1
|
Gao M, Dong H, Jiang S, Chen F, Fu Y, Luo Y. Activated platelet-derived exosomal LRG1 promotes multiple myeloma cell growth. Oncogenesis 2024; 13:21. [PMID: 38871685 DOI: 10.1038/s41389-024-00522-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
The hypercoagulable state is a hallmark for patients with multiple myeloma (MM) and is associated with disease progression. Activated platelets secrete exosomes and promote solid tumor growth. However, the role of platelet-derived exosomes in MM is not fully clear. We aim to study the underlying mechanism of how platelet-derived exosomes promote MM cell growth. Flow cytometry, Western blot, proteome analysis, co-immunoprecipitation, immunofluorescence staining, and NOD/SCID mouse subcutaneous transplantation model were performed to investigate the role of exosomal LRG1 on multiple myeloma cell growth. Peripheral blood platelets in MM patients were in a highly activated state, and platelet-rich plasma from MM patients significantly promoted cell proliferation and decreased apoptotic cells in U266 and RPMI8226 cells. Leucine-rich-alpha-2-glycoprotein 1 (LRG1) was significantly enriched in MM platelet-derived exosomes. Blocking LRG1 in recipient cells using LRG1 antibody could significantly eliminate the proliferation-promoting effect of platelet-derived exosomes on MM cells. And high exosomal LRG1 was associated with poor prognosis of patients with MM. Mechanistic studies revealed that LRG1 interacted with Olfactomedin 4 (OLFM4) to accelerate MM progression by activating the epithelial-to-mesenchymal transition (EMT) signaling pathway and promoting angiogenesis. Our results revealed that blocking LRG1 is a promising therapeutic strategy for the treatment of MM.
Collapse
Affiliation(s)
- Meng Gao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hang Dong
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Siyi Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Fangping Chen
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China.
| |
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
|
Wang J, Shan A, Shi F, Zheng Q. Molecular and clinical characterization of ANG expression in gliomas and its association with tumor-related immune response. Front Med (Lausanne) 2023; 10:1044402. [PMID: 37928479 PMCID: PMC10621067 DOI: 10.3389/fmed.2023.1044402] [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: 09/14/2022] [Accepted: 10/10/2023] [Indexed: 11/07/2023] Open
Abstract
Background Angiogenin (ANG) has been widely reported as a crucial molecular regulator in multiple malignancies. However, its role in gliomagenesis remains unclear. This study aimed to investigate the molecular and clinical characterization of ANG expression at transcriptome level and the association with glioma-related immune response. Methods A total of 301 glioma samples with mRNA microarray data (CGGA301) was obtained from the official website of CGGA project for yielding preliminary results, followed by validation in two independent RNAseq datasets, including TCGA with 697 samples and CGGA325 with 325 patients. Moreover, CGGA single-cell RNAseq (scRNAseq) data were analyzed to identify differential and dynamic ANG expression in different cells. Immunohistochemistry was performed to evaluate ANG protein expression across different WHO grades in a tissue microarray (TMA). Figure generation and statistical analysis were conducted using R software. Results ANG expression was associated with clinical features, malignant phenotypes, and genomic alterations. Based on significantly correlated genes of ANG, subsequent gene ontology (GO) and gene set enrichment analysis (GSEA) concordantly pointed to the significant association of ANG in immune-related biological processes. Moreover, ANG showed robust correlations with canonical immune checkpoint molecules, including PD1 signaling, CTLA4, TIM3, and B7H3. Gene sets variation analysis (GSVA) found that ANG was particularly associated with activities of macrophages and antigen presentation cells (APCs) in both LGG and GBM across different datasets. Furthermore, the higher-ANG milieu seemed to recruit monocyte-macrophage lineage and dendritic cells into the glioma microenvironment. According to scRNAseq analysis, ANG was mainly expressed by neoplastic cells and tumor-associated macrophages (TAMs) and was correlated with the initiation and progression of tumor cells and the polarization of TAMs. Finally, Kaplan-Meier plots demonstrated that higher expression of ANG was significantly correlated with shorter survival in gliomas. Cox regression analysis further confirmed ANG as an independent predictor of prognosis for gliomas of all three datasets. Conclusion ANG is significantly correlated with a range of malignant and aggressive characteristics in gliomas and reveals considerable prognostic value for glioma patients. ANG seems to be primarily associated with immune activities of macrophages and APCs in gliomas. Furthermore, ANG is mainly expressed in neoplastic cells and TAMs and is involved in the initiation and progression of neoplastic cells as well as macrophage polarization.
Collapse
Affiliation(s)
- Jin Wang
- Department of Emergency, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Aijun Shan
- Department of Emergency, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Fei Shi
- Department of Emergency, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| |
Collapse
|
4
|
Davoodvandi A, Rafiyan M, Asemi Z, Matini SA. An epigenetic modulator with promising therapeutic impacts against gastrointestinal cancers: A mechanistic review on microRNA-195. Pathol Res Pract 2023; 248:154680. [PMID: 37467635 DOI: 10.1016/j.prp.2023.154680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
Due to their high prevalence, gastrointestinal cancers are one of the key causes of cancer-related death globally. The development of drug-resistant cancer cell populations is a major factor in the high mortality rate, and it affects about half of all cancer patients. Because of advances in our understanding of cancer molecular biology, non-coding RNAs (ncRNAs) have emerged as critical factors in the initiation and development of gastrointestinal cancers. Gene expression can be controlled in several ways by ncRNAs, including through epigenetic changes, interactions between microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) and proteins, and the function of lncRNAs as miRNA precursors or pseudogenes. As lncRNAs may be detected in the blood, circulating ncRNAs have emerged as a promising new class of non-invasive cancer biomarkers for use in the detection, staging, and prognosis of gastrointestinal cancers, as well as in the prediction of therapy efficacy. In this review, we assessed the role lncRNAs play in the progression, and maintenance of colorectal cancer, and how they might be used as therapeutic targets in the future.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| | - Mahdi Rafiyan
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| | - Seyed Amirhassan Matini
- Department of Pathology, School of Medicine, Kashan University of Medical Sciences, Kashan, I.R. Iran.
| |
Collapse
|
5
|
Melaccio A, Reale A, Saltarella I, Desantis V, Lamanuzzi A, Cicco S, Frassanito MA, Vacca A, Ria R. Pathways of Angiogenic and Inflammatory Cytokines in Multiple Myeloma: Role in Plasma Cell Clonal Expansion and Drug Resistance. J Clin Med 2022; 11:jcm11216491. [PMID: 36362718 PMCID: PMC9658666 DOI: 10.3390/jcm11216491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, and despite the introduction of innovative therapies, remains an incurable disease. Identifying early and minimally or non-invasive biomarkers for predicting clinical outcomes and therapeutic responses is an active field of investigation. Malignant plasma cells (PCs) reside in the bone marrow (BM) microenvironment (BMME) which comprises cells (e.g., tumour, immune, stromal cells), components of the extracellular matrix (ECM) and vesicular and non-vesicular (soluble) molecules, all factors that support PCs’ survival and proliferation. The interaction between PCs and BM stromal cells (BMSCs), a hallmark of MM progression, is based not only on intercellular interactions but also on autocrine and paracrine circuits mediated by soluble or vesicular components. In fact, PCs and BMSCs secrete various cytokines, including angiogenic cytokines, essential for the formation of specialized niches called “osteoblastic and vascular niches”, thus supporting neovascularization and bone disease, vital processes that modulate the pathophysiological PCs–BMME interactions, and ultimately promoting disease progression. Here, we aim to discuss the roles of cytokines and growth factors in pathogenetic pathways in MM and as prognostic and predictive biomarkers. We also discuss the potential of targeted drugs that simultaneously block PCs’ proliferation and survival, PCs–BMSCs interactions and BMSCs activity, which may represent the future goal of MM therapy.
Collapse
Affiliation(s)
- Assunta Melaccio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| | - Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne 3004, Australia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- General Pathology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| |
Collapse
|
6
|
Circulating cytokines present in multiple myeloma patients inhibit the osteoblastic differentiation of adipose stem cells. Leukemia 2022; 36:540-548. [PMID: 34556797 DOI: 10.1038/s41375-021-01428-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 09/02/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Myeloma is characterized by bone lesions, which are related to both an increased osteoclast activity and a defect in the differentiation of medullary mesenchymal stem cells (MSCs) into osteoblasts. Outside the medullary environment, adipocyte-derived MSCs (ASCs) could represent a source of functional osteoblasts. However, we recently found a defect in the osteoblastic differentiation of ASCs from myeloma patients (MM-ASCs). We examined the effects of plasma from myeloma patients at diagnosis (MM-plasmas) and in complete remission (CR-plasmas) and from healthy donors on the osteoblastic differentiation of healthy donor-derived ASCs (HD-ASCs). Osteoblastogenesis in HD-ASCs was suppressed by MM-plasmas. Seven cytokines (ANG1, ENA-78, EGF, PDGF-AA/AB/BB, and TARC) were increased in MM-plasmas and separately inhibited the osteoblastic differentiation of HD-ASCs. Comparison of MM-ASCs and HD-ASCs by RNA sequencing showed that two master genes characterizing adipocyte differentiation, CD36 and PPARγ, were upregulated in MM-ASCs as compared to HD-ASCs. Finally, we demonstrated a significant increase in CD36 and PPARγ expression in HD-ASCs in the presence of MM-plasmas or the seven cytokines individually, similarly as in MM-ASCs. We conclude that specific cytokines in MM-plasmas, besides the well-known DKK1, inhibit the osteoblastic differentiation of MM- and HD-ASCs with a skewing towards adipocyte differentiation.
Collapse
|
7
|
Valsecchi C, Croce S, Maltese A, Montagna L, Lenta E, Nevone A, Girelli M, Milani P, Bosoni T, Massa M, Abbà C, Campanelli R, Ripepi J, De Silvestri A, Carolei A, Palladini G, Zecca M, Nuvolone M, Avanzini MA. Bone Marrow Microenvironment in Light-Chain Amyloidosis: In Vitro Expansion and Characterization of Mesenchymal Stromal Cells. Biomedicines 2021; 9:biomedicines9111523. [PMID: 34829752 PMCID: PMC8614719 DOI: 10.3390/biomedicines9111523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022] Open
Abstract
Immunoglobulin light-chain amyloidosis (AL) is caused by misfolded light chains produced by a small B cell clone. Mesenchymal stromal cells (MSCs) have been reported to affect plasma cell behavior. We aimed to characterize bone marrow (BM)-MSCs from AL patients, considering functional aspects, such as proliferation, differentiation, and immunomodulatory capacities. MSCs were in vitro expanded from the BM of 57 AL patients and 14 healthy donors (HDs). MSC surface markers were analyzed by flow cytometry, osteogenic and adipogenic differentiation capacities were in vitro evaluated, and co-culture experiments were performed in order to investigate MSC immunomodulatory properties towards the ALMC-2 cell line and HD peripheral blood mononuclear cells (PBMCs). AL-MSCs were comparable to HD-MSCs for morphology, immune-phenotype, and differentiation capacities. AL-MSCs showed a reduced proliferation rate, entering senescence at earlier passages than HD-MSCs. The AL-MSC modulatory effect on the plasma-cell line or circulating plasma cells was comparable to that of HD-MSCs. To our knowledge, this is the first study providing a comprehensive characterization of AL-MSCs. It remains to be defined if the observed abnormalities are the consequence of or are involved in the disease pathogenesis. BM microenvironment components in AL may represent the targets for the prevention/treatment of the disease in personalized therapies.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Stefania Croce
- General Surgery Department, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Alice Maltese
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Lorenza Montagna
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Elisa Lenta
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Alice Nevone
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maria Girelli
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Paolo Milani
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Tiziana Bosoni
- Clinical Chemistry Laboratory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Margherita Massa
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Carlotta Abbà
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Rita Campanelli
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Jessica Ripepi
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Annalisa De Silvestri
- Clinical Epidemiology and Biometry Unit, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy;
| | - Adriana Carolei
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
| | - Giovanni Palladini
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Marco Zecca
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
| | - Mario Nuvolone
- General Medicine 2—Center for Systemic Amyloidoses and High-Complexity Diseases, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (A.N.); (M.G.); (P.M.); (M.M.); (C.A.); (R.C.); (J.R.); (A.C.); (G.P.); (M.N.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology, Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy; (C.V.); (A.M.); (E.L.); (M.Z.)
- Correspondence: ; Tel.: +39-0382-502715
| |
Collapse
|
8
|
Immunological Prognostic Factors in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms22073587. [PMID: 33808304 PMCID: PMC8036885 DOI: 10.3390/ijms22073587] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm characterized by an abnormal proliferation of clonal, terminally differentiated B lymphocytes. Current approaches for the treatment of MM focus on developing new diagnostic techniques; however, the search for prognostic markers is also crucial. This enables the classification of patients into risk groups and, thus, the selection of the most optimal treatment method. Particular attention should be paid to the possible use of immune factors, as the immune system plays a key role in the formation and course of MM. In this review, we focus on characterizing the components of the immune system that are of prognostic value in MM patients, in order to facilitate the development of new diagnostic and therapeutic directions.
Collapse
|
9
|
Zarfati M, Avivi I, Brenner B, Katz T, Aharon A. Extracellular vesicles of multiple myeloma cells utilize the proteasome inhibitor mechanism to moderate endothelial angiogenesis. Angiogenesis 2018; 22:185-196. [PMID: 30386953 DOI: 10.1007/s10456-018-9649-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023]
Abstract
Bone marrow microenvironment is known to support angiogenesis, thus contributing to progression of multiple myeloma (MM). Bortezomib, a proteasome inhibitor (PI) widely used in MM treatment, has anti-angiogenic activity. Extracellular vesicles (EVs), shedding from cell surface, serve as mediators in cell-to-cell communication. We have hypothesized that MM cells (MMCs) treated with bortezomib generate EVs that could diminish angiogenesis, thus limiting MM progression. In the present study, EVs were obtained from MMCs (RPMI-8226), untreated (naïve) or pre-treated with bortezomib. EVs were outlined using NanoSight, FACS, protein arrays and proteasome activity assays. The impact of MMC-EVs on endothelial cell (EC) functions was assessed, employing XTT assay, Boyden chamber and Western blot. A high apoptosis level (annexin V binding 70.25 ± 16.37%) was observed in MMCs following exposure to bortezomib. Compared to naïve EVs, a large proportion of bortezomib-induced EVs (Bi-EVs) were bigger in size (> 300 nm), with higher levels of annexin V binding (p = 0.0043).They also differed in content, presenting with increased levels of pro-inflammatory proteins, reduced levels of pro-angiogenic growth factors (VEGFA, PDGF-BB, angiogenin), and displayed lower proteasome activity. Naïve EVs were found to promote EC migration and proliferation via ERK1/2 and JNK1/2/3 phosphorylation, whereas Bi-EVs inhibited these functions. Moreover, Bi-EVs appeared to reduce EC proteasome activity. EVs released from apoptotic MMCs following treatment with bortezomib can promote angiogenesis suppression by decreasing proliferation and migration of EC. These activities are found to be mediated by specific signal transduction pathways.
Collapse
Affiliation(s)
- Moran Zarfati
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Irit Avivi
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 7, Ha'Aliya St., Haifa, 3109601, Israel
| | - Benjamin Brenner
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 7, Ha'Aliya St., Haifa, 3109601, Israel
| | - Tami Katz
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.,Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 7, Ha'Aliya St., Haifa, 3109601, Israel
| | - Anat Aharon
- Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel. .,Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, 7, Ha'Aliya St., Haifa, 3109601, Israel.
| |
Collapse
|
10
|
Mondello P, Cuzzocrea S, Navarra M, Mian M. Bone marrow micro-environment is a crucial player for myelomagenesis and disease progression. Oncotarget 2017; 8:20394-20409. [PMID: 28099912 PMCID: PMC5386771 DOI: 10.18632/oncotarget.14610] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/05/2017] [Indexed: 01/06/2023] Open
Abstract
Despite the advent of many therapeutic agents, such as bortezomib and lenalidomide that have significantly improved the overall survival, multiple myeloma remains an incurable disease. Failure to cure is multifactorial and can be attributed to the underlying genetic heterogeneity of the cancer and to the surrounding micro-environment. Understanding the mutual interaction between myeloma cells and micro-environment may lead to the development of novel treatment strategies able to eradicate this disease. In this review we discuss the principal molecules involved in the micro-environment network in multiple myeloma and the currently available therapies targeting them.
Collapse
Affiliation(s)
- Patrizia Mondello
- Department of Human Pathology, University of Messina, Messina, Italy.,Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.,Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Michael Mian
- Department of Hematology and Center of Bone Marrow Transplantation, Hospital of Bolzano, Bolzano/Bozen, Italy.,Department of Internal Medicine V, Hematology & Oncology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Lia G, Brunello L, Bruno S, Carpanetto A, Omedè P, Festuccia M, Tosti L, Maffini E, Giaccone L, Arpinati M, Ciccone G, Boccadoro M, Evangelista A, Camussi G, Bruno B. Extracellular vesicles as potential biomarkers of acute graft-vs-host disease. Leukemia 2017; 32:765-773. [PMID: 28852198 DOI: 10.1038/leu.2017.277] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/31/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023]
Abstract
Acute graft-vs-host disease (GVHD) is a serious complication after allografting. We carried out an exploratory study to investigate a potential correlation of surface antigens on extracellular vesicles (EVs) and acute GVHD. EVs were extracted from serum samples from 41 multiple myeloma patients who underwent allografting. EVs were characterized by flow cytometry using a panel of 13 antibodies against specific membrane proteins that were reported to be predictive of acute GVHD. We observed a correlation between three potential biomarkers expressed on EV surface and acute GVHD onset by both logistic regression analysis and Cox proportional hazard model. In our study, CD146 (MCAM-1) was correlated with an increased risk-by almost 60%-of developing GVHD, whereas CD31 and CD140-α (PECAM-1 and PDGFR-α) with a decreased risk-by almost 40 and 60%, respectively. These biomarkers also showed a significant change in signal level from baseline to the onset of acute GVHD. Our novel study encourages future investigations into the potential correlation between EVs and acute GVHD. Larger prospective multicenter studies are currently in progress.
Collapse
Affiliation(s)
- G Lia
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - L Brunello
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - S Bruno
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Centro di Biotecnologie Molecolari, Torino, Italy
| | - A Carpanetto
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Centro di Biotecnologie Molecolari, Torino, Italy
| | - P Omedè
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy
| | - M Festuccia
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - L Tosti
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - E Maffini
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - L Giaccone
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - M Arpinati
- Dipartimento di Ematologia e Scienze Oncologiche 'L. e A. Seràgnoli', Università di Bologna, Bologna, Italy
| | - G Ciccone
- A.O.U. Città della Salute e della Scienza di Torino, Epidemiologia Clinica, Torino, Italy
| | - M Boccadoro
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| | - A Evangelista
- A.O.U. Città della Salute e della Scienza di Torino, Epidemiologia Clinica, Torino, Italy
| | - G Camussi
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Centro di Biotecnologie Molecolari, Torino, Italy
| | - B Bruno
- A.O.U. Città della Salute e della Scienza di Torino, Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università di Torino, School of Medicine, Torino, Italy
| |
Collapse
|
12
|
Ntellas P, Perivoliotis K, Dadouli K, Koukoulis GK, Ioannou M. Microvessel Density as a Surrogate Prognostic Marker in Patients with Multiple Myeloma: A Meta-Analysis. Acta Haematol 2017; 138:77-84. [PMID: 28796988 DOI: 10.1159/000478085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS Bone marrow (BM) angiogenesis is considered a hallmark of multiple myeloma (MM) development and progression, and can be quantified with the use of microvessel density (MVD). The purpose of this study is to provide a review and a meta-analysis of the current literature regarding the prognostic value of MVD in the overall survival (OS) of MM patients. METHODS MEDLINE was screened for studies evaluating the OS of MM patients with regard to their MVD count in BM trephine. The pooled hazard ratio (HR) and its associated 95% confidence interval (CI) among MM patients with a high and low MVD count was the primary end point. Secondary outcomes included odds ratios (OR) for 12-, 36-, and 60-month survival. RESULTS Ten eligible trials were identified for the analysis of the primary end point and 9 for the secondary end points. Pooled HR for OS was 1.85 (95% CI: 1.25-2.73, p = 0.002). The pooled OR of survival were 1.59 (95% CI: 1.02-2.46, p = 0.04) at 12 months, 2.90 (95% CI: 1.68-5.03, p = 0.0001) at 36 months, and 3.42 (95% CI: 2.41-4.85, p < 0.00001) at 60 months, in favor of the low MVD group. CONCLUSION This meta-analysis provides persuasive evidence that MVD has significant impact on the clinical outcome of MM patients.
Collapse
Affiliation(s)
- Panagiotis Ntellas
- Department of Pathology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | | | | | | |
Collapse
|
13
|
Ryu J, Koh Y, Park H, Kim DY, Kim DC, Byun JM, Lee HJ, Yoon SS. Highly Expressed Integrin-α8 Induces Epithelial to Mesenchymal Transition-Like Features in Multiple Myeloma with Early Relapse. Mol Cells 2016; 39:898-908. [PMID: 28008160 PMCID: PMC5223107 DOI: 10.14348/molcells.2016.0210] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/24/2016] [Accepted: 12/05/2016] [Indexed: 12/12/2022] Open
Abstract
Despite recent groundbreaking advances in multiple myeloma (MM) treatment, most MM patients ultimately experience relapse, and the relapse biology is not entirely understood. To define altered gene expression in MM relapse, gene expression profiles were examined and compared among 16 MM patients grouped by 12 months progression-free survival (PFS) after autologous stem cell transplantation. To maximize the difference between prognostic groups, patients at each end of the PFS spectrum (the four with the shortest PFS and four with the longest PFS) were chosen for additional analyses. We discovered that integrin-α8 (ITGA8) is highly expressed in MM patients with early relapse. The integrin family is well known to be involved in MM progression; however, the role of integrin-α8 is largely unknown. We functionally overexpressed integrin-α8 in MM cell lines, and surprisingly, stemness features including HIF1α, VEGF, OCT4, and Nanog, as well as epithelial mesenchymal transition (EMT)-related phenotypes, including N-cadherin, Slug, Snail and CXCR4, were induced. These, consequently, enhanced migration and invasion abilities, which are crucial to MM pathogenesis. Moreover, the gain of integrin-α8 expression mediated drug resistance against melphalan and bortezomib, which are the main therapeutic agents in MM. The cBioPortal genomic database revealed that ITGA8 have significant tendency to co-occur with PDGFRA and PDGFRB and their mRNA expression were up-regulated in ITGA8 overexpressed MM cells. In summary, integrin-α8, which was up-regulated in MM of early relapse, mediates EMT-like phenotype, enhancing migration and invasion; therefore, it could serve as a potential marker of MM relapse and be a new therapeutic target.
Collapse
Affiliation(s)
- Jiyeon Ryu
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
| | - Hyejoo Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Dae Yoon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Dong Chan Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Ja Min Byun
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
| | - Hyun Jung Lee
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang 10326,
Korea
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080,
Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080,
Korea
| |
Collapse
|
14
|
Agrawal S, Ganguly S, Hajian P, Cao JN, Agrawal A. PDGF upregulates CLEC-2 to induce T regulatory cells. Oncotarget 2016; 6:28621-32. [PMID: 26416420 PMCID: PMC4745681 DOI: 10.18632/oncotarget.5765] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/12/2015] [Indexed: 01/01/2023] Open
Abstract
The effect of platelet derived growth factor (PDGF) on immune cells is not elucidated. Here, we demonstrate PDGF inhibited the maturation of human DCs and induced IL-10 secretion. Culture of PDGF-DCs with T cells induced the polarization of T cells towards FoxP3 expressing T regulatory cells that secreted IL-10. Gene expression studies revealed that PDGF induced the expression of C-type lectin like receptor member 2, (CLEC-2) receptor on DCs. Furthermore, DCs transfected with CLEC-2 induced T regulatory cells in DC-T cell co-culture. CLEC-2 is naturally expressed on platelets. Therefore, to confirm whether CLEC-2 is responsible for inducing the T regulatory cells, T cells were cultured with either CLEC-2 expressing platelets or soluble CLEC-2. Both conditions resulted in the induction of regulatory T cells. The generation of T regulatory cells was probably due to the binding of CLEC-2 with its ligand podoplanin on T cells, since crosslinking of podoplanin on the T cells also resulted in the induction of T regulatory cells. These data demonstrate that PDGF upregulates the expression of CLEC-2 on cells to induce T regulatory cells.
Collapse
Affiliation(s)
- Sudhanshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Sreerupa Ganguly
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Pega Hajian
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Jia-Ning Cao
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
15
|
Insights into the molecular roles of heparan sulfate proteoglycans (HSPGs—syndecans) in autocrine and paracrine growth factor signaling in the pathogenesis of Hodgkin’s lymphoma. Tumour Biol 2016; 37:11573-11588. [DOI: 10.1007/s13277-016-5118-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/09/2016] [Indexed: 12/25/2022] Open
|
16
|
Antiangiogenesis and vascular disrupting agents in cancer: circumventing resistance and augmenting their therapeutic utility. Future Med Chem 2016; 8:443-62. [DOI: 10.4155/fmc.16.6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a process essential for tumor growth and metastasis. Inhibition of angiogenesis as an anticancer strategy has shown only moderately improved results and is beset with practical limitations, despite theoretical therapeutic advantages. Inevitably resistance develops, through redundancy of signaling pathways and selection for subclonal populations adapted for hypoxic conditions, with more invasive phenotypes. Antiangiogenic-targeted therapies may find improved efficacy in combination therapies; with others in this class, that directly or indirectly target separate pathways or different components of the same pathway, or with a separate class of tumor vasculature-disrupting agents. This review discusses the challenges and strategies for optimization of combination therapies including metronomic administration of drugs and the need for suitable prognostic and surrogate response biomarkers.
Collapse
|
17
|
Chen S, Guo X, Imarenezor O, Imoukhuede PI. Quantification of VEGFRs, NRP1, and PDGFRs on Endothelial Cells and Fibroblasts Reveals Serum, Intra-Family Ligand, and Cross-Family Ligand Regulation. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0411-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
18
|
Alexandrakis MG, Neonakis IK, Pappa CA, Konsolas I, Kokonozaki M, Vyzoukaki R, Soundoulounaki S, Xekalou A, Sfiridaki K, Tsirakis G. Immunohistochemical expression of endoglin offers a reliable estimation of bone marrow neoangiogenesis in multiple myeloma. J Cancer Res Clin Oncol 2015; 141:1503-9. [PMID: 25773125 DOI: 10.1007/s00432-015-1952-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/05/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE The aim of the present study was to evaluate CD105 tissue marker in the bone marrow (BM) of multiple myeloma (MM) patients. CD105 was evaluated using immunohistochemical method. An effort was made to correlate this marker with BM microvascular density (MVD) along with other known markers of angiogenesis in order to evaluate its clinical significance. METHODS BM MVD was estimated by CD31. CD105 in BM was estimated by immunohistochemical method in 54 newly diagnosed patients with MM. Circulating levels of known angiogenic factors such as basic fibroblast growth factor (b-FGF) and soluble CD105 (sCD105) were measured by ELISA in the same group of patients. All these factors were also measured in 20 age- and sex-matched healthy controls. RESULTS We found that CD105 MVD, along with the expected CD31 MVD, and serum levels of sCD105 and bFGF were increased, also in parallel with disease stage, and all were decreased after effective treatment. Moreover, CD105 MVD correlated with all the aforementioned markers of angiogenesis. CONCLUSIONS Our results indicated that CD105 MVD is following the behavior of CD31 MVD in MM, suggesting being a valid marker of BM neoangiogenesis in MM. Its prognostic impact remains to be proven.
Collapse
Affiliation(s)
- Michael G Alexandrakis
- Hematology Department, University Hospital of Heraklion, P.O. BOX 1352, 71110, Voutes, Heraklion, Greece,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Pappa CA, Alexandrakis MG, Boula A, Thanasia A, Konsolas I, Alegakis A, Tsirakis G. Prognostic impact of angiopoietin-2 in multiple myeloma. J Cancer Res Clin Oncol 2014; 140:1801-5. [PMID: 24906877 DOI: 10.1007/s00432-014-1731-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/30/2014] [Indexed: 12/20/2022]
Abstract
PURPOSE Angiogenesis is an essential process for the expansion of multiple myeloma (MM). Angiopoietin-2 (Ang-2), Ang-1 and their receptor possess important roles in this procedure. The aim of the study was to measure serum levels of Ang-2 along with known markers of angiogenesis and to estimate their prognostic impact on the survival. METHODS Bone marrow microvascular density (MVD), estimated by CD31, and circulating levels of known angiogenic factors Ang-2, interleukin-6, soluble CD105 and platelet-derived growth factor-AB, measured by ELISA, were measured in 77 newly diagnosed patients with active MM and in 57 of them who responded to chemotherapy. RESULTS All measured parameters were increased in MM patients, were also increasing in advanced disease and decreased after effective treatment. Ang-2 correlated positively with the other angiogenic factors and MVD. Moreover, Ang-2 values above the median were accompanied by worse survival. CONCLUSION Ang-2 correlates strongly with the angiogenic process and its serum levels are importantly prognostic for survival, highlighting the role of angiopoietins pathway in the biology of MM.
Collapse
Affiliation(s)
- Constantina A Pappa
- Hematology Department, Venizelion General Hospital of Heraklion, Heraklion, Greece
| | | | | | | | | | | | | |
Collapse
|
20
|
Changes in the peripheral blood and bone marrow from untreated advanced breast cancer patients that are associated with the establishment of bone metastases. Clin Exp Metastasis 2013; 31:213-32. [PMID: 24173696 DOI: 10.1007/s10585-013-9622-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/13/2013] [Indexed: 10/26/2022]
Abstract
Bone metastasis is an incurable complication of breast cancer affecting 70-80 % of advanced patients. It is a multistep process that includes tumour cell mobilisation, intravasation, survival in the circulation, extravasation, migration and proliferation in the bone marrow/bone. Although novel findings demonstrate the bone marrow microenvironment significance in bone metastatic progression, a majority of studies have focused on end-stage disease and little is known about how the pre-metastatic niche arises in the bone marrow/bone tissues. We demonstrated a significant increase in patients' peripheral blood plasma ability to induce transendothelial migration of MCF-7 cells compared with healthy volunteers. Moreover, high RANKL, MIF and OPG levels in patients' peripheral blood could play a role in the intravasation, angiogenesis, survival and epithelial-mesenchymal transition of circulating tumour cells. Also, we observed a significant increase in patients' bone marrow plasma capacity to induce transendothelial migration of MDA-MB231 and MCF-7 cells compared with healthy volunteers. Furthermore, patients' bone marrow mesenchymal stem cells could control the recruitment of tumour cells, modifying the MCF-7 and MDA-MB231 cell migration. In addition, we found a significantly higher MDA-MB231 cell proliferation when we used patients' bone marrow plasma compared with healthy volunteers. Interestingly, PDGF-AB, ICAM-1 and VCAM-1 levels in patients' bone marrow were significantly higher than the values of healthy volunteers, suggesting that they could be involved in the cancer cell extravasation, bone resorption and cancer cell proliferation. We believe that these results can reveal new information about what alterations happen in the bone marrow of advanced breast cancer patients before bone colonisation, changes that create optimal soil for the metastatic cascade progression.
Collapse
|
21
|
Lemancewicz D, Bolkun L, Mantur M, Semeniuk J, Kloczko J, Dzieciol J. Bone marrow megakaryocytes, soluble P-selectin and thrombopoietic cytokines in multiple myeloma patients. Platelets 2013; 25:181-7. [DOI: 10.3109/09537104.2013.805405] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
22
|
Lee C, Zhang F, Tang Z, Liu Y, Li X. PDGF-C: a new performer in the neurovascular interplay. Trends Mol Med 2013; 19:474-86. [PMID: 23714575 DOI: 10.1016/j.molmed.2013.04.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/15/2013] [Accepted: 04/26/2013] [Indexed: 12/30/2022]
Abstract
The importance of neurovascular crosstalk in development, normal physiology, and pathologies is increasingly being recognized. Although vascular endothelial growth factor (VEGF), a prototypic regulator of neurovascular interaction, has been studied intensively, defining other important regulators in this process is warranted. Recent studies have shown that platelet-derived growth factor C (PDGF-C) is both angiogenic and a neuronal survival factor, and it appears to be an important component of neurovascular crosstalk. Importantly, the expression pattern and functional properties of PDGF-C and its receptors differ from those of VEGF, and thus the PDGF-C-mediated neurovascular interaction may represent a new paradigm of neurovascular crosstalk.
Collapse
Affiliation(s)
- Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, P.R. China
| | | | | | | | | |
Collapse
|
23
|
Pappa CA, Tsirakis G, Boula A, Sfiridaki A, Psarakis FE, Alexandrakis MG, Stathopoulos EN. The Significance of non Correlation Between Interleukin-8 Serum Levels with Bone Marrow Microvascular Density in Patients with Myeloma Multiple. Pathol Oncol Res 2013; 19:539-43. [DOI: 10.1007/s12253-013-9614-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 02/11/2013] [Indexed: 02/07/2023]
|
24
|
In silico prediction and in vitro characterization of multifunctional human RNase3. BIOMED RESEARCH INTERNATIONAL 2013; 2013:170398. [PMID: 23484086 PMCID: PMC3581242 DOI: 10.1155/2013/170398] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/02/2012] [Indexed: 12/18/2022]
Abstract
Human ribonucleases A (hRNaseA) superfamily consists of thirteen members with high-structure similarities but exhibits divergent physiological functions other than RNase activity. Evolution of hRNaseA superfamily has gained novel functions which may be preserved in a unique region or domain to account for additional molecular interactions. hRNase3 has multiple functions including ribonucleolytic, heparan sulfate (HS) binding, cellular binding, endocytic, lipid destabilization, cytotoxic, and antimicrobial activities. In this study, three putative multifunctional regions, 34RWRCK38 (HBR1), 75RSRFR79 (HBR2), and 101RPGRR105 (HBR3), of hRNase3 have been identified employing in silico sequence analysis and validated employing in vitro activity assays. A heparin binding peptide containing HBR1 is characterized to act as a key element associated with HS binding, cellular binding, and lipid binding activities. In this study, we provide novel insights to identify functional regions of hRNase3 that may have implications for all hRNaseA superfamily members.
Collapse
|