1
|
Gonda L, Torner B, Ghansah H, Beke Debreceni I, Váróczy L, Pénzes-Daku K, Kappelmayer J. Monoclonal whole IgG impairs both fibrin and thrombin formation: hemostasis and surface plasmon resonance studies. Clin Chem Lab Med 2024; 62:1863-1869. [PMID: 38912717 DOI: 10.1515/cclm-2024-0252] [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: 02/26/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024]
Abstract
OBJECTIVES Monoclonal gammopathies frequently associate with hemostatic alterations. Thrombotic events occur with high incidence particularly upon treatment, while in rarer cases hemorrhagic diathesis can be observed. The pathology of these tendencies could be caused by thrombocytopenia or hyperviscosity burden of circulating monoclonal antibodies. Studies also suggest interference of monoclonal antibodies with primary hemostasis. We isolated monoclonal whole IgG paraproteins from two myeloma patients to observe their effect on thrombin formation and fibrin polymerization. METHODS Monoclonal whole IgG was prepared from sera of two newly diagnosed untreated multiple myeloma patients and control normal plasma samples. Fibrin formation was measured using thrombin time and dilute prothrombin time tests and thrombin formation was detected with a fluorimetric thrombin generation assay. In addition, molecular interactions were investigated by surface plasmon resonance (SPR). RESULTS Thrombin time was prolonged upon addition of monoclonal IgG even at 30 g/L by 12 %, increasing up to 36 % at 60 g/L concentration. Dilute prothrombin time was prolonged by 20 % even at 30 g/L. Thrombin generation assay indicated an impairment in thrombin formation at the presence of monoclonal IgG compared to polyclonal at equivalent concentration. By an SPR assay we determined that both clonality IgG preparations interacted with fibrinogen, however interaction with human thrombin was only detected with monoclonal immunoglobulins (KD=1.03 × 10-7 M). CONCLUSIONS Here we provide evidence that isolated monoclonal whole IgG from myeloma patients can impair both fibrin and thrombin formation and we demonstrate by SPR assay that it interacts with components of the final phase of the coagulation system.
Collapse
Affiliation(s)
- Lénárd Gonda
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Bernadett Torner
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Harriet Ghansah
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Váróczy
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Krisztina Pénzes-Daku
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Kappelmayer
- Department of Laboratory Medicine, Department of Hematology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Kulkarni A, Bazou D, Santos-Martinez MJ. Bleeding and Thrombosis in Multiple Myeloma: Platelets as Key Players during Cell Interactions and Potential Use as Drug Delivery Systems. Int J Mol Sci 2023; 24:15855. [PMID: 37958838 PMCID: PMC10647631 DOI: 10.3390/ijms242115855] [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: 09/27/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy originated in the bone marrow and characterized by unhindered plasma cell proliferation that results in several clinical manifestations. Although the main role of blood platelets lies in hemostasis and thrombosis, platelets also play a pivotal role in a number of other pathological conditions. Platelets are the less-explored components from the tumor microenvironment in MM. Although some studies have recently revealed that MM cells have the ability to activate platelets even in the premalignant stage, this phenomenon has not been widely investigated in MM. Moreover, thrombocytopenia, along with bleeding, is commonly observed in those patients. In this review, we discuss the hemostatic disturbances observed in MM patients and the dynamic interaction between platelets and myeloma cells, along with present and future potential avenues for the use of platelets for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Anushka Kulkarni
- The School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, The University of Dublin, D02 PN40 Dublin, Ireland;
| | - Despina Bazou
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Maria José Santos-Martinez
- The School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, The University of Dublin, D02 PN40 Dublin, Ireland;
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
3
|
Wright A, Snyder OL, He H, Christenson LK, Fleming S, Weiss ML. Procoagulant Activity of Umbilical Cord-Derived Mesenchymal Stromal Cells' Extracellular Vesicles (MSC-EVs). Int J Mol Sci 2023; 24:ijms24119216. [PMID: 37298168 DOI: 10.3390/ijms24119216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Many cell types, including cancer cells, release tissue factor (TF)-exposing extracellular vesicles (EVs). It is unknown whether MSC-EVs pose a thromboembolism risk due to TF expression. Knowing that MSCs express TF and are procoagulant, we hypothesize that MSC-EVs also might. Here, we examined the expression of TF and the procoagulant activity of MSC-EVs and the impact of EV isolation methods and cell culture expansion on EV yield, characterization, and potential risk using a design of experiments methodology. MSC-EVs were found to express TF and have procoagulant activity. Thus, when MSC-derived EVs are employed as a therapeutic agent, one might consider TF, procoagulant activity, and thromboembolism risk and take steps to prevent them.
Collapse
Affiliation(s)
- Adrienne Wright
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
- Midwest Institute of Comparative Stem Cell Biotechnology, Kansas State University, Manhattan, KS 66506, USA
| | - Orman Larry Snyder
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
- Midwest Institute of Comparative Stem Cell Biotechnology, Kansas State University, Manhattan, KS 66506, USA
| | - Hong He
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
- Midwest Institute of Comparative Stem Cell Biotechnology, Kansas State University, Manhattan, KS 66506, USA
| | - Lane K Christenson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sherry Fleming
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Mark L Weiss
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, USA
- Midwest Institute of Comparative Stem Cell Biotechnology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
4
|
Vulpis E, Cuollo L, Borrelli C, Antonangeli F, Masuelli L, Cippitelli M, Fionda C, Caracciolo G, Petrucci MT, Santoni A, Zingoni A, Soriani A. Doxorubicin–Mediated miR–433 Expression on Exosomes Promotes Bystander Senescence in Multiple Myeloma Cells in a DDR–Independent Manner. Int J Mol Sci 2023; 24:ijms24076862. [PMID: 37047835 PMCID: PMC10095495 DOI: 10.3390/ijms24076862] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Abstract
The success of senescence-based anticancer therapies relies on their anti-proliferative power and on their ability to trigger anti-tumor immune responses. Indeed, genotoxic drug-induced senescence increases the expression of NK cell-activating ligands on multiple myeloma (MM) cells, boosting NK cell recognition and effector functions. Senescent cells undergo morphological change and context-dependent functional diversification, acquiring the ability to secrete a vast pool of molecules termed the senescence-associated secretory phenotype (SASP), which affects neighboring cells. Recently, exosomes have been recognized as SASP factors, contributing to modulating a variety of cell functions. In particular, evidence suggests a key role for exosomal microRNAs in influencing many hallmarks of cancer. Herein, we demonstrate that doxorubicin treatment of MM cells leads to the enrichment of miR-433 into exosomes, which in turn induces bystander senescence. Our analysis reveals that the establishment of the senescent phenotype on neighboring MM cells is p53- and p21-independent and is related to CDK-6 down-regulation. Notably, miR-433-dependent senescence does not induce the up-regulation of activating ligands on MM cells. Altogether, our findings highlight the possibility of miR-433-enriched exosomes to reinforce doxorubicin-mediated cellular senescence.
Collapse
Affiliation(s)
- Elisabetta Vulpis
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Lorenzo Cuollo
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Cristiana Borrelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Fabrizio Antonangeli
- Institute of Molecular Biology and Pathology, National Research Council (CNR), 00161 Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Teresa Petrucci
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, 00161 Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
5
|
Iskrzak J, Zygmunciak P, Misiewicz-Krzemińska I, Puła B. Extracellular Vesicles in Multiple Myeloma-Cracking the Code to a Better Understanding of the Disease. Cancers (Basel) 2022; 14:cancers14225575. [PMID: 36428668 PMCID: PMC9688731 DOI: 10.3390/cancers14225575] [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: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell-derived malignancy that stands for around 1.5% of newly discovered cancer cases. Despite constantly improving treatment methods, the disease is incurable with over 13,000 deaths in the US and over 30,000 in Europe. Recent studies suggest that extracellular vesicles (EVs) might play a significant role in the pathogenesis and evolution of MM. Further investigation of their role could prove to be beneficial in establishing new therapies and hence, improve the prognosis of MM patients. What is more, EVs might serve as novel markers in diagnosing and monitoring the disease. Great advancements concerning the position of EVs in the pathophysiology of MM have recently been shown in research and in this review, we would like to delve into the still expanding state of knowledge.
Collapse
Affiliation(s)
- Justyna Iskrzak
- Medical University of Warsaw, 02-091 Warsaw, Poland
- Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Przemysław Zygmunciak
- Medical University of Warsaw, 02-091 Warsaw, Poland
- Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
| | - Irena Misiewicz-Krzemińska
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Chocimska Str. 5, 00-791 Warsaw, Poland
| | - Bartosz Puła
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indira Gandhi Str. 14, 02-776 Warsaw, Poland
- Correspondence: ; Tel.: +48-223-496-302; Fax: +48-223-496-335
| |
Collapse
|
6
|
Rezaie J, Ahmadi M, Ravanbakhsh R, Mojarad B, Mahbubfam S, Shaban SA, Shadi K, Berenjabad NJ, Etemadi T. Tumor-derived extracellular vesicles: The metastatic organotropism drivers. Life Sci 2022; 289:120216. [PMID: 34890589 DOI: 10.1016/j.lfs.2021.120216] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
The continuous growing, spreading, and metastasis of tumor cells depend on intercellular communication within cells resident in a tissue environment. Such communication is mediated through the secretion of particles from tumor cells and resident cells known as extracellular vesicles (EVs) within a microenvironment. EVs are a heterogeneous population of membranous vesicles released from tumor cells that transfer many types of active biomolecules to recipient cells and induce physiologic and phenotypic alterations in the tissue environment. Spreading the 'seeds' of metastasis needs the EVs that qualify the 'soil' at distant sites to promote the progress of arriving tumor cells. Growing evidence indicates that EVs have vital roles in tumorigenesis, including pre-metastatic niche formation and organotropic metastasis. These EVs mediate organotropic metastasis by modifying the pre-metastatic microenvironment through different pathways including induction of phenotypic alternation and differentiation of cells, enrolment of distinct supportive stromal cells, up-regulation of the expression of pro-inflammatory genes, and induction of immunosuppressive status. However, instead of pre-metastatic niche formation, evidence suggests that EVs may mediate reawakening of dormant niches. Findings regarding EVs function in tumor metastasis have led to growing interests in the interdisciplinary significance of EVs, including targeted therapy, cell-free therapy, drug-delivery system, and diagnostic biomarker. In this review, we discuss EVs-mediated pre-metastatic niche formation and organotropic metastasis in visceral such as lung, liver, brain, lymph node, and bone with a focus on associated signaling, causing visceral environment hospitable for metastatic cells. Furthermore, we present an overview of the possible therapeutic application of EVs in cancer management.
Collapse
Affiliation(s)
- Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahdi Ahmadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Ravanbakhsh
- Department of Aquatic Biotechnology, Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran
| | - Behnam Mojarad
- Biology Department, Faculty of Sciences, Urmia University, Urmia, Iran
| | - Shadi Mahbubfam
- Biology Department, Faculty of Sciences, Urmia University, Urmia, Iran
| | | | - Kosar Shadi
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Tahereh Etemadi
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| |
Collapse
|
7
|
Immune Checkpoint Inhibitor Therapy for Bone Metastases: Specific Microenvironment and Current Situation. J Immunol Res 2021; 2021:8970173. [PMID: 34877360 PMCID: PMC8645368 DOI: 10.1155/2021/8970173] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/19/2021] [Accepted: 11/03/2021] [Indexed: 12/20/2022] Open
Abstract
The treatment of bone metastases is a thorny issue. Immunotherapy may be one of the few hopes for patients with unresectable bone metastases. Immune checkpoint inhibitors are the most commonly used immunotherapy drugs currently. In this review, the characteristics and interaction of bone metastases and their immune microenvironment were systematically discussed, and the relevant research progress of the immunological mechanism of tumor bone metastasis was reviewed. On this basis, we expounded the clinical application of immune checkpoint inhibitors for bone metastasis of common tumors, including non-small-cell lung cancer, renal cell carcinoma, prostate cancer, melanoma, and breast cancer. Then, the deficiencies and limitations in current researches were summarized. In-depth basic research on bone metastases and optimization of clinical treatment is needed.
Collapse
|
8
|
Moreira R, Mendonça LS, Pereira de Almeida L. Extracellular Vesicles Physiological Role and the Particular Case of Disease-Spreading Mechanisms in Polyglutamine Diseases. Int J Mol Sci 2021; 22:ijms222212288. [PMID: 34830171 PMCID: PMC8621536 DOI: 10.3390/ijms222212288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Recent research demonstrated pathological spreading of the disease-causing proteins from one focal point across other brain regions for some neurodegenerative diseases, such as Parkinson's and Alzheimer's disease. Spreading mediated by extracellular vesicles is one of the proposed disease-spreading mechanisms. Extracellular vesicles are cell membrane-derived vesicles, used by cells for cell-to-cell communication and excretion of toxic components. Importantly, extracellular vesicles carrying pathological molecules, when internalized by "healthy" cells, may trigger pathological pathways and, consequently, promote disease spreading to neighboring cells. Polyglutamine diseases are a group of genetic neurodegenerative disorders characterized by the accumulation of mutant misfolded proteins carrying an expanded tract of glutamines, including Huntington's and Machado-Joseph disease. The pathological spread of the misfolded proteins or the corresponding mutant mRNA has been explored. The understanding of the disease-spreading mechanism that plays a key role in the pathology progression of these diseases can result in the development of effective therapeutic approaches to stop disease progression, arresting the spread of the toxic components and disease aggravation. Therefore, the present review's main focus is the disease-spreading mechanisms with emphasis on polyglutamine diseases and the putative role played by extracellular vesicles in this process.
Collapse
Affiliation(s)
- Ricardo Moreira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana S. Mendonça
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Correspondence: (L.S.M.); (L.P.d.A.); Tel.: +351-239-820-190 (L.S.M.)
| | - Luís Pereira de Almeida
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Correspondence: (L.S.M.); (L.P.d.A.); Tel.: +351-239-820-190 (L.S.M.)
| |
Collapse
|
9
|
Burgos-Ravanal R, Campos A, Díaz-Vesga MC, González MF, León D, Lobos-González L, Leyton L, Kogan MJ, Quest AFG. Extracellular Vesicles as Mediators of Cancer Disease and as Nanosystems in Theranostic Applications. Cancers (Basel) 2021; 13:3324. [PMID: 34283059 PMCID: PMC8268753 DOI: 10.3390/cancers13133324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer remains a leading cause of death worldwide despite decades of intense efforts to understand the molecular underpinnings of the disease. To date, much of the focus in research has been on the cancer cells themselves and how they acquire specific traits during disease development and progression. However, these cells are known to secrete large numbers of extracellular vesicles (EVs), which are now becoming recognized as key players in cancer. EVs contain a large number of different molecules, including but not limited to proteins, mRNAs, and miRNAs, and they are actively secreted by many different cell types. In the last two decades, a considerable body of evidence has become available indicating that EVs play a very active role in cell communication. Cancer cells are heterogeneous, and recent evidence reveals that cancer cell-derived EV cargos can change the behavior of target cells. For instance, more aggressive cancer cells can transfer their "traits" to less aggressive cancer cells and convert them into more malignant tumor cells or, alternatively, eliminate those cells in a process referred to as "cell competition". This review discusses how EVs participate in the multistep acquisition of specific traits developed by tumor cells, which are referred to as "the hallmarks of cancer" defined by Hanahan and Weinberg. Moreover, as will be discussed, EVs play an important role in drug resistance, and these more recent advances may explain, at least in part, why pharmacological therapies are often ineffective. Finally, we discuss literature proposing the use of EVs for therapeutic and prognostic purposes in cancer.
Collapse
Affiliation(s)
- Renato Burgos-Ravanal
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| | - América Campos
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane 4029, Australia
| | - Magda C. Díaz-Vesga
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Grupo de Investigación en Ciencias Básicas y Clínicas de la Salud, Pontificia Universidad Javeriana de Cali, Cali 760008, Colombia
| | - María Fernanda González
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| | - Daniela León
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Lorena Lobos-González
- Centro de Medicina Regenerativa, Facultad de Medicina, Universidad del Desarrollo-Clínica Alemana, Santiago 7590943, Chile;
| | - Lisette Leyton
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| | - Marcelo J. Kogan
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Andrew F. G. Quest
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| |
Collapse
|
10
|
Urabe F, Patil K, Ramm GA, Ochiya T, Soekmadji C. Extracellular vesicles in the development of organ-specific metastasis. J Extracell Vesicles 2021; 10:e12125. [PMID: 34295457 PMCID: PMC8287318 DOI: 10.1002/jev2.12125] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Distant organ metastasis, often termed as organotropic metastasis or metastatic organotropism, is a fundamental feature of malignant tumours and accounts for most cancer-related mortalities. This process is orchestrated by many complex biological interactions and processes that are mediated by a combination of anatomical, genetic, pathophysiological and biochemical factors. Recently, extracellular vesicles (EVs) are increasingly being demonstrated as critical mediators of bi-directional tumour-host cell interactions, controlling organ-specific infiltration, adaptation and colonization at the secondary site. EVs govern organotropic metastasis by modulating the pre-metastatic microenvironment through upregulation of pro-inflammatory gene expression and immunosuppressive cytokine secretion, induction of phenotype-specific differentiation and recruitment of specific stromal cell types. This review discusses EV-mediated metastatic organotropism in visceral (brain, lung, liver, and lymph node) and skeletal (bone) metastasis, and discusses how the pre-metastatic education by EVs transforms the organ into a hospitable, tumour cell-friendly milieu that supports the growth of metastatic cells. Decoding the organ-specific traits of EVs and their functions in organotropic metastasis is essential in accelerating the clinical application of EVs in cancer management.
Collapse
Affiliation(s)
- Fumihiko Urabe
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
- Department of Molecular and Cellular MedicineTokyo Medical UniversityTokyoJapan
| | - Kalyani Patil
- Department of Molecular PathophysiologyTranslational Research InstituteAcademic Health SystemHamad Medical CorporationDohaQatar
| | - Grant A. Ramm
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineTokyo Medical UniversityTokyoJapan
| | - Carolina Soekmadji
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| |
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: 1.0] [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
|
Nielsen T, Kristensen SR, Gregersen H, Teodorescu EM, Pedersen S. Prothrombotic abnormalities in patients with multiple myeloma and monoclonal gammopathy of undetermined significance. Thromb Res 2021; 202:108-118. [PMID: 33819778 DOI: 10.1016/j.thromres.2021.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Multiple myeloma (MM) and its precursor condition, monoclonal gammopathy of undetermined significance (MGUS) have an increased risk of thrombotic events, especially during anti-myeloma treatment. Many different underlying causes for this hypercoagulability have been suggested, but current techniques to identify abnormalities in these patients are sparse and inefficient. The aim of this study was to assess the hypercoagulability in MGUS and MM patients through various coagulation analyses and identify changes in the MM patients throughout their treatment regimen. MATERIALS AND METHODS Platelet-free plasma from 38 MM patients, 19 MGUS patients and 34 healthy controls were tested for hypercoagulability using calibrated automated thrombogram, a procoagulant phospholipid assay, a microvesicle-associated (MV) tissue factor (TF) assay, and a cell-free deoxyribonucleic acid (cf-DNA) assay as a surrogate measurement for neutrophil extracellular traps (NETs). RESULTS MGUS and MM patients both had elevated thrombin generation and procoagulant phospholipid activity in comparison to the control subjects. MM, and partly MGUS, showed increased MV-TF activity, however, only MM had increased levels of the cf-DNA. CONCLUSIONS Here we demonstrated that hypercoagulability was present in patients with MGUS and MM through increased thrombin generation, possibly due to higher TF and procoagulant phospholipids (PPL) activity. This may be associated to MVs and, for MM patients, be attributed to procoagulant NETs activity; however, this remains to be determined.
Collapse
Affiliation(s)
- Thøger Nielsen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | - Søren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | | | | | - Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
13
|
Forte D, Barone M, Palandri F, Catani L. The "Vesicular Intelligence" Strategy of Blood Cancers. Genes (Basel) 2021; 12:genes12030416. [PMID: 33805807 PMCID: PMC7999060 DOI: 10.3390/genes12030416] [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] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Blood cancers are a heterogeneous group of disorders including leukemia, multiple myeloma, and lymphoma. They may derive from the clonal evolution of the hemopoietic stem cell compartment or from the transformation of progenitors with immune potential. Extracellular vesicles (EVs) are membrane-bound nanovesicles which are released by cells into body fluids with a role in intercellular communication in physiology and pathology, including cancer. EV cargos are enriched in nucleic acids, proteins, and lipids, and these molecules can be delivered to target cells to influence their biological properties and modify surrounding or distant targets. In this review, we will describe the “smart strategy” on how blood cancer-derived EVs modulate tumor cell development and maintenance. Moreover, we will also depict the function of microenvironment-derived EVs in blood cancers and discuss how the interplay between tumor and microenvironment affects blood cancer cell growth and spreading, immune response, angiogenesis, thrombogenicity, and drug resistance. The potential of EVs as non-invasive biomarkers will be also discussed. Lastly, we discuss the clinical application viewpoint of EVs in blood cancers. Overall, blood cancers apply a ‘vesicular intelligence’ strategy to spread signals over their microenvironment, promoting the development and/or maintenance of the malignant clone.
Collapse
Affiliation(s)
- Dorian Forte
- IRCCS Azienda Ospedaliero—Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (D.F.); (M.B.)
| | - Martina Barone
- IRCCS Azienda Ospedaliero—Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (D.F.); (M.B.)
| | - Francesca Palandri
- IRCCS Azienda Ospedaliero—Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy
- Correspondence: (F.P.); (L.C.); Tel.: +39-5121-43044 (F.P.); +39-5121-43837 (L.C.)
| | - Lucia Catani
- IRCCS Azienda Ospedaliero—Department of Experimental, Diagnostic and Specialty Medicine, School of Medicine, Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy; (D.F.); (M.B.)
- IRCCS Azienda Ospedaliero—Institute of Hematology “Seràgnoli”, University of Bologna, 40138 Bologna, Italy
- Correspondence: (F.P.); (L.C.); Tel.: +39-5121-43044 (F.P.); +39-5121-43837 (L.C.)
| |
Collapse
|
14
|
Contribution of extracellular vesicles in normal hematopoiesis and hematological malignancies. Heliyon 2021; 7:e06030. [PMID: 33521365 PMCID: PMC7820922 DOI: 10.1016/j.heliyon.2021.e06030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/05/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed microparticles that have prominent roles in the intercellular crosstalk. EVs are secreted after fusion of endosomes with the plasma membrane (exosomes) or shed from the plasma membrane (microvesicles). These microparticles modulate bone marrow microenvironment and alter differentiation and expansion of normal hematopoietic cells. EVs originated from mesenchymal stromal cells have been shown to enhance expansion of myeloid-biased hematopoietic progenitor cells. In addition, megakaryocytic microparticles stimulate differentiation of hematopoietic stem and progenitor cells into mature megakaryocytes. The ability of EVs in induction of maturation and expansion of certain hematopoietic cells has implications in transfusion medicine and in targeted therapeutic modalities. Important prerequisites for these interventions are identification the specific targets of EVs, transferred biomolecules and molecular mechanisms underlying the fate decision in the target cells. EVs are also involved in the pathogenesis and progression of hematological malignancies including acute leukemia and multiples myeloma. In the current review, we provide a summary of studies which evaluated the significance of EVs in normal hematopoiesis and hematological malignancies.
Collapse
|
15
|
Scholl JN, Dias CK, Muller L, Battastini AMO, Figueiró F. Extracellular vesicles in cancer progression: are they part of the problem or part of the solution? Nanomedicine (Lond) 2020; 15:2625-2641. [PMID: 33094653 DOI: 10.2217/nnm-2020-0256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are released especially by cancer cells. They modulate the tumor microenvironment by interacting with immune cells while carrying immunosuppressive or immunostimulatory molecules. In this review, we will explore some conflicting reports regarding the immunological outcomes of EVs in cancer progression, in which they might initiate an antitumor immune response or an immunosuppressive response. Concerning immunosuppression, the role of tumor-derived EVs' in the adenosinergic system is underexplored. The enhancement of adenosine (ADO) levels in the tumor microenvironment impairs T-cell function and cytokine release. However, some tumor-derived EVs may deliver immunostimulatory factors, promoting immunogenic activity, even with ADO production. The modulatory role of ADO over the tumor progression represents a piece in an intricate microenvironment with anti and pro tumoral seesaw-like mechanisms.
Collapse
Affiliation(s)
- Juliete Nathali Scholl
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Camila Kehl Dias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Laurent Muller
- Department of Otolaryngology, Head & Neck Surgery, University of Basel, Basel, 4031, Switzerland
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Fabrício Figueiró
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| |
Collapse
|
16
|
Ellegaard Nielsen J, Sofie Pedersen K, Vestergård K, Georgiana Maltesen R, Christiansen G, Lundbye-Christensen S, Moos T, Risom Kristensen S, Pedersen S. Novel Blood-Derived Extracellular Vesicle-Based Biomarkers in Alzheimer's Disease Identified by Proximity Extension Assay. Biomedicines 2020; 8:E199. [PMID: 32645971 PMCID: PMC7400538 DOI: 10.3390/biomedicines8070199] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022] Open
Abstract
Easily accessible biomarkers for Alzheimer's dementia (AD) are lacking and established clinical markers are limited in applicability. Blood is a common biofluid for biomarker discoveries, and extracellular vesicles (EVs) may provide a matrix for exploring AD related biomarkers. Thus, we investigated proteins related to neurological and inflammatory processes in plasma and EVs. By proximity extension assay (PEA), 182 proteins were measured in plasma and EVs from patients with AD (n = 10), Mild Cognitive Impairment (MCI, n = 10), and healthy controls (n = 10). Plasma-derived EVs were enriched by 20,000× g, 1 h, 4 °C, and confirmed using nanoparticle tracking analysis (NTA), western blotting, and transmission electron microscopy with immunolabelling (IEM). Presence of CD9+ EVs was confirmed by western blotting and IEM. No group differences in particle concentration or size were detected by NTA. However, significant protein profiles were observed among subjects, particularly for EVs. Several proteins and their ratios could distinguish cognitively affected from healthy individuals. For plasma TGF-α│CCL20 (AUC = 0.96, 95% CI = 0.88-1.00, p = 0.001) and EVs CLEC1B│CCL11 (AUC = 0.95, 95% CI = 0.86-1.00, p = 0.001) showed diagnostic capabilities. Using PEA, we identified protein profiles capable of distinguishing healthy controls from AD patients. EVs provided additional biological information related to AD not observed in plasma alone.
Collapse
Affiliation(s)
- Jonas Ellegaard Nielsen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | | | - Karsten Vestergård
- Department of Neurology, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
| | - Raluca Georgiana Maltesen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark;
- Department of Health Science and Technology, Aalborg University, DK-9220 Aalborg, Denmark;
| | - Søren Lundbye-Christensen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Unit of Clinical Biostatistics, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Torben Moos
- Department of Health Science and Technology, Aalborg University, DK-9220 Aalborg, Denmark;
| | - Søren Risom Kristensen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Shona Pedersen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| |
Collapse
|
17
|
Ferreira B, Caetano J, Barahona F, Lopes R, Carneiro E, Costa-Silva B, João C. Liquid biopsies for multiple myeloma in a time of precision medicine. J Mol Med (Berl) 2020; 98:513-525. [PMID: 32246161 PMCID: PMC7198642 DOI: 10.1007/s00109-020-01897-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a challenging, progressive, and highly heterogeneous hematological malignancy. MM is characterized by multifocal proliferation of neoplastic plasma cells in the bone marrow (BM) and sometimes in extramedullary organs. Despite the availability of novel drugs and the longer median overall survival, some patients survive more than 10 years while others die rapidly. This heterogeneity is mainly driven by biological characteristics of MM cells, including genetic abnormalities. Disease progressions are mainly due to the inability of drugs to overcome refractory disease and inevitable drug-resistant relapse. In clinical practice, a bone marrow biopsy, mostly performed in one site, is still used to access the genetics of MM. However, BM biopsy use is limited by its invasive nature and by often not accurately reflecting the mutational profile of MM. Recent insights into the genetic landscape of MM provide a valuable opportunity to implement precision medicine approaches aiming to enable better patient profiling and selection of targeted therapies. In this review, we explore the use of the emerging field of liquid biopsies in myeloma patients considering current unmet medical needs, such as assessing the dynamic mutational landscape of myeloma, early predictors of treatment response, and a less invasive response monitoring.
Collapse
Affiliation(s)
- Bruna Ferreira
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana Caetano
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Filipa Barahona
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Raquel Lopes
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Emilie Carneiro
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cristina João
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Nova Medical School, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
18
|
Morandi F, Airoldi I, Marimpietri D, Bracci C, Faini AC, Gramignoli R. CD38, a Receptor with Multifunctional Activities: From Modulatory Functions on Regulatory Cell Subsets and Extracellular Vesicles, to a Target for Therapeutic Strategies. Cells 2019; 8:E1527. [PMID: 31783629 PMCID: PMC6953043 DOI: 10.3390/cells8121527] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
CD38 is a multifunctional cell surface protein endowed with receptor/enzymatic functions. The protein is generally expressed at low/intermediate levels on hematological tissues and some solid tumors, scoring the highest levels on plasma cells (PC) and PC-derived neoplasia. CD38 was originally described as a receptor expressed by activated cells, mainly T lymphocytes, wherein it also regulates cell adhesion and cooperates in signal transduction mediated by major receptor complexes. Furthermore, CD38 metabolizes extracellular NAD+, generating ADPR and cyclic ADPR. This ecto-enzyme controls extra-cellular nucleotide homeostasis and intra-cellular calcium fluxes, stressing its relevance in multiple physiopathological conditions (infection, tumorigenesis and aging). In clinics, CD38 was adopted as a cell activation marker and in the diagnostic/staging of leukemias. Quantitative surface CD38 expression by multiple myeloma (MM) cells was the basic criterion used for therapeutic application of anti-CD38 monoclonal antibodies (mAbs). Anti-CD38 mAbs-mediated PC depletion in autoimmunity and organ transplants is currently under investigation. This review analyzes different aspects of CD38's role in regulatory cell populations and how these effects are obtained. Characterizing CD38 functional properties may widen the extension of therapeutic applications for anti-CD38 mAbs. The availability of therapeutic mAbs with different effects on CD38 enzymatic functions may be rapidly translated to immunotherapeutic strategies of cell immune defense.
Collapse
Affiliation(s)
- Fabio Morandi
- Laboratory of Stem Cell and Cell Therapy, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (I.A.); (D.M.)
| | - Irma Airoldi
- Laboratory of Stem Cell and Cell Therapy, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (I.A.); (D.M.)
| | - Danilo Marimpietri
- Laboratory of Stem Cell and Cell Therapy, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (I.A.); (D.M.)
| | - Cristiano Bracci
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (C.B.); (A.C.F.)
- CeRMS, University of Torino, 10126 Torino, Italy
| | - Angelo Corso Faini
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Torino, 10126 Torino, Italy; (C.B.); (A.C.F.)
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden;
| |
Collapse
|
19
|
Taus F, Meneguzzi A, Castelli M, Minuz P. Platelet-Derived Extracellular Vesicles as Target of Antiplatelet Agents. What Is the Evidence? Front Pharmacol 2019; 10:1256. [PMID: 31780927 PMCID: PMC6857039 DOI: 10.3389/fphar.2019.01256] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Platelet-derived large extracellular vesicles (often referred to as microparticles in the field of cardiovascular disease) have been identified as effector in the atherothrombotic process, therefore representing a target of pharmacological intervention of potential interest. Despite that, limited evidence is so far available concerning the effects of antiplatelet agents on the release of platelet-derived extracellular vesicles. In the present narrative review, the mechanisms leading to vesiculation in platelets and the pathophysiological processes implicated will be discussed. This will be followed by a summary of the present evidence concerning the effects of antiplatelet agents under experimental conditions and in clinical settings.
Collapse
Affiliation(s)
- Francesco Taus
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Alessandra Meneguzzi
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Marco Castelli
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Pietro Minuz
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| |
Collapse
|
20
|
Natoni A, Bohara R, Pandit A, O'Dwyer M. Targeted Approaches to Inhibit Sialylation of Multiple Myeloma in the Bone Marrow Microenvironment. Front Bioeng Biotechnol 2019; 7:252. [PMID: 31637237 PMCID: PMC6787837 DOI: 10.3389/fbioe.2019.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/17/2019] [Indexed: 11/13/2022] Open
Abstract
Aberrant glycosylation modulates different aspects of tumor biology, and it has long been recognized as a hallmark of cancer. Among the different forms of glycosylation, sialylation, the addition of sialic acid to underlying oligosaccharides, is often dysregulated in cancer. Increased expression of sialylated glycans has been observed in many types of cancer, including multiple myeloma, and often correlates with aggressive metastatic behavior. Myeloma, a cancer of plasma cells, develops in the bone marrow, and colonizes multiple sites of the skeleton including the skull. In myeloma, the bone marrow represents an essential niche where the malignant cells are nurtured by the microenvironment and protected from chemotherapy. Here, we discuss the role of hypersialylation in the metastatic process focusing on multiple myeloma. In particular, we examine how increased sialylation modulates homing of malignant plasma cells into the bone marrow by regulating the activity of molecules important in bone marrow cellular trafficking including selectins and integrins. We also propose that inhibiting sialylation may represent a new therapeutic strategy to overcome bone marrow-mediated chemotherapy resistance and describe different targeted approaches to specifically deliver sialylation inhibitors to the bone marrow microenvironment.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Raghvendra Bohara
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Michael O'Dwyer
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
21
|
Colombo M, Giannandrea D, Lesma E, Basile A, Chiaramonte R. Extracellular Vesicles Enhance Multiple Myeloma Metastatic Dissemination. Int J Mol Sci 2019; 20:ijms20133236. [PMID: 31266187 PMCID: PMC6650870 DOI: 10.3390/ijms20133236] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) represent a heterogeneous group of membranous structures shed by all kinds of cell types, which are released into the surrounding microenvironment or spread to distant sites through the circulation. Therefore, EVs are key mediators of the communication between tumor cells and the surrounding microenvironment or the distant premetastatic niche due to their ability to transport lipids, transcription factors, mRNAs, non-coding regulatory RNAs, and proteins. Multiple myeloma (MM) is a hematological neoplasm that mostly relies on the bone marrow (BM). The BM represents a highly supportive niche for myeloma establishment and diffusion during the formation of distant bone lesions typical of this disease. This review represents a survey of the most recent evidence published on the role played by EVs in supporting MM cells during the multiple steps of metastasis, including travel and uptake at distant premetastatic niches, MM cell engraftment as micrometastasis, and expansion to macrometastasis thanks to EV-induced angiogenesis, release of angiocrine factors, activation of osteolytic activity, and mesenchymal cell support. Finally, we illustrate the first evidence concerning the dual effect of MM-EVs in promoting both anti-tumor immunity and MM immune escape, and the possible modulation operated by pharmacological treatments.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, I-20142 Milano, Italy
| | - Domenica Giannandrea
- Department of Health Sciences, Università degli Studi di Milano, I-20142 Milano, Italy
| | - Elena Lesma
- Department of Health Sciences, Università degli Studi di Milano, I-20142 Milano, Italy
| | - Andrea Basile
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, I-20122 Milano, Italy
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, I-20142 Milano, Italy.
| |
Collapse
|