowing significant discrimination between MM and HSs. Finally, EV associated biomarkers correlated with MM clinical parameters. CONCLUSION Overall, our cEV based procedure can play an important role in malignancy biomarker discovery and then in real-time tumor monitoring using minimal invasive samples. From a practical point of view, it is smart (small sample volume), rapid (two hours), easy (no specific expertise required) and requirements are widely available in clinical laboratories.
Collapse
Affiliation(s)
- Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Stefania Trino
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Marco Girasole
- Institute for the Study of the Structure of Matter, National Research Council (CNR), Rome, Italy
| | - Simone Dinarelli
- Institute for the Study of the Structure of Matter, National Research Council (CNR), Rome, Italy
| | - Angelo De Stradis
- Institute for Sustainable Plant Protection, National Research Council (CNR), Bari, Italy
| | - Vitina Grieco
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Maddalena Maietti
- Unit of Clinical Pathology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Antonio Traficante
- Unit of Clinical Pathology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Teodora Statuto
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Oreste Villani
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Pellegrino Musto
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Alessandro Sgambato
- Scientific Direction, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Luciana De Luca
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Antonella Caivano
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| |
Collapse
|
11
|
Human Plasma Extracellular Vesicle Isolation and Proteomic Characterization for the Optimization of Liquid Biopsy in Multiple Myeloma. Methods Mol Biol 2021; 2261:151-191. [PMID: 33420989 DOI: 10.1007/978-1-0716-1186-9_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer cells secrete membranous extracellular vesicles (EVs) which contain specific oncogenic molecular cargo (including oncoproteins, oncopeptides, and RNA) into their microenvironment and the circulation. As such, EVs including exosomes (small EVs) and microvesicles (large EVs) represent important circulating biomarkers for various diseases, including cancer and its progression. These circulating biomarkers offer a potentially minimally invasive and repeatable targets for analysis (liquid biopsy) that could aid in the diagnosis, risk stratification, and monitoring of cancer. Although their potential as cancer biomarkers has been promising, the identification and quantification of EVs in clinical samples remain challenging. Like EVs, other types of circulating biomarkers (including cell-free nucleic acids, cf-NAs; or circulating tumor cells, CTCs) may represent a complementary or alternative approach to cancer diagnosis. In the context of multiple myeloma (MM), a systemic cancer type that causes cancer cells to accumulate in the bone marrow, the specific role for EVs as biomarkers for diagnosis and monitoring remains undefined. Tumor heterogeneity along with the various subtypes of MM (such as non-secretory MM) that cannot be monitored using conventional testing (e.g. sequential serological testing and bone marrow biopsies) render liquid biopsy and circulating tumor-derived EVs a promising approach. In this protocol, we describe the isolation and purification of EVs from peripheral blood plasma (PBPL) collected from healthy donors and patients with MM for a biomarker discovery strategy. Our results demonstrate detection of circulating EVs from as little as 1 mL of MM patients' PBPL. High-resolution mass spectrometry (MS)-based proteomics promises to provide new avenues in identifying novel markers for detection, monitoring, and therapeutic intervention of disease. We describe biophysical characterization and quantitative proteomic profiling of disease-specific circulating EVs which may provide important implications for the development of cancer diagnostics in MM.
Collapse
|
12
|
Reale A, Carmichael I, Xu R, Mithraprabhu S, Khong T, Chen M, Fang H, Savvidou I, Ramachandran M, Bingham N, Simpson RJ, Greening DW, Spencer A. Human myeloma cell- and plasma-derived extracellular vesicles contribute to functional regulation of stromal cells. Proteomics 2021; 21:e2000119. [PMID: 33580572 DOI: 10.1002/pmic.202000119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Circulating small extracellular vesicles (sEV) represent promising non-invasive biomarkers that may aid in the diagnosis and risk-stratification of multiple myeloma (MM), an incurable blood cancer. Here, we comprehensively isolated and characterized sEV from human MM cell lines (HMCL) and patient-derived plasma (psEV) by specific EV-marker enrichment and morphology. Importantly, we demonstrate that HMCL-sEV are readily internalised by stromal cells to functionally modulate proliferation. psEV were isolated using various commercial approaches and pre-analytical conditions (collection tube types, storage conditions) assessed for sEV yield and marker enrichment. Functionally, MM-psEV was shown to regulate stromal cell proliferation and migration. In turn, pre-educated stromal cells favour HMCL adhesion. psEV isolated from patients with both pre-malignant plasma cell disorders (monoclonal gammopathy of undetermined significance [MGUS]; smouldering MM [SMM]) and MM have a similar ability to promote cell migration and adhesion, suggesting a role for both malignant and pre-malignant sEV in disease progression. Proteomic profiling of MM-psEV (305 proteins) revealed enrichment of oncogenic factors implicated in cell migration and adhesion, in comparison to non-disease psEV. This study describes a protocol to generate morphologically-intact and biologically functional sEV capable of mediating the regulation of stromal cells, and a model for the characterization of tumour-stromal cross-talk by sEV in MM.
Collapse
Affiliation(s)
- Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Irena Carmichael
- Monash Micro Imaging-AMREP, Monash University, Melbourne, Victoria, Australia
| | - Rong Xu
- Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Maoshan Chen
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Ioanna Savvidou
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Malarmathy Ramachandran
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Nicholas Bingham
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - David W Greening
- Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.,Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Monash University/Alfred Health, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, and Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Trino S, Lamorte D, Caivano A, De Luca L, Sgambato A, Laurenzana I. Clinical relevance of extracellular vesicles in hematological neoplasms: from liquid biopsy to cell biopsy. Leukemia 2021; 35:661-678. [PMID: 33299143 PMCID: PMC7932927 DOI: 10.1038/s41375-020-01104-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/30/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
In the era of precision medicine, liquid biopsy is becoming increasingly important in oncology. It consists in the isolation and analysis of tumor-derived biomarkers, including extracellular vesicles (EVs), in body fluids. EVs are lipid bilayer-enclosed particles, heterogeneous in size and molecular composition, released from both normal and neoplastic cells. In tumor context, EVs are valuable carriers of cancer information; in fact, their amount, phenotype and molecular cargo, including proteins, lipids, metabolites and nucleic acids, mirror nature and origin of parental cells rendering EVs appealing candidates as novel biomarkers. Translation of these new potential diagnostic tools into clinical practice could deeply revolutionize the cancer field mainly for solid tumors but for hematological neoplasms, too.
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy.
| | - Antonella Caivano
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Luciana De Luca
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Alessandro Sgambato
- Scientific Direction, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, PZ, Italy.
| |
Collapse
|
14
|
Recent Advancement and Technical Challenges in Developing Small Extracellular Vesicles for Cancer Drug Delivery. Pharm Res 2021; 38:179-197. [PMID: 33604783 DOI: 10.1007/s11095-021-02988-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer membrane-enclosed vesicles and act like 'messages in a bottle' in cell-cell communication by transporting their cargoes to recipient cells. Small EVs (sEVs, < 200 nm) are highly researched recently and have been harnessed as novel delivery systems for the treatment of various diseases, including neurodegenerative disorders, cardiovascular diseases, and most importantly cancer primarily because of their non-immunogenicity, tissue penetration and cell-tropism. This review will first provide a comprehensive overview of sEVs regarding the current understanding on their properties, biogenesis, new classification by the ISEV, composition, as well as their roles in cancer development (thereby called "oncosomes"). The primary focus will be given to the current state of sEVs as natural nanocarriers for cancer drug delivery, the technologies and challenges involved in sEV isolation and characterization, therapeutic cargo loading, and surface modification to enhance tumor-targeting. We will also provide examples of sEV products under clinical trials. Furthermore, the current challenges as well as the advance in "sEV mimetics" to address some of the sEVs limitations is briefly discussed. We seek to advance our understanding of sEVs to unlock their full potential as superior drug delivery vehicles in cancer therapy.
Collapse
|
15
|
Zhang PL, Liu ML. Extracellular vesicles mediate cellular interactions in renal diseases-Novel views of intercellular communications in the kidney. J Cell Physiol 2021; 236:5482-5494. [PMID: 33432614 DOI: 10.1002/jcp.30268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/23/2020] [Accepted: 12/26/2020] [Indexed: 12/28/2022]
Abstract
The kidney is a complicated and important internal organ receiving approximately 20% of the cardiac output and mediates numerous pathophysiologic actions. These include selectively filtering macromolecules of the blood, exquisite reclaimation of electrolyctes, urine concentration via an elegant osmotic mechanism, and excretion of an acid load. In addition, the renal tubules carry out secretory functions and produce hormones and cytokines. The kidney receives innervation and hormonal regulation. Therefore, dysfunction of the kidney leads to retention of metabolic waste products, and/or significant proteinuria and hematuria. In the past several decades, the role of extracellular vesicles (EVs) in intercellular communications, and the uptake of EVs by recipient cells through phagocytosis and endocytosis have been elucidated. The new knowledge on EVs expands over the classical mechanisms of cellular interaction, and may change our way of thinking of renal pathophysiology in the subcellular scale. Based on some ultrastructural discoveries in the kidney, this review will focus on the role of EVs in intercellular communications, their internalization by recipient cells, and their relationship to renal pathology.
Collapse
Affiliation(s)
- Ping L Zhang
- Division of Anatomic Pathology, Beaumont Laboratories, Beaumont Health, Royal Oak, Michigan, USA
| | - Ming-Lin Liu
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Gholipour E, Sarvarian P, Samadi P, Talebi M, Movassaghpour A, Motavalli R, Hojjat-Farsangi M, Yousefi M. Exosome: From leukemia progression to a novel therapeutic approach in leukemia treatment. Biofactors 2020; 46:698-715. [PMID: 32797698 DOI: 10.1002/biof.1669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Exosomes, as small vesicles, are released by tumor cells and tumor microenvironment (cells and function as key intercellular mediators and effects on different processes including tumorigenesis, angiogenesis, drug resistance, and evasion from immune system. These functions are due to exosomes' biomolecules which make them as efficient markers in early diagnosis of the disease. Also, exosomes have been recently applied in vaccination. The potential role of exosomes in immune response toward leukemic cells makes them efficient immunotherapeutic agents treating leukemia. Furthermore, variations in exosomes contents make them beneficial to be used in treating different diseases. This review introduces the role of exosomes in the development of hematological malignancies and evaluates their functional role in the treatment of these malignancies.
Collapse
Affiliation(s)
- Elham Gholipour
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Sarvarian
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Samadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roza Motavalli
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Immune and Gene Therapy Lab, Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Aging Research Institute, Tabriz university of Medical Sciences, Tabriz, Iran
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Emerging Insights on the Biological Impact of Extracellular Vesicle-Associated ncRNAs in Multiple Myeloma. Noncoding RNA 2020; 6:ncrna6030030. [PMID: 32764460 PMCID: PMC7549345 DOI: 10.3390/ncrna6030030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence indicates that extracellular vesicles (EVs) released from both tumor cells and the cells of the bone marrow microenvironment contribute to the pathobiology of multiple myeloma (MM). Recent studies on the mechanisms by which EVs exert their biological activity have indicated that the non-coding RNA (ncRNA) cargo is key in mediating their effect on MM development and progression. In this review, we will first discuss the role of EV-associated ncRNAs in different aspects of MM pathobiology, including proliferation, angiogenesis, bone disease development, and drug resistance. Finally, since ncRNAs carried by MM vesicles have also emerged as a promising tool for early diagnosis and therapy response prediction, we will report evidence of their potential use as clinical biomarkers.
Collapse
|
18
|
Pinto V, Bergantim R, Caires HR, Seca H, Guimarães JE, Vasconcelos MH. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers (Basel) 2020; 12:E407. [PMID: 32050631 PMCID: PMC7072128 DOI: 10.3390/cancers12020407] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is the second most common blood cancer. Treatments for MM include corticosteroids, alkylating agents, anthracyclines, proteasome inhibitors, immunomodulatory drugs, histone deacetylase inhibitors and monoclonal antibodies. Survival outcomes have improved substantially due to the introduction of many of these drugs allied with their rational use. Nonetheless, MM patients successively relapse after one or more treatment regimens or become refractory, mostly due to drug resistance. This review focuses on the main drugs used in MM treatment and on causes of drug resistance, including cytogenetic, genetic and epigenetic alterations, abnormal drug transport and metabolism, dysregulation of apoptosis, autophagy activation and other intracellular signaling pathways, the presence of cancer stem cells, and the tumor microenvironment. Furthermore, we highlight the areas that need to be further clarified in an attempt to identify novel therapeutic targets to counteract drug resistance in MM patients.
Collapse
Affiliation(s)
- Vanessa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- FCTUC–Faculty of Science and Technology of the University of Coimbra, 3030-790 Coimbra, Portugal
| | - Rui Bergantim
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hugo R. Caires
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Hugo Seca
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - M. Helena Vasconcelos
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP-Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
19
|
Faict S, Oudaert I, D’Auria L, Dehairs J, Maes K, Vlummens P, De Veirman K, De Bruyne E, Fostier K, Vande Broek I, Schots R, Vanderkerken K, Swinnen JV, Menu E. The Transfer of Sphingomyelinase Contributes to Drug Resistance in Multiple Myeloma. Cancers (Basel) 2019; 11:cancers11121823. [PMID: 31756922 PMCID: PMC6966559 DOI: 10.3390/cancers11121823] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/12/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma (MM) is well-known for the development of drug resistance, leading to relapse. Therefore, finding novel treatment strategies remains necessary. By performing a lipidomics assay on MM patient plasma, we aimed to identify new targets. We observed a dysregulation in the sphingolipid metabolism, with the upregulation of several ceramides and downregulation of sphingomyelin. This imbalance suggests an increase in sphingomyelinase, the enzyme responsible for hydrolyzing sphingomyelin into ceramide. We confirmed the upregulation of acid sphingomyelinase (ASM) in primary MM cells. Furthermore, we observed an increase in ASM expression in MM cell lines treated with melphalan or bortezomib, as well as in their exosomes. Exosomes high in ASM content were able to transfer the drug-resistant phenotype to chemosensitive cells, hereby suggesting a tumor-protective role for ASM. Finally, inhibition of ASM by amitriptyline improved drug sensitivity in MM cell lines and primary MM cells. In summary, this study is the first to analyze differences in plasma lipid composition of MM patients and match the observed differences to an upregulation of ASM. Moreover, we demonstrate that amitriptyline is able to inhibit ASM and increase sensitivity to anti-myeloma drugs. This study, therefore, provides a rational to include ASM-targeting-drugs in combination strategies in myeloma patients.
Collapse
Affiliation(s)
- Sylvia Faict
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Inge Oudaert
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Ludovic D’Auria
- Neurochemistry Unit, Institute of Neuroscience, Université Catholique de Louvain, B-1200 Brussels, Belgium;
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, LKI-Leuven Cancer Institute, KU Leuven, B-3000 Leuven, Belgium; (J.D.); (J.V.S.)
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Philip Vlummens
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
- Department of Clinical Hematology, Universitair Ziekenhuis Gent, B-9000 Ghent, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Karel Fostier
- Department of Clinical Hematology, Onze-Lieve-Vrouwziekenhuis Aalst, B-9300 Aalst, Belgium;
| | - Isabelle Vande Broek
- Department of Clinical Hematology, Algemeen Ziekenhuis Nikolaas, B-9100 Sint-Niklaas, Belgium;
| | - Rik Schots
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, LKI-Leuven Cancer Institute, KU Leuven, B-3000 Leuven, Belgium; (J.D.); (J.V.S.)
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, UZ Brussel, B-1090 Brussels, Belgium; (S.F.); (I.O.); (K.M.); (P.V.); (K.D.V.); (E.D.B.); (R.S.); (K.V.)
- Correspondence:
| |
Collapse
|
20
|
Valenzuela Alvarez M, Gutierrez LM, Correa A, Lazarowski A, Bolontrade MF. Metastatic Niches and the Modulatory Contribution of Mesenchymal Stem Cells and Its Exosomes. Int J Mol Sci 2019; 20:E1946. [PMID: 31010037 PMCID: PMC6515194 DOI: 10.3390/ijms20081946] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent an interesting population due to their capacity to release a variety of cytokines, chemokines, and growth factors, and due to their motile nature and homing ability. MSCs can be isolated from different sources, like adipose tissue or bone marrow, and have the capacity to differentiate, both in vivo and in vitro, into adipocytes, chondrocytes, and osteoblasts, making them even more interesting in the regenerative medicine field. Tumor associated stroma has been recognized as a key element in tumor progression, necessary for the biological success of the tumor, and MSCs represent a functionally fundamental part of this associated stroma. Exosomes represent one of the dominant signaling pathways within the tumor microenvironment. Their biology raises high interest, with implications in different biological processes involved in cancer progression, such as the formation of the pre-metastatic niche. This is critical during the metastatic cascade, given that it is the formation of a permissive context that would allow metastatic tumor cells survival within the new environment. In this context, we explored the role of exosomes, particularly MSCs-derived exosomes as direct or indirect modulators. All this points out a possible new tool useful for designing better treatment and detection strategies for metastatic progression, including the management of chemoresistance.
Collapse
Affiliation(s)
- Matias Valenzuela Alvarez
- Remodelative Processes and Cellular Niches Laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano Buenos Aires (HIBA), C1199ACL Buenos Aires, Argentina.
| | - Luciana M Gutierrez
- Remodelative Processes and Cellular Niches Laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano Buenos Aires (HIBA), C1199ACL Buenos Aires, Argentina.
| | | | - Alberto Lazarowski
- INFIBIOC, Clinical Biochemistry Department, School of Pharmacy and Biochemistry (FFyB), University of Buenos Aires (UBA), C1113AAD Buenos Aires, Argentina.
| | - Marcela F Bolontrade
- Remodelative Processes and Cellular Niches Laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano Buenos Aires (HIBA), C1199ACL Buenos Aires, Argentina.
| |
Collapse
|