1
|
Kim H, Oh J, Kim MK, Lee KH, Jeong D. Selenoprotein W engages in overactive osteoclast differentiation in multiple myeloma. Mol Biol Rep 2024; 51:587. [PMID: 38683225 PMCID: PMC11058866 DOI: 10.1007/s11033-024-09517-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Patients with multiple myeloma exhibit malignant osteolytic bone disease due to excessive osteoclast formation and function. We recently identified that osteoclastogenic stimulator selenoprotein W (SELENOW) is upregulated via ERK signaling and downregulated via p38 signaling during receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation. In the intrinsic physiological process, RANKL-induced downregulation of SELENOW maintains proper osteoclast differentiation; in contrast, forced overexpression of SELENOW leads to overactive osteoclast formation and function. METHODS AND RESULTS We observed that SELENOW is highly expressed in multiple myeloma-derived peripheral blood mononuclear cells (PBMCs) and mature osteoclasts when compared to healthy controls. Also, the level of tumor necrosis factor alpha (TNFα), a pathological osteoclastogenic factor, is increased in the PBMCs and serum of patients with multiple myeloma. ERK activation by TNFα was more marked and sustained than that by RANKL, allowing SELENOW upregulation. Excessive expression of SELENOW in osteoclast progenitors and mature osteoclasts derived from multiple myeloma facilitated efficient nuclear translocation of osteoclastogenic transcription factors NF-κB and NFATc1, which are favorable for osteoclast formation. CONCLUSION Our findings suggest a possibility that feedforward signaling of osteoclastogenic SELENOW by TNFα derived from multiple myeloma induces overactive osteoclast differentiation, leading to bone loss during multiple myeloma.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Laboratory of Bone Metabolism and Control, Department of Microbiology, Yeungnam University College of Medicine, Daegu, 42415, Korea
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA19104, USA
| | - Jiin Oh
- Laboratory of Bone Metabolism and Control, Department of Microbiology, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Min Kyoung Kim
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Kyung Hee Lee
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Daewon Jeong
- Laboratory of Bone Metabolism and Control, Department of Microbiology, Yeungnam University College of Medicine, Daegu, 42415, Korea.
- Company of The Bone Science, Yeungnam University College of Medicine, Daegu, 42415, Korea.
| |
Collapse
|
2
|
Misra S, Singh A, Kumar V. Plasma cell myeloma with double M bands on Serum protein electrophoresis: A diagnostic conundrum? J Cancer Res Ther 2024; 20:1026-1028. [PMID: 38102909 DOI: 10.4103/jcrt.jcrt_1362_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 08/23/2022] [Indexed: 12/17/2023]
Abstract
ABSTRACT Plasma cell myeloma (PCM) is a monoclonal gammopathy (MGM) characterized by proliferation of abnormal clone of plasma cells infiltrating the bone marrow with consequent end organ damage. The clonal plasma cells secrete a single clone of immunoglobulins (Ig) leading to presence of M-protein in the serum and/or urine. The M-protein is appreciated as a discrete band on serum protein electrophoresis (SPE) in the gamma globulin region, also called the M-band. Biclonal gammopathy (BGM) occurs due to neoplastic transformation of a plasma cell clone undergoing Ig class switching or due to an independent neoplastic transformation event yielding proliferation of unrelated plasma cell clones, therefore resulting in two distinct M-bands on SPE. It is, however, vital to distinguish a true BGM from an apparent one (MGM presenting with two distinct bands on SPE) so as to make an accurate diagnosis. Hereby, we report a case of a 61-year-old man, diagnosed with PCM and presenting with two discrete bands on SPE (simulating a BGM) which turned out to be monoclonal in nature.
Collapse
Affiliation(s)
- Sunayana Misra
- Department of Pathology, ABVIMS and Dr. RML Hospital, Delhi, India
| | | | | |
Collapse
|
3
|
Morales-Martínez M, Vega MI. p38 Molecular Targeting for Next-Generation Multiple Myeloma Therapy. Cancers (Basel) 2024; 16:256. [PMID: 38254747 PMCID: PMC10813990 DOI: 10.3390/cancers16020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Resistance to therapy and disease progression are the main causes of mortality in most cancers. In particular, the development of resistance is an important limitation affecting the efficacy of therapeutic alternatives for cancer, including chemotherapy, radiotherapy, and immunotherapy. Signaling pathways are largely responsible for the mechanisms of resistance to cancer treatment and progression, and multiple myeloma is no exception. p38 mitogen-activated protein kinase (p38) is downstream of several signaling pathways specific to treatment resistance and progression. Therefore, in recent years, developing therapeutic alternatives directed at p38 has been of great interest, in order to reverse chemotherapy resistance and prevent progression. In this review, we discuss recent findings on the role of p38, including recent advances in our understanding of its expression and activity as well as its isoforms, and its possible clinical role based on the mechanisms of resistance and progression in multiple myeloma.
Collapse
Affiliation(s)
- Mario Morales-Martínez
- Molecular Signal Pathway in Cancer Laboratory, UIMEO, Oncology Hospital, Siglo XXI National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
| | - Mario I. Vega
- Molecular Signal Pathway in Cancer Laboratory, UIMEO, Oncology Hospital, Siglo XXI National Medical Center, Mexican Institute of Social Security (IMSS), Mexico City 06720, Mexico
- Department of Medicine, Hematology-Oncology and Clinical Nutrition Division, Greater Los Angeles VA Healthcare Center, UCLA Medical Center, Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Bucheli OTM, Eyer K. Insights into the relationship between persistent antibody secretion and metabolic programming - A question for single-cell analysis. Immunol Lett 2023; 260:35-43. [PMID: 37315849 DOI: 10.1016/j.imlet.2023.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/28/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Vaccination aims to generate a protective and persisting antibody response. Indeed, humoral vaccine-mediated protection depends on the quality and quantity of the produced antigen-specific antibodies for its initial magnitude and the persistence of the plasma cells for its duration. Therefore, understanding the mechanisms behind the generation, selection and maintenance of long-lived plasma cells secreting protective antibodies is of fundamental importance for understanding long-term immunity, vaccine responses, therapeutical approaches for autoimmune disease and multiple myeloma. Recent studies have observed correlations between the generation, function and lifespan of plasma cells and their metabolism, with metabolism being both a main driver and primary consequence of changes in cellular behavior. This review introduces how metabolic programs influence and drive immune cell functions in general and plasma cell differentiation and longevity more specifically, summarizing the current knowledge on metabolic pathways and their influences on cellular fate. In addition, available technologies to profile metabolism and their limitations are discussed, leading to the unique and open technological challenges for further advancement of this research field.
Collapse
Affiliation(s)
- Olivia T M Bucheli
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, 8093 Zürich, Switzerland
| | - Klaus Eyer
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, 8093 Zürich, Switzerland.
| |
Collapse
|
5
|
Neri P, Nijhof I. Evidence-based mechanisms of synergy with IMiD agent-based combinations in multiple myeloma. Crit Rev Oncol Hematol 2023:104041. [PMID: 37268176 DOI: 10.1016/j.critrevonc.2023.104041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023] Open
Abstract
Treatment of multiple myeloma (MM) has seen great advances in recent years, and a key contributor to this change has been the effective use of combination therapies, which have improved both the depth and duration of patient responses. IMiD agents (lenalidomide and pomalidomide) have both tumoricidal and immunostimulatory functions, and due to their multiple mechanisms of action have become the backbone of numerous combination treatments in the newly diagnosed and relapsed/refractory settings. Although IMiD agent-based combination regimens provide improved clinical outcomes for patients with MM, the mechanisms underpinning these combinations are not well understood. In this review we describe the potential mechanisms of synergy leading to the enhanced activity observed when IMiD agents and other drug classes are used in combination through interrogation of the current knowledge surrounding their mechanism of actions.
Collapse
Affiliation(s)
- Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
| | - Inger Nijhof
- Department of Hematology, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Internal Medicine and Department of Hematology, St. Antonius Hospital Nieuwegein, Koekoekslaan 1, 3435CM, Nieuwegein, the Netherlands
| |
Collapse
|
6
|
Su Q, Li Q, Zhang W, Li B, Zhuang W. Integrative analysis of enrichment and prognostic value of ferroptosis-related genes and pathways in multiple myeloma. Carcinogenesis 2022; 43:1050-1058. [PMID: 36170154 DOI: 10.1093/carcin/bgac080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/31/2022] [Accepted: 09/27/2022] [Indexed: 02/04/2023] Open
Abstract
Ferroptosis is a non-apoptotic form of cell death caused by excessive iron exposure. The role played by the ferroptosis-related genes and pathways in multiple myeloma (MM) is poorly understood. Here, we show that the ferroptosis-related pathways might be involved in tumorigenesis and are closely correlated with the prognosis of MM. The ferroptosis suppressor genes are progressively enriched with the progression of plasma cell dyscrasias. Furthermore, high expression of ferroptosis suppressor genes is correlated with high International Staging System and Revised-ISS staging of MM, as well as the poor outcomes of poor outcomes in progression-free survival and overall survival . The ferroptosis driver genes and the ferroptosis suppressor genes have the opposite effects on the progression and prognosis of MM. Moreover, we reveal that ferroptosis-related genes are associated with cytogenetic abnormalities in MM. The ferroptosis-related pathways and genes might impact the osteogenic differentiation of mesenchymal stromal cells in MM patients. A better understanding of the participation of ferroptosis in MM will pave the way for design of new therapies.
Collapse
Affiliation(s)
- Qi Su
- Department of Cell Biology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qi Li
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Weimin Zhang
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bingzong Li
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenzhuo Zhuang
- Department of Cell Biology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
7
|
Solimando AG, Da Vià MC, Bolli N, Steinbrunn T. The Route of the Malignant Plasma Cell in Its Survival Niche: Exploring “Multiple Myelomas”. Cancers (Basel) 2022; 14:cancers14133271. [PMID: 35805041 PMCID: PMC9265748 DOI: 10.3390/cancers14133271] [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: 05/13/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Growing evidence points to multiple myeloma (MM) and its stromal microenvironment using several mechanisms to subvert effective immune and anti-tumor responses. Recent advances have uncovered the tumor-stromal cell influence in regulating the immune-microenvironment and have envisioned targeting these suppressive pathways to improve therapeutic outcomes. Nevertheless, some subgroups of patients include those with particularly unfavorable prognoses. Biological stratification can be used to categorize patient-, disease- or therapy-related factors, or alternatively, these biological determinants can be included in a dynamic model that customizes a given treatment to a specific patient. Genetic heterogeneity and current knowledge enforce a systematic and comprehensive bench-to-bedside approach. Given the increasing role of cancer stem cells (CSCs) in better characterizing the pathogenesis of solid and hematological malignancies, disease relapse, and drug resistance, identifying and describing CSCs is of paramount importance in the management of MM. Even though the function of CSCs is well-known in other cancer types, their role in MM remains elusive. With this review, we aim to provide an update on MM homing and resilience in the bone marrow micro milieu. These data are particularly interesting for clinicians facing unmet medical needs while designing novel treatment approaches for MM.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine ‘G. Baccelli’, University of Bari Medical School, 70124 Bari, Italy
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| | - Matteo Claudio Da Vià
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.C.D.V.); (N.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Torsten Steinbrunn
- Department of Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (A.G.S.); (T.S.); Tel.: +39-3395626475 (A.G.S.)
| |
Collapse
|
8
|
Jalloul M, Sater AA, Ballout I, Annan K, Mokdad T, Lakis ZA, Khachfe HH. Multiple myeloma in Lebanon: Trend analysis, 10-year projections and comparisons to other countries. Cancer Treat Res Commun 2022; 30:100513. [PMID: 35026534 DOI: 10.1016/j.ctarc.2022.100513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/23/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Multiple myeloma (MM) is a common hematological malignancy in aging populations. This study aims to inspect the trends from 2005 to 2016 and future projections of MM in Lebanon and to compare its incidence-rates to other MENA and non-MENA countries. METHODS The data concerning MM cases over the studied period was extracted from the National Cancer Registry (NCR) of Lebanon. The online database "Cancer Incidence in Five Continents" was screened and data of other countries were collected. The age-specific and age-standardized incidence rates (ASR(w)) were computed and analyzed using Joinpoint regression. 10 year projections were predicted by employing a logarithmic model. RESULTS During the time period 2005-2016, MM was significantly more common in males. Both genders had a cancer peak in patients older than 75 years. MM demonstrated a significantly increasing trend in both genders over the 12 years of study. Compared to other countries in the MENA region, Lebanon ranked first in females and second in males, while it came among the lowest when compared to randomly selected developed countries from non-MENA regions. Projecting to 2026, incidence rates of MM in Lebanon are expected to rise in both males and females. CONCLUSION Incidence of MM in Lebanon is continually rising. The elderly population, especially males, is much more affected than the younger one. Different risk factors, specifically obesity and toxic exposures, can explain the escalating burden of MM among the population. A collaboration of efforts between the government and health organization is expected for an effective disease control.
Collapse
Affiliation(s)
- Mohammad Jalloul
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - AliH Abdel Sater
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - Ibrahim Ballout
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - KhalilEl Annan
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - Taha Mokdad
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - Zeina A Lakis
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - Hussein H Khachfe
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
9
|
Zeng Z, Lin J, Zhang K, Guo X, Zheng X, Yang A, Chen J. Single cell RNA-seq data and bulk gene profiles reveal a novel signature of disease progression in multiple myeloma. Cancer Cell Int 2021; 21:511. [PMID: 34563174 PMCID: PMC8465778 DOI: 10.1186/s12935-021-02190-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/01/2021] [Indexed: 01/22/2023] Open
Abstract
Background The development of multiple myeloma (MM) is considered to involve a multistep transformation process, but the role of cytogenetic abnormalities and molecular alterations in determining the cell fate of multiple myeloma (MM) remains unclear. Here, we have analyzed single cell RNA-seq data and bulk gene profiles to reveal a novel signature associated with MM development. Methods The scRNA-seq data from GSE118900 was used to profile the transcriptomes of cells from MM patients at different stages. Pseudotemporal ordering of the single cells was performed using Monocle package to feature distinct transcriptomic states of the developing MM cells. The bulk microarray profiles from GSE24080 and GSE9782 were applied to identify a signature associated with MM development. Results The 597 cells were divided into 7 clusters according to different risk levels. They were initiated mainly from monoclonal gammopathy of undetermined significance (MGUS), newly diagnosed MM (NDMM), or relapsed and/or refractory myeloma (RRMM) with cytogenetically favorable t(11;14), moved towards the cells from smoldering MM (SMM) or NDMM without t(11;14) or t(4;14), and then finally to cells from SMM or RRMM with t(4;14). Based on the markers identified in the late stage, the bulk data was used to develop a 20-gene signature stratifying patients into high and low-risk groups (GSE24080: HR = 3.759, 95% CI 2.746–5.145; GSE9782: HR = 2.612, 95% CI 1.894–3.603), which was better than the previously published gene signatures (EMC92, UAMS70, and UAMS17) and International Staging System. This signature also succeeded in predicting the clinical outcome of patients treated with bortezomib (HR = 2.884, 95% CI 1.994–4.172, P = 1.89e−8). The 20 genes were further verified by quantitative real-time polymerase chain reaction using samples obtained from the patients with MM. Conclusion Our comprehensive analyses offered new insights in MM development, and established a 20-gene signature as an independent biomarker for MM. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02190-6.
Collapse
Affiliation(s)
- Zhiyong Zeng
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Junfang Lin
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Kejie Zhang
- Department of Hematology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Xizhe Guo
- Department of Hematology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiaoqiang Zheng
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Apeng Yang
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Junmin Chen
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
10
|
Desantis V, Solimando AG, Saltarella I, Sacco A, Giustini V, Bento M, Lamanuzzi A, Melaccio A, Frassanito MA, Paradiso A, Montagnani M, Vacca A, Roccaro AM. MicroRNAs as a Potential New Preventive Approach in the Transition from Asymptomatic to Symptomatic Multiple Myeloma Disease. Cancers (Basel) 2021; 13:cancers13153650. [PMID: 34359551 PMCID: PMC8344971 DOI: 10.3390/cancers13153650] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Multiple myeloma (MM) is the second most common haematologic malignancy, and it remains an incurable disease despite the advances of novel therapies. It is characterised by a multistep process that arises from a pre-malignant asymptomatic status-defined monoclonal gammopathy of undetermined significance (MGUS), evolves to a middle stage named smouldering myeloma phase (SMM), and culminates in the active disease (MM). Identification of early and non-invasive markers of the disease progression is currently an active field of investigation. In this review, we discuss the role and significance of microRNAs (miRNAs) as potential diagnostic biomarkers to predict the clinical transition from MGUS/SMM status to MM. Abstract Multiple myeloma (MM) is a hematological malignancy characterised by proliferation of clonal plasma cells (PCs) within the bone marrow (BM). Myelomagenesis is a multi-step process which goes from an asymptomatic phase, defined as monoclonal gammopathy of undetermined significance (MGUS), to a smouldering myeloma (SMM) stage, to a final active MM disease, characterised by hypercalcemia, renal failure, bone lesions anemia, and higher risk of infections. Overall, microRNAs (miRNAs) have shown to significantly impact on MM tumorigenesis, as a result of miRNA-dependent modulation of genes involved in pathways known to be crucial for MM pathogenesis and disease progression. We aim to revise the literature related to the role of miRNAs as potential diagnostic and prognostic biomarkers, thus highlighting their key role as novel players within the field of MM and related premalignant conditions.
Collapse
Affiliation(s)
- Vanessa Desantis
- Unit of Internal Medicine and Clinical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.D.); (A.G.S.); (I.S.); (A.L.); (A.M.)
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Antonio Giovanni Solimando
- Unit of Internal Medicine and Clinical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.D.); (A.G.S.); (I.S.); (A.L.); (A.M.)
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Ilaria Saltarella
- Unit of Internal Medicine and Clinical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.D.); (A.G.S.); (I.S.); (A.L.); (A.M.)
| | - Antonio Sacco
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.S.); (V.G.)
| | - Viviana Giustini
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.S.); (V.G.)
| | - Marta Bento
- Centro Hospitalar Lisboa Norte, Department of Hematology and Transplantation, Institute of Molecular Medicine, University of Lisbon, 1649-035 Lisbon, Portugal;
| | - Aurelia Lamanuzzi
- Unit of Internal Medicine and Clinical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.D.); (A.G.S.); (I.S.); (A.L.); (A.M.)
| | - Assunta Melaccio
- Unit of Internal Medicine and Clinical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.D.); (A.G.S.); (I.S.); (A.L.); (A.M.)
| | - Maria Antonia Frassanito
- Unit of General Pathology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Angelo Paradiso
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, Medical School, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Angelo Vacca
- Unit of Internal Medicine and Clinical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.D.); (A.G.S.); (I.S.); (A.L.); (A.M.)
- Correspondence: (A.V.); (A.M.R.)
| | - Aldo M. Roccaro
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (A.S.); (V.G.)
- Correspondence: (A.V.); (A.M.R.)
| |
Collapse
|
11
|
Giannakoulas N, Ntanasis-Stathopoulos I, Terpos E. The Role of Marrow Microenvironment in the Growth and Development of Malignant Plasma Cells in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms22094462. [PMID: 33923357 PMCID: PMC8123209 DOI: 10.3390/ijms22094462] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/16/2022] Open
Abstract
The development and effectiveness of novel therapies in multiple myeloma have been established in large clinical trials. However, multiple myeloma remains an incurable malignancy despite significant therapeutic advances. Accumulating data have elucidated our understanding of the genetic background of the malignant plasma cells along with the role of the bone marrow microenvironment. Currently, the interaction among myeloma cells and the components of the microenvironment are considered crucial in multiple myeloma pathogenesis. Adhesion molecules, cytokines and the extracellular matrix play a critical role in the interplay among genetically transformed clonal plasma cells and stromal cells, leading to the proliferation, progression and survival of myeloma cells. In this review, we provide an overview of the multifaceted role of the bone marrow microenvironment in the growth and development of malignant plasma cells in multiple myeloma.
Collapse
Affiliation(s)
- Nikolaos Giannakoulas
- Department of Hematology of University Hospital of Larisa, Faculty of Medicine, University of Thessaly, 41110 Larisa, Greece;
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
- Correspondence:
| |
Collapse
|
12
|
Targeted Therapies for Multiple Myeloma. J Pers Med 2021; 11:jpm11050334. [PMID: 33922567 PMCID: PMC8145732 DOI: 10.3390/jpm11050334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/30/2022] Open
Abstract
Multiple myeloma continues to be a challenging disorder to treat despite improved therapies and the widespread use of proteasome inhibitors and immunomodulatory drugs. Although patient outcomes have improved, the disease continues to invariably relapse, and in the majority of cases, a cure remains elusive. In the last decade, there has been an explosion of novel drugs targeting cellular proteins essential for malignant plasma cell proliferation and survival. In this review, we focus on novel druggable targets leading to the development of monoclonal antibodies and cellular therapies against surface antigens (CD38, CD47, CD138, BCMA, SLAMF7, GPRC5D, FcRH5), inhibitors of epigenetic regulators such as histone deacetylase (HDAC), and agents targeting anti-apoptotic (BCL-2), ribosomal (eEF1A2) and nuclear export (XPO1) proteins.
Collapse
|
13
|
Telomere Architecture Correlates with Aggressiveness in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13081969. [PMID: 33921898 PMCID: PMC8073772 DOI: 10.3390/cancers13081969] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Multiple myeloma (MM) remains an incurable blood cancer. One of the current challenges in patient management is the risk assessment and subsequent treatment management for each patient with MM. Patients with an identical diagnosis may present very different disease courses and outcomes. This challenge of MM is a current focus of the scientific and medical communities. In our research, we have used an imaging approach to determine the risk of MM patients to progressive/aggressive disease. Using three-dimensional (3D) imaging of telomeres, the ends of chromosomes, we report that specific telomeric profiles are associated with aggressive disease. Abstract The prognosis of multiple myeloma (MM), an incurable B-cell malignancy, has significantly improved through the introduction of novel therapeutic modalities. Myeloma prognosis is essentially determined by cytogenetics, both at diagnosis and at disease progression. However, for a large cohort of patients, cytogenetic analysis is not always available. In addition, myeloma patients with favorable cytogenetics can display an aggressive clinical course. Therefore, it is necessary to develop additional prognostic and predictive markers for this disease to allow for patient risk stratification and personalized clinical decision-making. Genomic instability is a prominent characteristic in MM, and we have previously shown that the three-dimensional (3D) nuclear organization of telomeres is a marker of both genomic instability and genetic heterogeneity in myeloma. In this study, we compared in a longitudinal prospective study blindly the 3D telomeric profiles from bone marrow samples of 214 initially treatment-naïve patients with either monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), or MM, with a minimum follow-up of 5 years. Here, we report distinctive 3D telomeric profiles correlating with disease aggressiveness and patient response to treatment in MM patients, and also distinctive 3D telomeric profiles for disease progression in smoldering multiple myeloma patients. In particular, lower average intensity (telomere length, below 13,500 arbitrary units) and increased number of telomere aggregates are associated with shorter survival and could be used as a prognostic factor to identify high-risk SMM and MM patients.
Collapse
|
14
|
Liu Y, Jelloul F, Zhang Y, Bhavsar T, Ho C, Rao M, Lewis NE, Cimera R, Baik J, Sigler A, Sen F, Yabe M, Roshal M, Landgren O, Dogan A, Xiao W. Genetic Basis of Extramedullary Plasmablastic Transformation of Multiple Myeloma. Am J Surg Pathol 2020; 44:838-848. [PMID: 32118627 DOI: 10.1097/pas.0000000000001459] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In patients with multiple myeloma, plasmablastic transformation in the bone marrow is rare and associated with poor outcomes. The significance of discordant extramedullary plasmablastic transformation in patients with small, mature clonal plasma cells in the bone marrow has not been well studied. Here, we report the clinicopathologic, cytogenetic, and molecular features of 10 such patients (male/female: 6/4, median age: 65 y, range: 48 to 76 y) with an established diagnosis of multiple myeloma in the bone marrow composed of small, mature plasma cells in parallel with a concurrent or subsequent extramedullary plasmablastic transformation. Eight patients with available survival data showed an overall aggressive clinical course with a median survival of 4.5 months after the diagnosis of extramedullary plasmablastic transformation, despite aggressive treatment and even in patients with low-level bone marrow involvement. Pathologically, the extramedullary plasmablastic myeloma were clonally related to the corresponding bone marrow plasma cells, showed high levels of CMYC and/or P53 expression with a high Ki-67 proliferation index by immunohistochemistry and harbored more complex genomic aberrations including frequent mutations in the RAS pathway and MYC rearrangements compared with their bone marrow counterparts. In summary, although genetic and immunohistochemical studies were not uniformly performed on all cases due to the retrospective nature of this study, our data suggest that discordant extramedullary plasmablastic transformation of multiple myeloma has an aggressive clinical course and is characterized by frequent mutations in the RAS pathway and more complex genomic abnormalities.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pathology, Hematopathology Service
| | | | | | - Tapan Bhavsar
- Department of Pathology, Wayne State University School of Medicine, Detroit Receiving Hospital, Detroit, MI
| | - Caleb Ho
- Department of Pathology, Hematopathology Service.,Department of Pathology, Diagnostic Molecular Pathology Service
| | - Mamta Rao
- Department of Pathology, Cytogenetic Laboratory
| | | | | | - Jeeyeon Baik
- Department of Pathology, Hematopathology Service
| | | | - Filiz Sen
- Department of Pathology, Hematopathology Service
| | - Mariko Yabe
- Department of Pathology, Hematopathology Service
| | | | - Ola Landgren
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Dogan
- Department of Pathology, Hematopathology Service
| | - Wenbin Xiao
- Department of Pathology, Hematopathology Service
| |
Collapse
|
15
|
Huang J, Huang LQ, He HS, Yan J, Huang C, Wang R, Guan Y, Huang DP. Overexpression of heme oxygenase-1 in bone marrow stromal cells promotes multiple myeloma resistance through the JAK2/STAT3 pathway. Life Sci 2020; 257:118088. [PMID: 32663573 DOI: 10.1016/j.lfs.2020.118088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/09/2020] [Accepted: 07/08/2020] [Indexed: 01/16/2023]
Abstract
AIMS Bone marrow stromal cells (BMSCs) have been reported to interact with multiple myeloma (MM) and exert a vital function of the survival of MM cells. Heme oxygenase-1 (HO-1), a cytoprotective enzyme, has the potential to become a hematological malignancies targeted gene. This study aimed to investigate the role of HO-1 in MM resistance of BMSCs and its possible mechanisms. MAIN METHODS In this study, the expression of related proteins was detected by RT-qPCR and Western blot. HO-1 expression was regulated by lentivirus transfection. Cell viability and apoptosis were detected by Flow cytometry and CCK-8. Cytokine secretion was assayed by ELISA. The survival and carcinogenic abilities was detected by clone formation assay. KEY FINDINGS HO-1 expression in the BMSCs of stage III MM patients was substantially increased, compared with that of healthy donors and stage I/II patients. The results of co-culture of BMSCs and MM cells indicated that, the upregulated HO-1 inhibited the apoptosis of co-cultured MM cells, while downregulated HO-1 promoted the chemosensitivity of co-cultured MM cells, moreover, the upregulated HO-1 in BMSCs increased the colony-formation ability of MM cells. This protective capability may be regulated by CXCL12/CXCR4 signaling. High HO-1 expression in BMSCs can promote the phosphorylation of the JAK2/STAT3 pathway, thereby increasing secretion of SDF-1 in BMSCs and activating CXCL12/CXCR4 signaling. In addition, direct contact between BMSCs and MM cells may cause drug resistance. SIGNIFICANCE These results indicated that the regulation of HO-1 in BMSCs may be a new effective method of MM therapy.
Collapse
Affiliation(s)
- Jun Huang
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China
| | - Lai-Quan Huang
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China
| | - He-Sheng He
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China
| | - Jiawei Yan
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China
| | - Chen Huang
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China
| | - Ran Wang
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China
| | - Yan Guan
- Wannan Medical College, Wuhu 241001, China
| | - Dong-Ping Huang
- Department of Hematology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu 241001, China.
| |
Collapse
|
16
|
Kapoor R, Kumar R, Dubey AP. Risk Stratification in Multiple Myeloma in Indian Settings. Indian J Hematol Blood Transfus 2020; 36:464-472. [PMID: 32647419 DOI: 10.1007/s12288-019-01240-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/06/2019] [Indexed: 01/03/2023] Open
Abstract
Multiple myeloma (MM) constitutes 10% of all hematological malignancies. The last one decade has seen a phenomenal progress in the therapeutic options available for the management. Although it still remains incurable, with the advent of newer therapies, the median survival in many risk groups is now around 10 years. Conventional karyotyping of bone marrow samples has a positivity rate of 20-30% at diagnosis in patients of Multiple Myeloma. However, array Comparative Genomic Hybridisation (aCGH) has revealed that almost all MM patients have cytogenetic abnormalities which may affect the pathophysiology, selection of therapy and outcomes of the disease. The progress in the field of exploring the genetic landscape of multiple myeloma with multiple tools like Fluorescent in-situ hybridization, aCGH, Next Generation Sequencing, Flow cytometry, etc., combined with the traditional risk stratification markers like albumin, β2 microglobulin and LDH, is gradually leading towards a risk-adapted therapy. The recent R-ISS risk stratification has combined these two group of information to validate a prognostic score which is an improvement over the past tools like DSS and ISS. In view of the plethora of information available on the multitude of cytogenetic markers there is a tendency to evaluate for all of them at diagnosis, especially in research centers. This leads to a significant increase in the cost of therapy of Multiple Myeloma in day-to-day clinical practice and an increased out-of-pocket spending to the patient, especially in resource-limited settings like India. Also, there is a variable approach to pre-therapy cytogenetic evaluation and risk stratification at different Hematology centres in the country, often dictated by financial constraints and availability of specialized tests. This review discusses the risk stratification markers and tools available in MM in 2019 and how it can be adapted in the resource constraint settings so as to derive the maximum prognostic information from a minimal prognostic panel, as well as lead to standardization of the prognostic protocols in resource limited settings across various Hematology centres in India.
Collapse
Affiliation(s)
- Rajan Kapoor
- Medicine and Clinical Hematology, Command Hospital (EC), Kolkata, India
| | - Rajiv Kumar
- Department of Medicine and Clinical Hematology, INHS Asvini, R C Church, Colaba, Mumbai, 400005 India
| | - A P Dubey
- Medical Oncology, Clear Medi Hospitals and Cancer Centre, Ghaziabad, UP India
| |
Collapse
|
17
|
Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e752-e768. [PMID: 32651110 DOI: 10.1016/j.clml.2020.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of the plasma cells within the bone marrow (BM). Studies have shown that the cellular and noncellular components of the BM milieu, such as cytokines and exosomes, play an integral role in MM pathogenesis and progression by mediating drug resistance and inducing MM proliferation. Moreover, the BM microenvironment of patients with MM facilitates cancer tolerance and immune evasion through the expansion of regulatory immune cells, inhibition of antitumor effector cells, and disruption of the antigen presentation machinery. These are of special relevance, especially in the current era of cancer immunotherapy. An improved understanding of the supportive role of the MM BM microenvironment will allow for the development of future therapies targeting MM in the context of the BM milieu to elicit deeper and more durable responses. In the present review, we have discussed our current understanding of the role of the BM microenvironment in MM progression and resistance to therapy and discuss novel potential approaches to alter its pro-MM function.
Collapse
|
18
|
Méndez-Ferrer S, Bonnet D, Steensma DP, Hasserjian RP, Ghobrial IM, Gribben JG, Andreeff M, Krause DS. Bone marrow niches in haematological malignancies. Nat Rev Cancer 2020; 20:285-298. [PMID: 32112045 PMCID: PMC9912977 DOI: 10.1038/s41568-020-0245-2] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
Abstract
Haematological malignancies were previously thought to be driven solely by genetic or epigenetic lesions within haematopoietic cells. However, the niches that maintain and regulate daily production of blood and immune cells are now increasingly being recognized as having an important role in the pathogenesis and chemoresistance of haematological malignancies. Within haematopoietic cells, the accumulation of a small number of recurrent mutations initiates malignancy. Concomitantly, specific alterations of the niches, which support haematopoietic stem cells and their progeny, can act as predisposition events, facilitating mutant haematopoietic cell survival and expansion as well as contributing to malignancy progression and providing protection of malignant cells from chemotherapy, ultimately leading to relapse. In this Perspective, we summarize our current understanding of the composition and function of the specialized haematopoietic niches of the bone marrow during health and disease. We discuss disease mechanisms (rather than malignancy subtypes) to provide a comprehensive description of key niche-associated pathways that are shared across multiple haematological malignancies. These mechanisms include primary driver mutations in bone marrow niche cells, changes associated with increased hypoxia, angiogenesis and inflammation as well as metabolic reprogramming by stromal niche cells. Consequently, remodelling of bone marrow niches can facilitate immune evasion and activation of survival pathways favouring malignant haematopoietic cell maintenance, defence against excessive reactive oxygen species and protection from chemotherapy. Lastly, we suggest guidelines for the handling and biobanking of patient samples and analysis of the niche to ensure that basic research identifying therapeutic targets can be more efficiently translated to the clinic. The hope is that integrating knowledge of how bone marrow niches contribute to haematological disease predisposition, initiation, progression and response to therapy into future clinical practice will likely improve the treatment of these disorders.
Collapse
Affiliation(s)
- Simón Méndez-Ferrer
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Cambridge, UK.
- National Health Service Blood and Transplant, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - David P Steensma
- Harvard Medical School, Boston, MA, USA
- The Center for Prevention of Progression of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Robert P Hasserjian
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Irene M Ghobrial
- Harvard Medical School, Boston, MA, USA
- The Center for Prevention of Progression of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Medicine, Frankfurt, Germany
- Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
19
|
Ishibashi M, Takahashi R, Tsubota A, Sasaki M, Handa H, Imai Y, Tanaka N, Tsukune Y, Tanosaki S, Ito S, Asayama T, Sunakawa M, Kaito Y, Kuribayashi-Hamada Y, Onodera A, Moriya K, Komatsu N, Tanaka J, Odajima T, Sugimori H, Inokuchi K, Tamura H. SLAMF3-Mediated Signaling via ERK Pathway Activation Promotes Aggressive Phenotypic Behaviors in Multiple Myeloma. Mol Cancer Res 2020; 18:632-643. [PMID: 31974290 DOI: 10.1158/1541-7786.mcr-19-0391] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 12/03/2019] [Accepted: 01/16/2020] [Indexed: 11/16/2022]
Abstract
The signaling lymphocytic activation molecule family 3 (SLAMF3) is a member of the immunoglobulin superfamily expressed on T, B, and natural killer cells and modulates the activation and cytotoxicity of these cells. SLAMF3 is also expressed on plasma cells from patients with multiple myeloma (MM), although its role in MM pathogenesis remains unclear. This study found that SLAMF3 is highly and constitutively expressed on MM cells regardless of disease stage and that SLAMF3 knockdown/knockout suppresses proliferative potential and increases drug-induced apoptosis with decreased levels of phosphorylated ERK protein in MM cells. SLAMF3-overexpressing MM cells promote aggressive myeloma behavior in comparison with cytoplasmic domain-truncated SLAMF3 (ΔSLAMF3) cells. SLAMF3 interacts directly with adaptor proteins SH2 domain-containing phosphatase 2 (SHP2) and growth factor receptor bound 2 (GRB2), which also interact with each other. SLAMF3 knockdown, knockout, ΔSLAMF3, and SHP2 inhibitor-treated MM cells decreased phosphorylated ERK protein levels. Finally, serum soluble SLAMF3 (sSLAMF3) levels were markedly increased in advanced MM. Patients with high levels of sSLAMF3 progressed to the advanced stage significantly more often and had shorter progression-free survival times than those with low levels. This study revealed that SLAMF3 molecules consistently expressed on MM cells transmit MAPK/ERK signals mediated via the complex of SHP2 and GRB2 by self-ligand interaction between MM cells and induce a high malignant potential in MM. Furthermore, high levels of serum sSLAMF3 may reflect MM disease progression and be a useful prognostic factor. IMPLICATIONS: SLAMF3 may be a new therapeutic target for immunotherapy and novel agents such as small-molecule inhibitors.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Department of Hematology, Nippon Medical School, Tokyo, Japan.,Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Risa Takahashi
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Asako Tsubota
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Makoto Sasaki
- Division of Hematology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiroshi Handa
- Department of Hematology, Gunma University, Gunma, Japan
| | - Yoichi Imai
- Department of Hematology and Oncology, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Norina Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yutaka Tsukune
- Division of Hematology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Sakae Tanosaki
- Department of Hematology, The Fraternity Memorial Hospital, Tokyo, Japan
| | - Shigeki Ito
- Department of Hematology/Oncology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Toshio Asayama
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Mika Sunakawa
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Yuta Kaito
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | | | - Asaka Onodera
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Keiichi Moriya
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Norio Komatsu
- Division of Hematology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | - Junji Tanaka
- Department of Hematology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takeshi Odajima
- Faculty of Health Science, Daito Bunka University School of Sports and Health Science, Saitama, Japan
| | - Hiroki Sugimori
- Department of Preventive Medicine, Daito Bunka University Graduate School of Sports and Health Science, Saitama, Japan
| | - Koiti Inokuchi
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Hideto Tamura
- Department of Hematology, Nippon Medical School, Tokyo, Japan.
| |
Collapse
|
20
|
D'Souza L, Bhattacharya D. Plasma cells: You are what you eat. Immunol Rev 2019; 288:161-177. [PMID: 30874356 DOI: 10.1111/imr.12732] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022]
Abstract
Plasma cells are terminally differentiated B lymphocytes that constitutively secrete antibodies. These antibodies can provide protection against pathogens, and their quantity and quality are the best clinical correlates of vaccine efficacy. As such, plasma cell lifespan is the primary determinant of the duration of humoral immunity. Yet dysregulation of plasma cell function can cause autoimmunity or multiple myeloma. The longevity of plasma cells is primarily dictated by nutrient uptake and non-transcriptionally regulated metabolic pathways. We have previously shown a positive effect of glucose uptake and catabolism on plasma cell longevity and function. In this review, we discuss these findings with an emphasis on nutrient uptake and its effects on respiratory capacity, lifespan, endoplasmic reticulum stress, and antibody secretion in plasma cells. We further discuss how some of these pathways may be dysregulated in multiple myeloma, potentially providing new therapeutic targets. Finally, we speculate on the connection between plasma cell intrinsic metabolism and systemic changes in nutrient availability and metabolic diseases.
Collapse
Affiliation(s)
- Lucas D'Souza
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
21
|
A multiple myeloma classification system that associates normal B-cell subset phenotypes with prognosis. Blood Adv 2019; 2:2400-2411. [PMID: 30254104 DOI: 10.1182/bloodadvances.2018018564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Despite the recent progress in treatment of multiple myeloma (MM), it is still an incurable malignant disease, and we are therefore in need of new risk stratification tools that can help us to understand the disease and optimize therapy. Here we propose a new subtyping of myeloma plasma cells (PCs) from diagnostic samples, assigned by normal B-cell subset associated gene signatures (BAGS). For this purpose, we combined fluorescence-activated cell sorting and gene expression profiles from normal bone marrow (BM) Pre-BI, Pre-BII, immature, naïve, memory, and PC subsets to generate BAGS for assignment of normal BM subtypes in diagnostic samples. The impact of the subtypes was analyzed in 8 available data sets from 1772 patients' myeloma PC samples. The resulting tumor assignments in available clinical data sets exhibited similar BAGS subtype frequencies in 4 cohorts from de novo MM patients across 1296 individual cases. The BAGS subtypes were significantly associated with progression-free and overall survival in a meta-analysis of 916 patients from 3 prospective clinical trials. The major impact was observed within the Pre-BII and memory subtypes, which had a significantly inferior prognosis compared with other subtypes. A multiple Cox proportional hazard analysis documented that BAGS subtypes added significant, independent prognostic information to the translocations and cyclin D classification. BAGS subtype analysis of patient cases identified transcriptional differences, including a number of differentially spliced genes. We identified subtype differences in myeloma at diagnosis, with prognostic impact and predictive potential, supporting an acquired B-cell trait and phenotypic plasticity as a pathogenetic hallmark of MM.
Collapse
|
22
|
Scherger AK, Al-Maarri M, Maurer HC, Schick M, Maurer S, Öllinger R, Gonzalez-Menendez I, Martella M, Thaler M, Pechloff K, Steiger K, Sander S, Ruland J, Rad R, Quintanilla-Martinez L, Wunderlich FT, Rose-John S, Keller U. Activated gp130 signaling selectively targets B cell differentiation to induce mature lymphoma and plasmacytoma. JCI Insight 2019; 4:128435. [PMID: 31391340 DOI: 10.1172/jci.insight.128435] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/09/2019] [Indexed: 12/22/2022] Open
Abstract
Aberrant activity of the glycoprotein 130 130/JAK/STAT3 (gp130/JAK/STAT3) signaling axis is a recurrent event in inflammation and cancer. In particular, it is associated with a wide range of hematological malignancies, including multiple myeloma and leukemia. Novel targeted therapies have only been successful for some subtypes of these malignancies, underlining the need for developing robust mouse models to better dissect the role of this pathway in specific tumorigenic processes. Here, we investigated the role of selective gp130/JAK/STAT3 activation by generating a conditional mouse model. This model targeted constitutively active, cell-autonomous gp130 activity to B cells, as well as to the entire hematopoietic system. We found that regardless of the timing of activation in B cells, constitutively active gp130 signaling resulted in the formation specifically of mature B cell lymphomas and plasma cell disorders with full penetrance, only with different latencies, where infiltrating CD138+ cells were a dominant feature in every tumor. Furthermore, constitutively active gp130 signaling in all adult hematopoietic cells also led to the development specifically of largely mature, aggressive B cell cancers, again with a high penetrance of CD138+ tumors. Importantly, gp130 activity abrogated the differentiation block induced by a B cell-targeted Myc transgene and resulted in a complete penetrance of the gp130-associated, CD138+, mature B cell lymphoma phenotype. Thus, gp130 signaling selectively provides a strong growth and differentiation advantage for mature B cells and directs lymphomagenesis specifically toward terminally differentiated B cell cancers.
Collapse
Affiliation(s)
- Anna K Scherger
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Mona Al-Maarri
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Preventive Medicine and Diabetes, Cologne, Germany
| | | | - Markus Schick
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Sabine Maurer
- Internal Medicine III, Technische Universität München, Munich, Germany
| | - Rupert Öllinger
- Internal Medicine II.,Center for Translational Cancer Research, and.,Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany
| | | | - Manuela Martella
- Institute of Pathology, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Markus Thaler
- Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany
| | - Konstanze Pechloff
- Center for Translational Cancer Research, and.,Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Katja Steiger
- German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,Institute of Pathology, Technische Universität München, Munich, Germany
| | - Sandrine Sander
- Adaptive Immunity and Lymphoma, German Cancer Research Center/National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Jürgen Ruland
- Center for Translational Cancer Research, and.,Institute of Clinical Chemistry and Pathobiochemistry, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Roland Rad
- Internal Medicine II.,Center for Translational Cancer Research, and.,Institute of Molecular Oncology and Functional Genomics, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | | | - Frank T Wunderlich
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Preventive Medicine and Diabetes, Cologne, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Ulrich Keller
- Internal Medicine III, Technische Universität München, Munich, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany.,Department of Hematology, Oncology and Tumor Immunology (Campus Benjamin Franklin), Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
23
|
Zhang ZY, Li YC, Geng CY, Zhou HX, Gao W, Chen WM. Serum exosomal microRNAs as novel biomarkers for multiple myeloma. Hematol Oncol 2019; 37:409-417. [PMID: 31102419 DOI: 10.1002/hon.2639] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/29/2019] [Accepted: 05/14/2019] [Indexed: 12/11/2022]
Abstract
Accumulating studies have focused on circulating microRNAs, which might be potential biomarkers for different malignancies. The aim of this study was to investigate the potential of serum exosomal microRNAs to be novel serum biomarkers for smouldering myeloma (SMM) or even multiple myeloma (MM). The levels of serum exosomal microRNAs and serum circulating microRNAs were measured in healthy individuals and patients with SMM (n = 20) or MM (n = 20). Serum exosomal microRNAs and serum circulating microRNAs were extracted from serum, and the expression levels of selected microRNAs were quantified by real-time polymerase chain reaction (PCR). The levels of serum exosome-derived miR-20a-5p, miR-103a-3p, and miR-4505 were significantly different among patients with MM, patients with SMM, and healthy individuals, while there were differences in the levels of let-7c-5p, miR-185-5p, and miR-4741 in patients with MM relative to those in SMM patients or healthy controls. Additionally, a significant correlation was rarely found between the levels of serum and exosomal microRNAs. This study shows that serum exosomal microRNAs can be used independently as novel serum biomarkers for MM.
Collapse
Affiliation(s)
- Zhi-Yao Zhang
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yan-Chen Li
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Chuan-Ying Geng
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hui-Xing Zhou
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wen Gao
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wen-Ming Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Harmer D, Falank C, Reagan MR. Interleukin-6 Interweaves the Bone Marrow Microenvironment, Bone Loss, and Multiple Myeloma. Front Endocrinol (Lausanne) 2019; 9:788. [PMID: 30671025 PMCID: PMC6333051 DOI: 10.3389/fendo.2018.00788] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
The immune system is strongly linked to the maintenance of healthy bone. Inflammatory cytokines, specifically, are crucial to skeletal homeostasis and any dysregulation can result in detrimental health complications. Interleukins, such as interleukin 6 (IL-6), act as osteoclast differentiation modulators and as such, must be carefully monitored and regulated. IL-6 encourages osteoclastogenesis when bound to progenitors and can cause excessive osteoclastic activity and osteolysis when overly abundant. Numerous bone diseases are tied to IL-6 overexpression, including rheumatoid arthritis, osteoporosis, and bone-metastatic cancers. In the latter, IL-6 can be released with growth factors into the bone marrow microenvironment (BMM) during osteolysis from bone matrix or from cancer cells and osteoblasts in an inflammatory response to cancer cells. Thus, IL-6 helps create an ideal microenvironment for oncogenesis and metastasis. Multiple myeloma (MM) is a blood cancer that homes to the BMM and is strongly tied to overexpression of IL-6 and bone loss. The roles of IL-6 in the progression of MM are discussed in this review, including roles in bone homing, cancer-associated bone loss, disease progression and drug resistance. MM disease progression often includes the development of drug-resistant clones, and patients commonly struggle with reoccurrence. As such, therapeutics that specifically target the microenvironment, rather than the cancer itself, are ideal and IL-6, and its myriad of downstream signaling partners, are model targets. Lastly, current and potential therapeutic interventions involving IL-6 and connected signaling molecules are discussed in this review.
Collapse
Affiliation(s)
- Danielle Harmer
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
| | - Carolyne Falank
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Michaela R. Reagan
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- School of Medicine, Tufts University, Boston, MA, United States
| |
Collapse
|
25
|
Ding W, Tan Y, Qian Y, Xue W, Wang Y, Xi C, Gu K, Xu Y, Xu X. Primary plasmablastic plasmacytoma in the stomach of an immunocompetent adult: A case report. Medicine (Baltimore) 2019; 98:e14235. [PMID: 30681607 PMCID: PMC6358359 DOI: 10.1097/md.0000000000014235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/24/2018] [Accepted: 01/01/2019] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Extramedullary plasmacytomas (EMP) are tumors composed by a monoclonal population of plasma cells that arise in extraosseus tissues, occupying <5% of all plasma cell neoplasms. Gastrointestinal solitary extramedullary plasmacytoma is rare, just comprises about 5% of all EMPs. The most common site is small intestine. The gastric incidence is much rare and especially the plasmablastic plasmacytoma in the stomach. PATIENT CONCERNS A 65-year-old man had an epigastric discomfort and mass for about 2 months. Gastroscopy revealed a malignant tumor in the gastric body. Abdomen computed tomography (CT) showed that the gastric cavity was filled, and the irregular soft tissue shadow was seen in the greater curvature, and the enhancement was obvious. To get more tissue, we conducted stomach puncture biopsy. Pathology showed small-round cell malignant tumors. And immunohistochemical examinations revealed that the tumor tend to be a plasma cell tumor. DIAGNOSIS Gastric plasma cell tumor. INTERVENTIONS Distal gastrectomy was performed to treat the tumor. OUTCOMES In addition to ascites caused by hypoproteinemia, there were no postoperative complications. Postoperative pathologic report showed plasmablastic plasmacytoma. Histopathologic examination of the specimen revealed plasmablastic plasmacytoma originating in the stomach with transmural extension, but without lymph node metastasis. The patient is regularly followed up at a postoperative clinic and is doing well, and at present there is no plan for adjuvant treatment. LESSONS Surgical resection is good option for gastrointestinal EMP.
Collapse
Affiliation(s)
| | | | - Yan Qian
- Department of Respiration, Changzhou Second People's Hospital Affiliated to Nanjing Medical University
| | | | | | | | - Kefeng Gu
- Department of Pathology, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu Province, China
| | | | | |
Collapse
|
26
|
Carmel B, Delost GR, Stern J, Honda K. Cutaneous plasmacytoma: Metastasis of multiple myeloma and invasion of sternotomy scar. JAAD Case Rep 2018; 5:94-97. [PMID: 30581944 PMCID: PMC6297258 DOI: 10.1016/j.jdcr.2018.10.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Benjamin Carmel
- Lake Erie College of Osteopathic Medicine, Bradenton, Florida
| | - Gregory R Delost
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Jason Stern
- Department of Hematology & Oncology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Kord Honda
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
27
|
Kim C, Lee SG, Yang WM, Arfuso F, Um JY, Kumar AP, Bian J, Sethi G, Ahn KS. Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Lett 2018; 431:123-141. [DOI: 10.1016/j.canlet.2018.05.038] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 01/13/2023]
|
28
|
Fresco R, Saldombide L, Suárez L. Synchronous Presentation of an Askin Tumor and a Plasmacytoma in an Adult Patient. TUMORI JOURNAL 2018; 89:324-7. [PMID: 12908792 DOI: 10.1177/030089160308900318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Askin tumor, or malignant small round cell tumor of the thoracopulmonary region, is an extremely infrequent entity occurring primarily in children and adolescents. Its histopathologic and cytogenetic features suggest that it belongs to the family of Ewing's sarcoma and primitive neuroectodermal tumors. We report the case of a 43-year-old woman affected by an Askin tumor with bone metastases at diagnosis, presenting synchronously with a plasmacytoma. This is the first reported case of the simultaneous occurrence of an Askin tumor and a malignant hemopathy. The progression of the former and the remission of the plasmacytoma during chemotherapy were remarkable, since Askin tumor treatment shares drugs used for the treatment of plasma cell tumors. Given the infrequent presentation of these diseases in a young adult and the coexistence of two neoplasias characterized by typical chromosomal abnormalities, we consider the possibility of a genetic cancer susceptibility in our patient.
Collapse
Affiliation(s)
- Rodrigo Fresco
- Servicio de Oncología Clínica, Hospital de Clínicas, Facultad de Medicina, Montevideo, Uruguay.
| | | | | |
Collapse
|
29
|
Byun JM, Shin DY, Hong J, Kim I, Kim HK, Lee DS, Koh Y, Yoon SS. Distinct predictive impact of FISH abnormality in proteasome inhibitors and immunomodulatory agents response: redefining high-risk multiple myeloma in Asian patients. Cancer Med 2018; 7:831-841. [PMID: 29380550 PMCID: PMC5852362 DOI: 10.1002/cam4.1340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/27/2017] [Accepted: 12/21/2017] [Indexed: 12/25/2022] Open
Abstract
For risk-adaptive therapeutic approaches in multiple myeloma (MM) treatment, we analyzed treatment outcome according to in situ hybridization (FISH) profiles to investigate the prognostic and predictive values of structural variations in a large series of Asian population. A total of 565 newly diagnosed patients with multiple myeloma between January 2005 and June 2015 were evaluated. FISH results showed del(17p13) in 8.8% (29/331), del(13q14) in 35.5% (184/519), t(14;16) in 2.5% (8/326), t(4;14) in 27.9% (109/390), IgH rearrangement in 47.7% (248/520), and +1q21 in 40.8% (211/517). The presence of del(17p13), IgH rearrangement, and t(14;16) was associated with worse overall survival. Interestingly, however, the presence of t(4;14) conferred little prognostic impact. Treatment-specific analysis revealed the presence of del(17p13), t(14;16), IgH rearrangement, and trisomy 1q21 was predictive of unsatisfactory response to bortezomib. On the other hand, patients with del(13q14) and del(9p21) were less likely to benefit from lenalidomide. Autologous stem cell transplantation (autoSCT) was less effective in patients with del(17p13), t(14;16), and trisomy 1q21. Predictive values of del(17p13) and t(14;16) to bortezomib and autoSCT are seemingly universal, but predictive marker del(13q14) and del(9p21) for lenalidomide response appears ethnicity-specific. Thus, FISH profiles in MM treatment should be interpreted with regards to patient's ethnicity.
Collapse
Affiliation(s)
- Ja Min Byun
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Inho Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hyun Kyung Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Soon Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
30
|
Kim SJ, Kim ES, Kim S, Uhm J, Won YW, Park BB, Choi JH, Lee YY. Antitumoral effect of arsenic compound, sodium metaarsenite (KML001), on multiple myeloma cells. Int J Oncol 2017; 51:1739-1746. [DOI: 10.3892/ijo.2017.4161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/21/2017] [Indexed: 11/06/2022] Open
|
31
|
Inflammatory and Anti-Inflammatory Equilibrium, Proliferative and Antiproliferative Balance: The Role of Cytokines in Multiple Myeloma. Mediators Inflamm 2017; 2017:1852517. [PMID: 29089667 PMCID: PMC5635476 DOI: 10.1155/2017/1852517] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is typically exemplified by a desynchronized cytokine system with increased levels of inflammatory cytokines. We focused on the contrast between inflammatory and anti-inflammatory systems by assessing the role of cytokines and their influence on MM. The aim of this review is to summarize the available information to date concerning this equilibrium to provide an overview of the research exploring the roles of serum cytokines in MM. However, the association between MM and inflammatory cytokines appears to be inadequate, and other functions, such as pro-proliferative or antiproliferative effects, can assume the role of cytokines in the genesis and progression of MM. It is possible that inflammation, when guided by cancer-specific Th1 cells, may inhibit tumour onset and progression. In a Th1 microenvironment, proinflammatory cytokines (e.g., IL-6 and IL-1) may contribute to tumour eradication by attracting leucocytes from the circulation and by increasing CD4 + T cell activity. Hence, caution should be used when considering therapies that target factors with pro- or anti-inflammatory activity. Drugs that may reduce the tumour-suppressive Th1-driven inflammatory immune response should be avoided. A better understanding of the relationship between inflammation and myeloma will ensure more effective therapeutic interventions.
Collapse
|
32
|
Wang W, Zhang Y, Chen R, Tian Z, Zhai Y, Janz S, Gu C, Yang Y. Chromosomal instability and acquired drug resistance in multiple myeloma. Oncotarget 2017; 8:78234-78244. [PMID: 29100463 PMCID: PMC5652852 DOI: 10.18632/oncotarget.20829] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/26/2017] [Indexed: 12/29/2022] Open
Abstract
Chromosomal instability (CIN) is an important hallmark of human cancer. CIN not only contributes to all stages of tumor development (initiation, promotion and progression) but also drives, in large measure, the acquisition of drug resistance by cancer cells. Although CIN is a cornerstone of the complex mutational architecture that underlies neoplastic cell development and tumor heterogeneity and has been tightly associated with treatment responses and survival of cancer patients, it may be one of the least understood features of the malignant phenotype in terms of genetic pathways and molecular mechanisms. Here we review new insights into the type of CIN seen in multiple myeloma (MM), a blood cancer of terminally differentiated, immunoglobulin-producing B-lymphocytes called plasma cells that remains incurable in the great majority of cases. We will consider bona fide myeloma CIN genes, methods for measuring CIN in myeloma cells, and novel approaches to CIN-targeted treatments of patients with myeloma. The new findings generate optimism that enhanced understanding of CIN will lead to the design and testing of new therapeutic strategies to overcome drug resistance in MM in the not-so-distant future.
Collapse
Affiliation(s)
- Wang Wang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Ruini Chen
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhidan Tian
- Department of Pathology, Nanjing First Hospital, Nanjing, 210006, China
| | - Yongpin Zhai
- Department of Hematology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China
| | - Siegfried Janz
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, 52242, USA
| | - Chunyan Gu
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Yang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
33
|
Effenberger M, Bommert KS, Kunz V, Kruk J, Leich E, Rudelius M, Bargou R, Bommert K. Glutaminase inhibition in multiple myeloma induces apoptosis via MYC degradation. Oncotarget 2017; 8:85858-85867. [PMID: 29156762 PMCID: PMC5689652 DOI: 10.18632/oncotarget.20691] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/04/2017] [Indexed: 01/03/2023] Open
Abstract
Multiple Myeloma (MM) is an incurable hematological malignancy affecting millions of people worldwide. As in all tumor cells both glucose and more recently glutamine have been identified as important for MM cellular metabolism, however there is some dispute as to the role of glutamine in MM cell survival. Here we show that the small molecule inhibitor compound 968 effectively inhibits glutaminase and that this inhibition induces apoptosis in both human multiple myeloma cell lines (HMCLs) and primary patient material. The HMCL U266 which does not express MYC was insensitive to both glutamine removal and compound 968, but ectopic expression of MYC imparted sensitivity. Finally, we show that glutamine depletion is reflected by rapid loss of MYC protein which is independent of MYC transcription and post translational modifications. However, MYC loss is dependent on proteasomal activity, and this loss was paralleled by an equally rapid induction of apoptosis. These findings are in contrast to those of glucose depletion which largely affected rates of proliferation in HMCLs, but had no effects on either MYC expression or viability. Therefore, inhibition of glutaminolysis is effective at inducing apoptosis and thus serves as a possible therapeutic target in MM.
Collapse
Affiliation(s)
- Madlen Effenberger
- Comprehensive Cancer Center Mainfranken University Hospital Würzburg, Würzburg, Germany
| | - Kathryn S Bommert
- Comprehensive Cancer Center Mainfranken University Hospital Würzburg, Würzburg, Germany
| | - Viktoria Kunz
- Comprehensive Cancer Center Mainfranken University Hospital Würzburg, Würzburg, Germany
| | - Jessica Kruk
- Comprehensive Cancer Center Mainfranken University Hospital Würzburg, Würzburg, Germany
| | - Ellen Leich
- Institute of Pathology and Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Martina Rudelius
- Institute of Pathology and Comprehensive Cancer Center Mainfranken University of Würzburg, Würzburg, Germany
| | - Ralf Bargou
- Comprehensive Cancer Center Mainfranken University Hospital Würzburg, Würzburg, Germany
| | - Kurt Bommert
- Comprehensive Cancer Center Mainfranken University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
34
|
Zhou H, Luo W, Zeng C, Zhang Y, Wang L, Yao W, Nie C. PP2A mediates apoptosis or autophagic cell death in multiple myeloma cell lines. Oncotarget 2017; 8:80770-80789. [PMID: 29113343 PMCID: PMC5655238 DOI: 10.18632/oncotarget.20415] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/27/2017] [Indexed: 02/05/2023] Open
Abstract
The crosstalk between apoptosis and autophagy contributes to tumorigenesis and cancer therapy. The process by which BetA (betulinic acid), a naturally occurring triterpenoid, regulates apoptosis and autophagy as a cancer therapy is unclear. In this study, we show for the first time that protein phosphatase 2A (PP2A) acts as a switch to regulate apoptosis and autophagic cell death mediated by BetA. Under normal conditions, caspase-3 is activated by the mitochondrial pathway upon BetA treatment. Activated caspase-3 cleaves the A subunit of PP2A (PP2A/A), resulting in the association of PP2A and Akt. This association inactivates Akt to initiate apoptosis. Overexpression of Bcl-2 attenuates the mitochondrial apoptosis pathway, resulting in caspase-3 inactivation and the dissociation of PP2A and Akt. PP2A isolated from Akt binds with DAPK to induce autophagic cell death. Meanwhile, in vivo tumor experiments have demonstrated that BetA initiates different types of cell death in a myeloma xenograft model. Thus, PP2A can shift myeloma cells from apoptosis to autophagic cell death. These findings have important implications for the therapeutic application of BetA, particularly against apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Chemotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Luo
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chao Zeng
- Department of Gastroenterology, the Third People's Hospital of Chengdu, Chengdu, China
| | - Yu Zhang
- Department of Oncology, Guizhou People's Hospital, Guizhou, China
| | - Liyang Wang
- Department of Chemotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenxiu Yao
- Department of Chemotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunlai Nie
- Department of Chemotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
35
|
Styles CT, Bazot Q, Parker GA, White RE, Paschos K, Allday MJ. EBV epigenetically suppresses the B cell-to-plasma cell differentiation pathway while establishing long-term latency. PLoS Biol 2017; 15:e2001992. [PMID: 28771465 PMCID: PMC5542390 DOI: 10.1371/journal.pbio.2001992] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/10/2017] [Indexed: 11/29/2022] Open
Abstract
Mature human B cells infected by Epstein-Barr virus (EBV) become activated, grow, and proliferate. If the cells are infected ex vivo, they are transformed into continuously proliferating lymphoblastoid cell lines (LCLs) that carry EBV DNA as extra-chromosomal episomes, express 9 latency-associated EBV proteins, and phenotypically resemble antigen-activated B-blasts. In vivo similar B-blasts can differentiate to become memory B cells (MBC), in which EBV persistence is established. Three related latency-associated viral proteins EBNA3A, EBNA3B, and EBNA3C are transcription factors that regulate a multitude of cellular genes. EBNA3B is not necessary to establish LCLs, but EBNA3A and EBNA3C are required to sustain proliferation, in part, by repressing the expression of tumour suppressor genes. Here we show, using EBV-recombinants in which both EBNA3A and EBNA3C can be conditionally inactivated or using virus completely lacking the EBNA3 gene locus, that-after a phase of rapid proliferation-infected primary B cells express elevated levels of factors associated with plasma cell (PC) differentiation. These include the cyclin-dependent kinase inhibitor (CDKI) p18INK4c, the master transcriptional regulator of PC differentiation B lymphocyte-induced maturation protein-1 (BLIMP-1), and the cell surface antigens CD38 and CD138/Syndecan-1. Chromatin immunoprecipitation sequencing (ChIP-seq) and chromatin immunoprecipitation quantitative PCR (ChIP-qPCR) indicate that in LCLs inhibition of CDKN2C (p18INK4c) and PRDM1 (BLIMP-1) transcription results from direct binding of EBNA3A and EBNA3C to regulatory elements at these loci, producing stable reprogramming. Consistent with the binding of EBNA3A and/or EBNA3C leading to irreversible epigenetic changes, cells become committed to a B-blast fate <12 days post-infection and are unable to de-repress p18INK4c or BLIMP-1-in either newly infected cells or conditional LCLs-by inactivating EBNA3A and EBNA3C. In vitro, about 20 days after infection with EBV lacking functional EBNA3A and EBNA3C, cells develop a PC-like phenotype. Together, these data suggest that EBNA3A and EBNA3C have evolved to prevent differentiation to PCs after infection by EBV, thus favouring long-term latency in MBC and asymptomatic persistence.
Collapse
Affiliation(s)
- Christine T. Styles
- Molecular Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Quentin Bazot
- Molecular Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Gillian A. Parker
- Molecular Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Robert E. White
- Molecular Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Kostas Paschos
- Molecular Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Martin J. Allday
- Molecular Virology, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
36
|
Russignan A, Spina C, Tamassia N, Cassaro A, Rigo A, Bagnato A, Rosanò L, Bonalumi A, Gottardi M, Zanatta L, Giacomazzi A, Scupoli MT, Tinelli M, Salvadori U, Mosna F, Zamò A, Cassatella MA, Vinante F, Tecchio C. Endothelin-1 receptor blockade as new possible therapeutic approach in multiple myeloma. Br J Haematol 2017; 178:781-793. [DOI: 10.1111/bjh.14771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/14/2017] [Indexed: 01/31/2023]
Affiliation(s)
- Anna Russignan
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Cecilia Spina
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Nicola Tamassia
- Section of General Pathology; Department of Medicine; Verona University; Verona Italy
| | - Adriana Cassaro
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Antonella Rigo
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Anna Bagnato
- Preclinical Models and New Therapeutic Agents Unit; Regina Elena National Cancer Institute; Rome Italy
| | - Laura Rosanò
- Preclinical Models and New Therapeutic Agents Unit; Regina Elena National Cancer Institute; Rome Italy
| | - Angela Bonalumi
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | | | - Lucia Zanatta
- Pathology Unit; Ospedale Regionale Cà Foncello; Treviso Italy
| | - Alice Giacomazzi
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Maria Teresa Scupoli
- Interdepartmental Laboratory for Medical Research (LURM); Verona University; Verona Italy
| | - Martina Tinelli
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Ugo Salvadori
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Federico Mosna
- Haematology Unit; Ospedale Regionale Cà Foncello; Treviso Italy
| | - Alberto Zamò
- Section of Pathology; Department of Pathology and Diagnostic; Verona University; Verona Italy
| | - Marco A. Cassatella
- Section of General Pathology; Department of Medicine; Verona University; Verona Italy
| | - Fabrizio Vinante
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| | - Cristina Tecchio
- Haematology and Bone-Marrow Transplant Unit; Department of Medicine; Verona University; Verona Italy
| |
Collapse
|
37
|
Issa ME, Takhsha FS, Chirumamilla CS, Perez-Novo C, Vanden Berghe W, Cuendet M. Epigenetic strategies to reverse drug resistance in heterogeneous multiple myeloma. Clin Epigenetics 2017; 9:17. [PMID: 28203307 PMCID: PMC5303245 DOI: 10.1186/s13148-017-0319-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/26/2017] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy, which remains incurable because most patients eventually relapse or become refractory to current treatments. Due to heterogeneity within the cancer cell microenvironment, cancer cell populations employ a dynamic survival strategy to chemotherapeutic treatments, which frequently results in a rapid acquisition of therapy resistance. Besides resistance-conferring genetic alterations within a tumor cell population selected during drug treatment, recent findings also reveal non-mutational mechanisms of drug resistance, involving a small population of "cancer stem cells" (CSCs) which are intrinsically more refractory to the effects of a variety of anticancer drugs. Other studies have implicated epigenetic mechanisms in reversible drug tolerance to protect the population from eradication by potentially lethal exposures, suggesting that acquired drug resistance does not necessarily require a stable heritable genetic alteration. Clonal evolution of MM cells and the bone marrow microenvironment changes contribute to drug resistance. MM-CSCs may not be a static population and survive as phenotypically and functionally different cell types via the transition between stem-like and non-stem-like states in local microenvironments, as observed in other types of cancers. Targeting MM-CSCs is clinically relevant, and different approaches have been suggested to target molecular, metabolic and epigenetic signatures, and the self-renewal signaling characteristic of MM CSC-like cells. Here, we summarize epigenetic strategies to reverse drug resistance in heterogeneous multiple myeloma.
Collapse
Affiliation(s)
- Mark E Issa
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CH-1211 Geneva 4, Switzerland
| | - Farnaz Sedigheh Takhsha
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk, Belgium
| | - Chandra Sekhar Chirumamilla
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk, Belgium
| | - Claudina Perez-Novo
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk, Belgium
| | - Wim Vanden Berghe
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical sciences, University of Antwerp, Campus Drie Eiken, Universiteitsplein 1, Wilrijk, Belgium
| | - Muriel Cuendet
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
38
|
Agarwal P, Alzrigat M, Párraga AA, Enroth S, Singh U, Ungerstedt J, Österborg A, Brown PJ, Ma A, Jin J, Nilsson K, Öberg F, Kalushkova A, Jernberg-Wiklund H. Genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in multiple myeloma reveals the importance of Polycomb gene targeting and highlights EZH2 as a potential therapeutic target. Oncotarget 2017; 7:6809-23. [PMID: 26755663 PMCID: PMC4872750 DOI: 10.18632/oncotarget.6843] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 12/29/2015] [Indexed: 02/02/2023] Open
Abstract
Multiple myeloma (MM) is a malignancy of the antibody-producing plasma cells. MM is a highly heterogeneous disease, which has hampered the identification of a common underlying mechanism for disease establishment as well as the development of targeted therapy. Here we present the first genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in MM patient samples, defining a common set of active H3K4me3-enriched genes and silent genes marked by H3K27me3 (H3K27me3 alone or bivalent) unique to primary MM cells, when compared to normal bone marrow plasma cells. Using this epigenome profile, we found increased silencing of H3K27me3 targets in MM patients at advanced stages of the disease, and the expression pattern of H3K27me3-marked genes correlated with poor patient survival. We also demonstrated that pharmacological inhibition of EZH2 had anti-myeloma effects in both MM cell lines and CD138+ MM patient cells. In addition, EZH2 inhibition decreased the global H3K27 methylation and induced apoptosis. Taken together, these data suggest an important role for the Polycomb repressive complex 2 (PRC2) in MM, and highlights the PRC2 component EZH2 as a potential therapeutic target in MM.
Collapse
Affiliation(s)
- Prasoon Agarwal
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Mohammad Alzrigat
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Alba Atienza Párraga
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Stefan Enroth
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Umashankar Singh
- Department of Biological Engineering, Indian Institute of Technology, Gandhinagar, Gujarat, India
| | - Johanna Ungerstedt
- Department of Medicine, Center for Hematology and Regenerative Medicine (HERM), Karolinska Institute Huddinge, Stockholm, Sweden
| | - Anders Österborg
- Department of Oncology-Pathology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Peter J Brown
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, Canada
| | - Anqi Ma
- Departments of Structural and Chemical Biology, Oncological Sciences, and Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jian Jin
- Departments of Structural and Chemical Biology, Oncological Sciences, and Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth Nilsson
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Fredrik Öberg
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Antonia Kalushkova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
39
|
Abstract
3D-dynamic culture models represent an invaluable tool for a better comprehension of tumor biology and drug response, as they accurately re-create/preserve the complex multicellular organization and the dynamic interactions of the parental microenvironment, which can affect tumor fate and drug sensitivity. Hence, development of models that recapitulate tumor within its embedding microenvironment is an imperative need. This is particularly true for multiple myeloma (MM), which survives almost exclusively in the bone marrow (BM). To meet this need, we have previously exploited and validated an innovative 3D-dynamic culture technology, based on the use of the Rotary Cell Culture System (RCCS ™) bioreactor . Here, we describe, step by step, the procedures we have employed to establish two human MM ex vivo models, i.e., the culture of human BM-derived isolated cells and of MM tissues from patients.
Collapse
|
40
|
Kim M, Ju YS, Lee EJ, Kang HJ, Kim HS, Cho HC, Kim HJ, Kim JA, Lee DS, Lee YK. Abnormalities in Chromosomes 1q and 13 Independently Correlate With Factors of Poor Prognosis in Multiple Myeloma. Ann Lab Med 2016; 36:573-82. [PMID: 27578511 PMCID: PMC5011111 DOI: 10.3343/alm.2016.36.6.573] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/09/2016] [Accepted: 06/28/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND We comprehensively profiled cytogenetic abnormalities in multiple myeloma (MM) and analyzed the relationship between cytogenetic abnormalities of undetermined prognostic significance and established prognostic factors. METHODS The karyotype of 333 newly diagnosed MM cases was analyzed in association with established prognostic factors. Survival analysis was also performed. RESULTS MM with abnormal karyotypes (41.1%) exhibited high international scoring system (ISS) stage, frequent IgA type, elevated IgG or IgA levels, elevated calcium levels, elevated creatine (Cr) levels, elevated β2-microglobulin levels, and decreased Hb levels. Structural abnormalities in chromosomes 1q, 4, and 13 were independently associated with elevated levels of IgG or IgA, calcium, and Cr, respectively. Chromosome 13 abnormalities were associated with poor prognosis and decreased overall survival. CONCLUSIONS This is the first study to demonstrate that abnormalities in chromosomes 1q, 4, and 13 are associated with established factors for poor prognosis, irrespective of the presence of other concurrent chromosomal abnormalities. Chromosome 13 abnormalities have a prognostic impact on overall survival in association with elevated Cr levels. Frequent centromeric breakpoints appear to be related to MM pathogenesis.
Collapse
Affiliation(s)
- Miyoung Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Young Su Ju
- Department of Occupational and Environmental Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Eun Jin Lee
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Han Sung Kim
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Hyoun Chan Cho
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Hyo Jung Kim
- Department of Internal Medicine, Hallym University College of Medicine, Anyang, Korea
| | - Jung Ah Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Soon Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Young Kyung Lee
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, Korea.
| |
Collapse
|
41
|
Zahedi S, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P. NF-Kβ Activation in U266 Cells on Mesenchymal Stem Cells. Adv Pharm Bull 2016; 6:415-422. [PMID: 27766226 DOI: 10.15171/apb.2016.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 08/27/2016] [Accepted: 08/30/2016] [Indexed: 01/09/2023] Open
Abstract
Purpose: Mesenchymal Stem Cells (MSCs) are one of the essential members of Bone Marrow (BM) microenvironment and the cells affect normal and malignant cells in BM milieu. One of the most important hematological malignancies is Multiple Myeloma (MM). Numerous studies reported various effects of MSCs on myeloma cells. MSCs initiate various signaling pathways in myeloma cells, particularly NF-kβ. NF-kβ signaling pathway plays pivotal role in the survival, proliferation and resistance of myeloma cells to the anticancer drugs, therefore this pathway can be said to be a vital target for cancer therapy. This study examined the relationship between U266 cells and MSCs. Methods: U266 cells were cultured with Umbilical Cord Blood derived-MSCs (UCB-MSCs) and Conditioned Medium (C.M). Effect of UCB-MSCs and C.M on proliferation rate and CD54 expression of U266 cells were examined with MTT assay and Flowcytometry respectively. Furthermore, expression of CXCL1, PECAM-1, JUNB, CCL2, CD44, CCL4, IL-6, and IL-8 were analyzed by Real Time-PCR (RT-PCR). Moreover, status of p65 protein in NF-kβ pathway assessed by western blotting. Results: Our findings confirm that UCB-MSCs support U266 cells proliferation and they increase CD54 expression. In addition, we demonstrate that UCB-MSCs alter the expression of CCL4, IL-6, IL-8, CXCL1 and the levels of phosphorylated p65 in U266 cells. Conclusion: Our study provides a novel sight to the role of MSCs in the activation of NF-kβ signaling pathway. So, NF-kβ signaling pathway will be targeted in future therapies against MM.
Collapse
Affiliation(s)
- Sara Zahedi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Liu H, Dong Y, Gao Y, Du Z, Wang Y, Cheng P, Chen A, Huang H. The Fascinating Effects of Baicalein on Cancer: A Review. Int J Mol Sci 2016; 17:ijms17101681. [PMID: 27735841 PMCID: PMC5085714 DOI: 10.3390/ijms17101681] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide and a major global health problem. In recent decades, the rates of both mortality and morbidity of cancer have rapidly increased for a variety of reasons. Despite treatment options, there are serious side effects associated with chemotherapy drugs and multiple forms of drug resistance that significantly reduce their effects. There is an accumulating amount of evidence on the pharmacological activities of baicalein (e.g., anti-inflammatory, antioxidant, antiviral, and antitumor effects). Furthermore, there has been great progress in elucidating the target mechanisms and signaling pathways of baicalein's anti-cancer potential. The anti-tumor functions of baicalein are mainly due to its capacities to inhibit complexes of cyclins to regulate the cell cycle, to scavenge oxidative radicals, to attenuate mitogen activated protein kinase (MAPK), protein kinase B (Akt) or mammalian target of rapamycin (mTOR) activities, to induce apoptosis by activating caspase-9/-3 and to inhibit tumorinvasion and metastasis by reducing the expression of matrix metalloproteinase-2/-9 (MMP-2/-9). In this review, we focused on the relevant biological mechanisms of baicalein involved in inhibiting various cancers, such as bladder cancer, breast cancer, and ovarian cancer. Moreover, we also summarized the specific mechanisms by which baicalein inhibited the growth of various tumors in vivo. Taken together, baicalein may be developed as a potential, novel anticancer drug to treat tumors.
Collapse
Affiliation(s)
- Hui Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yonghui Dong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yutong Gao
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhipeng Du
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yuting Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
43
|
Li Y, Zhang B, Li W, Wang L, Yan Z, Li H, Yao Y, Yao R, Xu K, Li Z. MiR-15a/16 regulates the growth of myeloma cells, angiogenesis and antitumor immunity by inhibiting Bcl-2, VEGF-A and IL-17 expression in multiple myeloma. Leuk Res 2016; 49:73-9. [PMID: 27596960 DOI: 10.1016/j.leukres.2016.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/21/2016] [Accepted: 08/26/2016] [Indexed: 01/13/2023]
Abstract
miRNAs have been reported to be involved in the pathogenesis of many cancers. In this article, we investigated the role and the mechanisms of miR-15a/16 in the pathogenesis of multiple myeloma (MM). We found that miR-15a/16 was down-regulated in bone marrow-derived mononuclear cells (BM-MNCs) of newly diagnosed patients with MM and the downregulation of miR-15a/16 was correlated with International Staging System (ISS) stage. We then demonstrated miR-15a/16 inhibited myeloma cells proliferation, and increased apoptosis rate of U266 cells by suppressing the expression of anti-apoptosis protein Bcl-2. We also found miR-15a/16 could decrease VEGF-A and IL-17 levels in the supernatant of myeloma cells. These results indicate that miR-15a/16 may function as a tumor suppressor in MM through multiple regulatory mechanisms and they may be potential targets for the therapy of MM.
Collapse
Affiliation(s)
- Yanjie Li
- Laboratory Center of Diagnostics, Xuzhou Medical University
| | - Bingyun Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Wenjing Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Lijin Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Zhiling Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Hujun Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Yao Yao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Ruosi Yao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University.
| |
Collapse
|
44
|
Merza H, Sarkar R. Solitary extraosseous plasmacytoma. Clin Case Rep 2016; 4:851-4. [PMID: 27648261 PMCID: PMC5018587 DOI: 10.1002/ccr3.609] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 03/22/2016] [Accepted: 04/25/2016] [Indexed: 11/24/2022] Open
Abstract
Plasma cell neoplasms are characterized by a neoplastic plasma cell lineage which produces a monoclonal immunoglobulin. These neoplasms can present as a single lesion (solitary plasmacytoma) or as multiple lesions (multiple myeloma). Solitary plasmacytomas most frequently occur in bone (plasmacytomas of bone), but can also be found outside bone in soft tissues (extramedullary plasmacytomas).
Collapse
Affiliation(s)
- Hussein Merza
- Department of Internal Medicine Fox Chase Cancer Center Philadelphia Pennsylvania USA
| | - Raj Sarkar
- Department of Internal Medicine Fox Chase Cancer Center Philadelphia Pennsylvania USA
| |
Collapse
|
45
|
Hawley TS, Linsley PS, Hawley RG. Co-expression of B7–1 with Interleukin-12 Enhances Vaccine-induced Antitumour Immunity in Experimental Myeloma. Hematology 2016; 3:365-74. [DOI: 10.1080/10245332.1998.11746410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Teresa S. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| | - Peter S. Linsley
- Bristol-Meyers Squibb Pharmaceutical Research Institute, Seattle, Washington, USA
| | - Robert G. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Alexandrakis MG, Passam FH, Pappa CA, Dambaki C, Sfakiotaki G, Alegakis AK, Kyriakou DS, Stathopoulos E. Expression of Proliferating Cell Nuclear Antigen (PCNA) in Multiple Myeloma: Its Relationship to Bone Marrow Microvessel Density and other Factors of Disease Activity. Int J Immunopathol Pharmacol 2016; 17:49-56. [PMID: 15000866 DOI: 10.1177/039463200401700107] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The expression of the proliferating cell nuclear antigen (PCNA) was studied in plasma cells in bone marrow biopsies from patients with multiple myeloma (MM) using a double immunostaining method. In the same samples, microvessel density (MVD), after staining with anti-CD34 antibodies, was determined before and after chemotherapy. The correlation of PCNA expression and MVD with other myeloma parameters (clinical stage, bone marrow plasma cell infiltration and serum interleukin –6 (IL-6)) was also investigated. The study population included 51 newly diagnosed MM patients, 12 patients in plateau phase after treatment and 15 normal controls. Pretreatment mean ± SE values of PCNA, MVD, plasma cell infiltration and serum IL-6 were significantly higher than post treatment values and controls. Pretreatment PCNA expression correlated significantly with bone marrow MVD (p<0.05) plasma cell infiltration (p<0.01) and IL-6 (p<0.01). These findings show that the proliferative activity of plasma cells is related to the angiogenic activity in the bone marrow of multiple myeloma patients. Both PCNA and MVD correlate with markers of disease activity thus may provide additional information when included in the initial evaluation of myeloma bone marrow biopsies.
Collapse
Affiliation(s)
- M G Alexandrakis
- Department of Hematology, University Hospital of Heraklion, Medical School of Crete, Greece.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
The KISS1 Receptor as an In Vivo Microenvironment Imaging Biomarker of Multiple Myeloma Bone Disease. PLoS One 2016; 11:e0155087. [PMID: 27158817 PMCID: PMC4861277 DOI: 10.1371/journal.pone.0155087] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/22/2016] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma is one of the most common hematological diseases and is characterized by an aberrant proliferation of plasma cells within the bone marrow. As a result of crosstalk between cancer cells and the bone microenvironment, bone homeostasis is disrupted leading to osteolytic lesions and poor prognosis. Current diagnostic strategies for myeloma typically rely on detection of excess monoclonal immunoglobulins or light chains in the urine or serum. However, these strategies fail to localize the sites of malignancies. In this study we sought to identify novel biomarkers of myeloma bone disease which could target the malignant cells and/or the surrounding cells of the tumor microenvironment. From these studies, the KISS1 receptor (KISS1R), a G-protein-coupled receptor known to play a role in the regulation of endocrine functions, was identified as a target gene that was upregulated on mesenchymal stem cells (MSCs) and osteoprogenitor cells (OPCs) when co-cultured with myeloma cells. To determine the potential of this receptor as a biomarker, in vitro and in vivo studies were performed with the KISS1R ligand, kisspeptin, conjugated with a fluorescent dye. In vitro microscopy showed binding of fluorescently-labeled kisspeptin to both myeloma cells as well as MSCs under direct co-culture conditions. Next, conjugated kisspeptin was injected into immune-competent mice containing myeloma bone lesions. Tumor-burdened limbs showed increased peak fluorescence compared to contralateral controls. These data suggest the utility of the KISS1R as a novel biomarker for multiple myeloma, capable of targeting both tumor cells and host cells of the tumor microenvironment.
Collapse
|
48
|
IL-6 Contributes to the Defective Osteogenesis of Bone Marrow Stromal Cells from the Vertebral Body of the Glucocorticoid-Induced Osteoporotic Mouse. PLoS One 2016; 11:e0154677. [PMID: 27128729 PMCID: PMC4851291 DOI: 10.1371/journal.pone.0154677] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/18/2016] [Indexed: 01/28/2023] Open
Abstract
Osteoporosis is one of the most prevalent skeletal system diseases. It is characterized by a decrease in bone mass and microarchitectural changes in bone tissue that lead to an attenuation of bone resistance and susceptibility to fracture. Vertebral fracture is by far the most prevalent osteoporotic fracture. In the musculoskeletal system, osteoblasts, originated from bone marrow stromal cells (BMSC), are responsible for osteoid synthesis and mineralization. In osteoporosis, BMSC osteogenic differentiation is defective. However, to date, what leads to the defective BMSC osteogenesis in osteoporosis remains an open question. In the current study, we made attempts to answer this question. A mouse model of glucocorticoid-induced osteoporosis (GIO) was established and BMSC were isolated from vertebral body. The impairment of osteogenesis was observed in BMSC of osteoporotic vertebral body. The expression profiles of thirty-six factors, which play important roles in bone metabolisms, were compared through antibody array between normal and osteoporotic BMSC. Significantly higher secretion level of IL-6 was observed in osteoporotic BMSCs compared with normal control. We provided evidences that IL-6 over-secretion impaired osteogenesis of osteoporotic BMSC. Further, it was observed that β-catenin activity was inhibited in response to IL-6 over-secretion. More importantly, in vivo administration of IL-6 neutralizing antibody was found to be helpful to rescue the osteoporotic phenotype of mouse vertebral body. Our study provides a deeper insight into the pathophysiology of osteoporosis and identifies IL-6 as a promising target for osteoporosis therapy.
Collapse
|
49
|
Khan OS, Bhat AA, Krishnankutty R, Mohammad RM, Uddin S. Therapeutic Potential of Resveratrol in Lymphoid Malignancies. Nutr Cancer 2016; 68:365-373. [PMID: 27028800 DOI: 10.1080/01635581.2016.1152386] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Natural products have always been sought as a dependable source for the cure of many fatal diseases including cancer. Resveratrol (RSV), a naturally occurring plant polyphenol, has been of recent research interest and is being investigated for its beneficial biological properties that include antioxidant, anti-inflammatory, proapoptotic, and growth inhibitory activities. These effects are mainly mediated by cell cycle arrest, upregulation of proapoptotic proteins, loss of mitochondrial potential, and generation of reactive oxygen species. Among the beneficial properties of RSV, the anticancer property has been of the prime focus and extensively explored during the last few years. Although reports exist on the chemopreventive role of RSV in many solid tumors, limited information is available on the antiproliferative activity of RSV in human lymphoma cells and experimental models. Potential mechanisms for its antiproliferative effect include induction of cell differentiation, apoptosis, and inhibition of DNA synthesis. In this review, the different kinds of lymphoid malignancies and the main mechanisms of cell death induced by resveratrol are discussed. The challenges are limiting in vivo experimental studies involving resveratrol. An attempt for the translation of this compound into a clinical drug also forms a part of this review.
Collapse
Affiliation(s)
- Omar S Khan
- a Health Science, College of Science, Benedictine University , Lisle , Illinois , USA
| | - Ajaz A Bhat
- b Translational Research Institute, Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Roopesh Krishnankutty
- b Translational Research Institute, Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Ramzi M Mohammad
- b Translational Research Institute, Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Shahab Uddin
- b Translational Research Institute, Academic Health System, Hamad Medical Corporation , Doha , Qatar
| |
Collapse
|
50
|
Low M, Infantino S, Grigoriadis G, Tarlinton D. Targeting plasma cells: are we any closer to a panacea for diseases of antibody-secreting cells? Immunol Rev 2016; 270:78-94. [DOI: 10.1111/imr.12388] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Michael Low
- Immunology Division; Walter and Eliza Hall Institute of Medical Research; University of Melbourne; Parkville Vic. Australia
- Department of Haematology; Monash Health; Monash Hospital; Clayton Vic. Australia
- Department of Medical Biology; The University of Melbourne; Parkville Vic. Australia
| | - Simona Infantino
- Immunology Division; Walter and Eliza Hall Institute of Medical Research; University of Melbourne; Parkville Vic. Australia
- Department of Medical Biology; The University of Melbourne; Parkville Vic. Australia
| | - George Grigoriadis
- Department of Haematology; Monash Health; Monash Hospital; Clayton Vic. Australia
- School of Clinical Sciences at Monash Health; Monash University; Clayton Vic. Australia
- Centre for Cancer Research; Hudson Institute of Medical Research; Clayton Vic. Australia
- Malignant Haematology and Stem Cell Transplantation Service and Alfred Pathology Service; The Alfred; Melbourne Vic. Australia
| | - David Tarlinton
- Immunology Division; Walter and Eliza Hall Institute of Medical Research; University of Melbourne; Parkville Vic. Australia
- Department of Haematology; Monash Health; Monash Hospital; Clayton Vic. Australia
| |
Collapse
|