51
|
Lê GN, Bones J, Coyne M, Bazou D, Dowling P, O'Gorman P, Larkin AM. Current and future biomarkers for risk-stratification and treatment personalisation in multiple myeloma. Mol Omics 2019; 15:7-20. [PMID: 30652172 DOI: 10.1039/c8mo00193f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple myeloma, an incurable malignancy of the plasma cells in the bone marrow, has a complex pathogenesis due to clonal heterogeneity. Over the years, many clinical trials and researches have led to the development of effective myeloma treatments, resulting in survival prolongation. Molecular prognostic markers for risk-stratification to predict survival, and predictive markers for treatment response are being extensively explored. This review discusses the current risk-adaptive strategies based on genetic and molecular risk signatures that are in practice to predict survival and describes the future prognostic and predictive biomarkers across the fields of genomics, proteomics, and glycomics in myeloma. Gene expression profiling and next generation sequencing are coming to the forefront of risk-stratification and therapeutic-response prediction. Similarly, proteomic and glycomic-based platforms are gaining momentum in biomarker discovery to predict drug resistance and disease progression.
Collapse
Affiliation(s)
- Giao N Lê
- NIBRT - The National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merion, Blackrock Co., Dublin A94 X099, Ireland.
| | | | | | | | | | | | | |
Collapse
|
52
|
Chandler KB, Costello CE, Rahimi N. Glycosylation in the Tumor Microenvironment: Implications for Tumor Angiogenesis and Metastasis. Cells 2019; 8:E544. [PMID: 31195728 PMCID: PMC6627046 DOI: 10.3390/cells8060544] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/31/2019] [Accepted: 06/01/2019] [Indexed: 01/27/2023] Open
Abstract
Just as oncogene activation and tumor suppressor loss are hallmarks of tumor development, emerging evidence indicates that tumor microenvironment-mediated changes in glycosylation play a crucial functional role in tumor progression and metastasis. Hypoxia and inflammatory events regulate protein glycosylation in tumor cells and associated stromal cells in the tumor microenvironment, which facilitates tumor progression and also modulates a patient's response to anti-cancer therapeutics. In this review, we highlight the impact of altered glycosylation on angiogenic signaling and endothelial cell adhesion, and the critical consequences of these changes in tumor behavior.
Collapse
Affiliation(s)
- Kevin Brown Chandler
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Catherine E Costello
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Nader Rahimi
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
53
|
Daly J, Carlsten M, O'Dwyer M. Sugar Free: Novel Immunotherapeutic Approaches Targeting Siglecs and Sialic Acids to Enhance Natural Killer Cell Cytotoxicity Against Cancer. Front Immunol 2019; 10:1047. [PMID: 31143186 PMCID: PMC6521797 DOI: 10.3389/fimmu.2019.01047] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/24/2019] [Indexed: 12/20/2022] Open
Abstract
Natural Killer (NK) cells are cytotoxic lymphocytes that play a key role in the immune system, targeting and destroying invading pathogens and malignantly transformed cells. Evading NK cell-mediated immunosurveillance is therefore critical to facilitating cancer cell survival and metastasis. Signals from a range of inhibitory and activating receptors located on the NK cell surface regulate NK cell cytotoxicity. Recently, attention has turned to the role of hypersialylated tumor cell surfaces in mediating immune-evasion of NK cells. Two inhibitory sialic acid-binding immunoglobulin-like lectin (Siglec) receptors are expressed by NK cells: Siglec-7 and Siglec-9. The abundance of sialic acids on tumor cell surface is hypothesized to regulate NK cell-mediated cytotoxicity by interacting with Siglec-7 and Siglec-9, causing a dampening of NK cell activation pathways. Targeting Siglec-7 and Siglec-9, or the sialic acid coated tumor cell surface is therefore being investigated as a novel therapeutic approach to enhance the NK cell response against cancer. In this review we report on the currently published documentation of the role for Siglec-7 and Siglec-9 receptors on NK cells and their ligands expressed by tumor cells. We also discuss the strategies currently explored to target Siglec-7, Siglec-9 and the sialylated tumor cell surface as well as the impact abrogation of these interactions have on NK cell cytotoxicity against several cancer types.
Collapse
Affiliation(s)
- John Daly
- Department of Hematology, Biomedical Sciences, National University of Ireland Galway, Galway, Ireland
| | - Mattias Carlsten
- Department of Medicine, Huddinge, Center for Haematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michael O'Dwyer
- Department of Hematology, Biomedical Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
54
|
Zhang Z, Westhrin M, Bondt A, Wuhrer M, Standal T, Holst S. Serum protein N-glycosylation changes in multiple myeloma. Biochim Biophys Acta Gen Subj 2019; 1863:960-970. [DOI: 10.1016/j.bbagen.2019.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 10/27/2022]
|
55
|
Wang M, Zhu J, Lubman DM, Gao C. Aberrant glycosylation and cancer biomarker discovery: a promising and thorny journey. Clin Chem Lab Med 2019; 57:407-416. [PMID: 30138110 PMCID: PMC6785348 DOI: 10.1515/cclm-2018-0379] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 07/15/2018] [Indexed: 12/12/2022]
Abstract
Glycosylation is among the most important post-translational modifications for proteins and is of intrinsic complex character compared with DNAs and naked proteins. Indeed, over 50%-70% of proteins in circulation are glycosylated, and the "sweet attachments" have versatile structural and functional implications. Both the configuration and composition of the attached glycans affect the biological activities of consensus proteins significantly. Glycosylation is generated by complex biosynthetic pathways comprising hundreds of glycosyltransferases, glycosidases, transcriptional factors, transporters and the protein backbone. In addition, lack of direct genetic templates and glyco-specific antibodies such as those commonly used in DNA amplification and protein capture makes research on glycans and glycoproteins even more difficult, thus resulting in sparse knowledge on the pathophysiological implications of glycosylation. Fortunately, cutting-edge technologies have afforded new opportunities and approaches for investigating cancer-related glycosylation. Thus, glycans as well as aberrantly glycosylated protein-based cancer biomarkers have been increasingly recognized. This mini-review highlights the most recent developments in glyco-biomarker studies in an effort to discover clinically relevant cancer biomarkers using advanced analytical methodologies such as mass spectrometry, high-performance liquid chromatographic/ultra-performance liquid chromatography, capillary electrophoresis, and lectin-based technologies. Recent clinical-centered glycobiological studies focused on determining the regulatory mechanisms and the relation with diagnostics, prognostics and even therapeutics are also summarized. These studies indicate that glycomics is a treasure waiting to be mined where the growth of cancer-related glycomics and glycoproteomics is the next great challenge after genomics and proteomics.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, P.R. China
| | - Jianhui Zhu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David M. Lubman
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Chunfang Gao
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, P.R. China
| |
Collapse
|
56
|
Windisch R, Pirschtat N, Kellner C, Chen-Wichmann L, Lausen J, Humpe A, Krause DS, Wichmann C. Oncogenic Deregulation of Cell Adhesion Molecules in Leukemia. Cancers (Basel) 2019; 11:E311. [PMID: 30841639 PMCID: PMC6468598 DOI: 10.3390/cancers11030311] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 01/01/2023] Open
Abstract
Numerous cell⁻cell and cell⁻matrix interactions within the bone marrow microenvironment enable the controlled lifelong self-renewal and progeny of hematopoietic stem and progenitor cells (HSPCs). On the cellular level, this highly mutual interaction is granted by cell adhesion molecules (CAMs) integrating differentiation, proliferation, and pro-survival signals from the surrounding microenvironment to the inner cell. However, cell⁻cell and cell⁻matrix interactions are also critically involved during malignant transformation of hematopoietic stem/progenitor cells. It has become increasingly apparent that leukemia-associated gene products, such as activated tyrosine kinases and fusion proteins resulting from chromosomal translocations, directly regulate the activation status of adhesion molecules, thereby directing the leukemic phenotype. These observations imply that interference with adhesion molecule function represents a promising treatment strategy to target pre-leukemic and leukemic lesions within the bone marrow niche. Focusing on myeloid leukemia, we provide a current overview of the mechanisms by which leukemogenic gene products hijack control of cellular adhesion to subsequently disturb normal hematopoiesis and promote leukemia development.
Collapse
Affiliation(s)
- Roland Windisch
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Nina Pirschtat
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Linping Chen-Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Johann-Wolfgang-Goethe University and German Red Cross Blood Service, 60528 Frankfurt am Main, Germany.
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| | - Daniela S Krause
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt am Main, Germany.
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
57
|
Redondo-Muñoz J, García-Pardo A, Teixidó J. Molecular Players in Hematologic Tumor Cell Trafficking. Front Immunol 2019; 10:156. [PMID: 30787933 PMCID: PMC6372527 DOI: 10.3389/fimmu.2019.00156] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
The trafficking of neoplastic cells represents a key process that contributes to progression of hematologic malignancies. Diapedesis of neoplastic cells across endothelium and perivascular cells is facilitated by adhesion molecules and chemokines, which act in concert to tightly regulate directional motility. Intravital microscopy provides spatio-temporal views of neoplastic cell trafficking, and is crucial for testing and developing therapies against hematologic cancers. Multiple myeloma (MM), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL) are hematologic malignancies characterized by continuous neoplastic cell trafficking during disease progression. A common feature of these neoplasias is the homing and infiltration of blood cancer cells into the bone marrow (BM), which favors growth and survival of the malignant cells. MM cells traffic between different BM niches and egress from BM at late disease stages. Besides the BM, CLL cells commonly home to lymph nodes (LNs) and spleen. Likewise, ALL cells also infiltrate extramedullary organs, such as the central nervous system, spleen, liver, and testicles. The α4β1 integrin and the chemokine receptor CXCR4 are key molecules for MM, ALL, and CLL cell trafficking into and out of the BM. In addition, the chemokine receptor CCR7 controls CLL cell homing to LNs, and CXCR4, CCR7, and CXCR3 contribute to ALL cell migration across endothelia and the blood brain barrier. Some of these receptors are used as diagnostic markers for relapse and survival in ALL patients, and their level of expression allows clinicians to choose the appropriate treatments. In CLL, elevated α4β1 expression is an established adverse prognostic marker, reinforcing its role in the disease expansion. Combining current chemotherapies with inhibitors of malignant cell trafficking could represent a useful therapy against these neoplasias. Moreover, immunotherapy using humanized antibodies, CAR-T cells, or immune check-point inhibitors together with agents targeting the migration of tumor cells could also restrict their survival. In this review, we provide a view of the molecular players that regulate the trafficking of neoplastic cells during development and progression of MM, CLL, and ALL, together with current therapies that target the malignant cells.
Collapse
Affiliation(s)
- Javier Redondo-Muñoz
- Department of Immunology, Ophthalmology and ERL, Hospital 12 de Octubre Health Research Institute (imas12), School of Medicine, Complutense University, Madrid, Spain.,Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Angeles García-Pardo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
58
|
Pang X, Li H, Guan F, Li X. Multiple Roles of Glycans in Hematological Malignancies. Front Oncol 2018; 8:364. [PMID: 30237983 PMCID: PMC6135871 DOI: 10.3389/fonc.2018.00364] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023] Open
Abstract
The three types of blood cells (red blood cells for carrying oxygen, white blood cells for immune protection, and platelets for wound clotting) arise from hematopoietic stem/progenitor cells in the adult bone marrow, and function in physiological regulation and communication with local microenvironments to maintain systemic homeostasis. Hematological malignancies are relatively uncommon malignant disorders derived from the two major blood cell lineages: myeloid (leukemia) and lymphoid (lymphoma). Malignant clones lose their regulatory mechanisms, resulting in production of a large number of dysfunctional cells and destruction of normal hematopoiesis. Glycans are one of the four major types of essential biological macromolecules, along with nucleic acids, proteins, and lipids. Major glycan subgroups are N-glycans, O-glycans, glycosaminoglycans, and glycosphingolipids. Aberrant expression of glycan structures, resulting from dysregulation of glycan-related genes, is associated with cancer development and progression in terms of cell signaling and communication, tumor cell dissociation and invasion, cell-matrix interactions, tumor angiogenesis, immune modulation, and metastasis formation. Aberrant glycan expression occurs in most hematological malignancies, notably acute myeloid leukemia, myeloproliferative neoplasms, and multiple myeloma, etc. Here, we review recent research advances regarding aberrant glycans, their related genes, and their roles in hematological malignancies. Our improved understanding of the mechanisms that underlie aberrant patterns of glycosylation will lead to development of novel, more effective therapeutic approaches targeted to hematological malignancies.
Collapse
Affiliation(s)
- Xingchen Pang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Hongjiao Li
- College of Life Science, Northwest University, Xi'an, China
| | - Feng Guan
- School of Biotechnology, Jiangnan University, Wuxi, China.,College of Life Science, Northwest University, Xi'an, China
| | - Xiang Li
- College of Life Science, Northwest University, Xi'an, China.,Wuxi Medical School, Jiangnan University, Wuxi, China
| |
Collapse
|
59
|
Rodrigues E, Macauley MS. Hypersialylation in Cancer: Modulation of Inflammation and Therapeutic Opportunities. Cancers (Basel) 2018; 10:cancers10060207. [PMID: 29912148 PMCID: PMC6025361 DOI: 10.3390/cancers10060207] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Cell surface glycosylation is dynamic and often changes in response to cellular differentiation under physiological or pathophysiological conditions. Altered glycosylation on cancers cells is gaining attention due its wide-spread occurrence across a variety of cancer types and recent studies that have documented functional roles for aberrant glycosylation in driving cancer progression at various stages. One change in glycosylation that can correlate with cancer stage and disease prognosis is hypersialylation. Increased levels of sialic acid are pervasive in cancer and a growing body of evidence demonstrates how hypersialylation is advantageous to cancer cells, particularly from the perspective of modulating immune cell responses. Sialic acid-binding receptors, such as Siglecs and Selectins, are well-positioned to be exploited by cancer hypersialylation. Evidence is also mounting that Siglecs modulate key immune cell types in the tumor microenvironment, particularly those responsible for maintaining the appropriate inflammatory environment. From these studies have come new and innovative ways to block the effects of hypersialylation by directly reducing sialic acid on cancer cells or blocking interactions between sialic acid and Siglecs or Selectins. Here we review recent works examining how cancer cells become hypersialylated, how hypersialylation benefits cancer cells and tumors, and proposed therapies to abrogate hypersialylation of cancer.
Collapse
Affiliation(s)
- Emily Rodrigues
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
60
|
Association of cytosolic sialidase Neu2 with plasma membrane enhances Fas-mediated apoptosis by impairing PI3K-Akt/mTOR-mediated pathway in pancreatic cancer cells. Cell Death Dis 2018; 9:210. [PMID: 29434218 PMCID: PMC5833727 DOI: 10.1038/s41419-017-0191-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 12/27/2022]
Abstract
Modulation of sialylation by sialyltransferases and sialidases plays essential role in carcinogenesis. There are few reports on sialyltransferase, however, the contribution of cytosolic sialidase (Neu2) remains unexplored in pancreatic ductal adenocarcinoma (PDAC). We observed lower expression of Neu2 in different PDAC cells, patient tissues, and a significant strong association with clinicopathological characteristics. Neu2 overexpression guided drug-resistant MIAPaCa2 and AsPC1 cells toward apoptosis as evidenced by decreased Bcl2/Bax ratio, activation of caspase-3/caspase-6/caspase-8, PARP reduction, reduced CDK2/CDK4/CDK6, and cyclin-B1/cyclin-E with unaffected caspase-9. Neu2-overexpressed cells exhibited higher expression of Fas/CD95-death receptor, FasL, FADD, and Bid cleavage confirming extrinsic pathway-mediated apoptosis. α2,6-linked sialylation of Fas helps cancer cells to survive, which is a substrate for Neu2. Therefore, their removal should enhance Fas-mediated apoptosis. Neu2-overexpressed cells indeed showed increased enzyme activity even on membrane. Interestingly, this membrane-bound Neu2 exhibited enhanced association with Fas causing its desialylation and activation as corroborated by decreased association of Fas with α2,6-sialic acid-binding lectin. Additionally, enhanced cytosolic Neu2 inhibited the expression of several growth factor-mediated signaling molecules involved in PI3K/Akt–mTOR pathway probably through desialylation which in turn also causes Fas activation. Furthermore, Neu2-overexpressed cells exhibited reduced cell migration, invasion with decreased VEGF, VEGFR, and MMP9 levels. To the best of our knowledge, this is the first report of cytosolic Neu2 on membrane, its association with Fas, enhanced desialylation, activation, and Fas-mediated apoptosis. Taken together, our study ascertains a novel concept by which the function of Fas/CD95 could be modulated indicating a critical role of upstream Neu2 as a promising target for inducing apoptosis in pancreatic cancer.
Collapse
|
61
|
Sitarski AM, Fairfield H, Falank C, Reagan MR. 3d Tissue Engineered In Vitro Models Of Cancer In Bone. ACS Biomater Sci Eng 2018; 4:324-336. [PMID: 29756030 PMCID: PMC5945209 DOI: 10.1021/acsbiomaterials.7b00097] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biological models are necessary tools for gaining insight into underlying mechanisms governing complex pathologies such as cancer in the bone. Models range from in vitro tissue culture systems to in vivo models and can be used with corresponding epidemiological and clinical data to understand disease etiology, progression, driver mutations, and signaling pathways. In bone cancer, as with many other cancers, in vivo models are often too complex to study specific cell-cell interactions or protein roles, and 2D models are often too simple to accurately represent disease processes. Consequently, researchers have increasingly turned to 3D in vitro tissue engineered models as a useful compromise. In this review, tissue engineered 3D models of bone and cancer are described in depth and compared to 2D models. Biomaterials and cell types used are described, and future directions in the field of tissue engineered bone cancer models are proposed.
Collapse
Affiliation(s)
- Anna M. Sitarski
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
- University of Maine, Orono, Maine 04469, USA
| | - Heather Fairfield
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
- University of Maine, Orono, Maine 04469, USA
- School of Medicine, Tufts University, Boston, Massachusetts 02111, USA
| | - Carolyne Falank
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
- University of Maine, Orono, Maine 04469, USA
- School of Medicine, Tufts University, Boston, Massachusetts 02111, USA
| | - Michaela R. Reagan
- Maine Medical Center Research Institute, Scarborough, Maine 04074, USA
- University of Maine, Orono, Maine 04469, USA
- School of Medicine, Tufts University, Boston, Massachusetts 02111, USA
| |
Collapse
|
62
|
Shen Y, Feng Y, Chen H, Huang L, Wang F, Bai J, Yang Y, Wang J, Zhao W, Jia Y, Peng Y, Lei X, He A. Focusing on long non-coding RNA dysregulation in newly diagnosed multiple myeloma. Life Sci 2018; 196:133-142. [PMID: 29459023 DOI: 10.1016/j.lfs.2018.01.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
AIMS Multiple myeloma (MM) is an incurable hematological cancer with a higher rate of relapse. Alterations in the function of long non-coding RNAs (lncRNAs) promote the progression and metastasis of cancer. We carry out this study to explore the expression profile of differently expressed lncRNAs in newly diagnosed MM. MAIN METHODS The Bone marrows we analyzed were obtained from five MM and five IDA patients (serving as controls). Arraystar Human LncRNA Array V4.0 was used to profile expression of lncRNAs and mRNAs. Gene ontology (GO) and pathway analysis were utilized to understand the biological roles of differently expressed genes, while Database for Annotation, Visualization and Integrated Discovery (DAVID) was used for constructing the lncRNA-mRNA co-expression network. Quantitative polymerase chain reaction (qRT-PCR) was performed to confirm the expressions of dysregulated lncRNAs. KEY FINDINGS Bioinformatic analysis of the lncRNA expression identified >3000 dysregulated lncRNAs (difference ≥ 2-fold) in MM samples. GO and pathway analysis revealed that ECM-receptor and cell cycle pathway-related genes were significantly associated with MM. Four dysregulated lncRNAs were confirmed by qRT-PCR. Among them, the expression of ST3GAL6-AS1, LAMA5-AS1and RP11-175D17.3wereassociated with stage and risk status of MM. On the basis of GEO public database analysis, LAMA5-AS1 was related with an overall survival rate of MM patients. SIGNIFICANCE These results reveal the feasible functions of lncRNAs in pathogenesis of MM. Further studies are required to explore whether these lncRNAs could serve as candidate therapeutic targets and new molecular biomarkers for MM.
Collapse
Affiliation(s)
- Ying Shen
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuandong Feng
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hongli Chen
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lingjuan Huang
- Department of Geriatrics, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Fangxia Wang
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ju Bai
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yun Yang
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianli Wang
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wanhong Zhao
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yachun Jia
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yan Peng
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaoru Lei
- Institute of Hematology of Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Aili He
- Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
63
|
Chen J, Fang M, Chen X, Yi C, Ji J, Cheng C, Wang M, Gu X, Sun Q, Gao C. N-glycosylation of serum proteins for the assessment of patients with IgD multiple myeloma. BMC Cancer 2017; 17:881. [PMID: 29268706 PMCID: PMC5740902 DOI: 10.1186/s12885-017-3891-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 12/08/2017] [Indexed: 01/16/2023] Open
Abstract
Background Because glycosylation is one of the most common post-translational modifications of proteins and because changes in glycosylation have been shown to have a significant correlation with the development of many cancer types, we investigated the serum N-glycome used to diagnose, stage and evaluate the pathological outcomes in IgD multiple myeloma. Methods Serum samples were available for 20 patients with IgD multiple myeloma, 41 patients with light chain multiple myeloma and 42 healthy control subjects. Serum N-glycans were released and analysed using DNA sequencer-assisted fluorophore-assisted capillary electrophoresis. Results Characteristic changes were revealed in the serum N-glycome of IgD myeloma. In particular, three N-glycans (NG1(6)A2F, Peak3; NG1(3)A2F, Peak4; NA2FB, Peak7) showed increased clinical value. The best area under the ROC curve of NG1(6)A2F to diagnose IgD myeloma was 0.981, with a 95.0% sensitivity and 95.2% specificity, and that of NG1(3)A2F was 0.936, with a 95.0% sensitivity and 78.6% specificity. The best area under the ROC curve of NA2FB/NG1(3)A2F to differentially diagnose IgD myeloma versus light chain myeloma was 0.744, with a 95.3% sensitivity and 50.0% specificity. The level of NG1(3)A2F was correlated with the international staging system, while the higher abundance of NA2FB presented in IgD myeloma was predictive of a shorter progression-free survival. Conclusions The advent of serum N-glycan signatures may play a role in the diagnosis, staging and prognosis of IgD myeloma and will serve as the foundation for a precision medicine approach to this rare subtype of multiple myeloma. Electronic supplementary material The online version of this article (10.1186/s12885-017-3891-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Chen
- Department of Laboratory Medicine, Shanghai Jingan District Zhabei Central Hospital, 619 Zhonghuaxin Road, Shanghai, China
| | - Meng Fang
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Xiaoling Chen
- Department of Hematology, Shanghai Jingan District Zhabei Central Hospital, 619 Zhonghuaxin Road, Shanghai, China
| | - Changhong Yi
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Jun Ji
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Cheng Cheng
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Mengmeng Wang
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Xing Gu
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Quansheng Sun
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China
| | - Chunfang Gao
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, China.
| |
Collapse
|
64
|
Rengasamy M, Zhang F, Vashisht A, Song WM, Aguilo F, Sun Y, Li S, Zhang W, Zhang B, Wohlschlegel JA, Walsh MJ. The PRMT5/WDR77 complex regulates alternative splicing through ZNF326 in breast cancer. Nucleic Acids Res 2017; 45:11106-11120. [PMID: 28977470 PMCID: PMC5737218 DOI: 10.1093/nar/gkx727] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/11/2017] [Indexed: 12/22/2022] Open
Abstract
We observed overexpression and increased intra-nuclear accumulation of the PRMT5/WDR77 in breast cancer cell lines relative to immortalized breast epithelial cells. Utilizing mass spectrometry and biochemistry approaches we identified the Zn-finger protein ZNF326, as a novel interaction partner and substrate of the nuclear PRMT5/WDR77 complex. ZNF326 is symmetrically dimethylated at arginine 175 (R175) and this modification is lost in a PRMT5 and WDR77-dependent manner. Loss of PRMT5 or WDR77 in MDA-MB-231 cells leads to defects in alternative splicing, including inclusion of A-T rich exons in target genes, a phenomenon that has previously been observed upon loss of ZNF326. We observed that the alternatively spliced transcripts of a subset of these genes, involved in proliferation and tumor cell migration like REPIN1/AP4, ST3GAL6, TRNAU1AP and PFKM are degraded upon loss of PRMT5. In summary, we have identified a novel mechanism through which the PRMT5/WDR77 complex maintains the balance between splicing and mRNA stability through methylation of ZNF326.
Collapse
Affiliation(s)
- Madhumitha Rengasamy
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fan Zhang
- Department of Medicine, Division of Nephrology, Bioinformatics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Center for Life Sciences, School of Life Sciences and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Ajay Vashisht
- Departmentof Biological Chemistry and the Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Francesca Aguilo
- Wallenberg Centre for Molecular Medicine, Department of Medical Biosciences, University of Umeå, Försörjningsvägen 19073, Umeå, Sweden
| | - Yifei Sun
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mount Sinai Center for RNA Biology and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - SiDe Li
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mount Sinai Center for RNA Biology and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Weijia Zhang
- Department of Medicine, Division of Nephrology, Bioinformatics Laboratory, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James A Wohlschlegel
- Departmentof Biological Chemistry and the Institute of Genomics and Proteomics, University of California, Los Angeles, CA 90095, USA
| | - Martin J Walsh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,The Mount Sinai Center for RNA Biology and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
65
|
Falank C, Fairfield H, Reagan MR. Reflections on Cancer in the Bone Marrow: Adverse Roles of Adipocytes. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:254-262. [PMID: 29399440 PMCID: PMC5791905 DOI: 10.1007/s40610-017-0074-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This review highlights the recent advances in our understanding of adipocyte contributions to carcinogenesis or cancer disease progression for cancers in the bone. PURPOSE In this review, we aim to describe bone marrow adipose tissue and discuss the soluble adipocyte-derived cytokines (adipokines) or endocrine factors, adipocyte-derived lipids, and the actual or putative juxtacrine signaling between bone marrow adipocytes and tumor cells in the bone marrow. This relationship likely affects tumor cell initiation, proliferation, metastasis, and/or drug resistance. RECENT FINDINGS Bone marrow adipose may affect tumor proliferation, drug resistance, or cancer-induced bone disease and hence may be a new target in the fight against cancer. SUMMARY Overall, evidence is mixed regarding the role of bone marrow adipose and adipocytes in cancer progression, and more research in this arena is necessary to determine how these bone marrow microenvironmental cells contribute to malignancies in the marrow to identify novel, potentially targetable pathways.
Collapse
Affiliation(s)
- Carolyne Falank
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Boston, MA, USA
| | - Heather Fairfield
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Boston, MA, USA
| | - Michaela R. Reagan
- Maine Medical Research Institute, Scarborough, ME, USA
- University of Maine, Orono, ME, USA
- Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
66
|
Hewett DR, Vandyke K, Lawrence DM, Friend N, Noll JE, Geoghegan JM, Croucher PI, Zannettino ACW. DNA Barcoding Reveals Habitual Clonal Dominance of Myeloma Plasma Cells in the Bone Marrow Microenvironment. Neoplasia 2017; 19:972-981. [PMID: 29091798 PMCID: PMC5678743 DOI: 10.1016/j.neo.2017.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/30/2017] [Indexed: 02/07/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy resulting from the uncontrolled proliferation of antibody-producing plasma cells in the bone marrow. At diagnosis, independent plasma cell tumors are found throughout the skeleton. The recirculation of mutant plasma cells from the initial lesion and their recolonization of distant marrow sites are thought to occur by a process similar to solid tumor metastasis. However, the efficiency of this bone marrow homing process and the proportion of disseminated cells that actively divide and contribute to new tumor growth in MM are both unknown. We used the C57BL/KaLwRij mouse model of myeloma, lentiviral-mediated DNA barcoding of 5TGM1 myeloma cells, and next-generation sequencing to investigate the relative efficiency of plasma cell migration to, and growth within, the bone marrow. This approach revealed three major findings: firstly, establishment of metastasis within the bone marrow was extremely inefficient, with approximately 0.01% of circulating myeloma cells becoming resident long term in the bone marrow of each long bone; secondly, the individual cells of each metastasis exhibited marked differences in their proliferative fates, with the majority of final tumor burden within a bone being attributable to the progeny of between 1 and 8 cells; and, thirdly, the proliferative fate of individual clonal plasma cells differed at each bone marrow site in which the cells “landed.” These findings suggest that individual myeloma plasma cells are subjected to vastly different selection pressures within the bone marrow microenvironment, highlighting the importance of niche-driven factors, which determine the disease course and outcome.
Collapse
Affiliation(s)
- Duncan R Hewett
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia.
| | - Kate Vandyke
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia
| | - David M Lawrence
- Centre for Cancer Biology, Australian Cancer Research Fund Cancer Genomics Facility, SA Pathology, Adelaide, Australia; School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Natasha Friend
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia
| | - Jacqueline E Noll
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia
| | - Joel M Geoghegan
- Centre for Cancer Biology, Australian Cancer Research Fund Cancer Genomics Facility, SA Pathology, Adelaide, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales, 2010
| | - Andrew C W Zannettino
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, 5000, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, 5000, Australia
| |
Collapse
|
67
|
GFAPδ/GFAPα ratio directs astrocytoma gene expression towards a more malignant profile. Oncotarget 2017; 8:88104-88121. [PMID: 29152145 PMCID: PMC5675697 DOI: 10.18632/oncotarget.21540] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022] Open
Abstract
Astrocytomas are the most common malignant brain tumours and are to date incurable. It is unclear how astrocytomas progress into higher malignant grades. The intermediate filament cytoskeleton is emerging as an important regulator of malignancy in several tumours. The majority of the astrocytomas express the intermediate filament protein Glial Fibrillary Acidic Protein (GFAP). Several GFAP splice variants have been identified and the main variants expressed in human astrocytoma are the GFAPα and GFAPδ isoforms. Here we show a significant downregulation of GFAPα in grade IV astrocytoma compared to grade II and III, resulting in an increased GFAPδ/α ratio. Mimicking this increase in GFAPδ/α ratio in astrocytoma cell lines and comparing the subsequent transcriptomic changes with the changes in the patient tumours, we have identified a set of GFAPδ/α ratio-regulated high-malignant and low-malignant genes. These genes are involved in cell proliferation and protein phosphorylation, and their expression correlated with patient survival. We additionally show that changing the ratio of GFAPδ/α, by targeting GFAP expression, affected expression of high-malignant genes. Our data imply that regulating GFAP expression and splicing are novel therapeutic targets that need to be considered as a treatment for astrocytoma.
Collapse
|
68
|
Moschetta M, Kawano Y, Sacco A, Belotti A, Ribolla R, Chiarini M, Giustini V, Bertoli D, Sottini A, Valotti M, Ghidini C, Serana F, Malagola M, Imberti L, Russo D, Montanelli A, Rossi G, Reagan MR, Maiso P, Paiva B, Ghobrial IM, Roccaro AM. Bone Marrow Stroma and Vascular Contributions to Myeloma Bone Homing. Curr Osteoporos Rep 2017; 15:499-506. [PMID: 28889371 DOI: 10.1007/s11914-017-0399-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF THE REVIEW Herein we dissect mechanisms behind the dissemination of cancer cells from primary tumor site to the bone marrow, which are necessary for metastasis development, with a specific focus on multiple myeloma. RECENT FINDINGS The ability of tumor cells to invade vessels and reach the systemic circulation is a fundamental process for metastasis development; however, the interaction between clonal cells and the surrounding microenvironment is equally important for supporting colonization, survival, and growth in the secondary sites of dissemination. The intrinsic propensity of tumor cells to recognize a favorable milieu where to establish secondary growth is the basis of the "seed and soil" theory. This theory assumes that certain tumor cells (the "seeds") have a specific affinity for the milieu of certain organs (the "soil"). Recent literature has highlighted the important contributions of the vascular niche to the hospitable "soil" within the bone marrow. In this review, we discuss the crucial role of stromal cells and endothelial cells in supporting primary growth, homing, and metastasis to the bone marrow, in the context of multiple myeloma, a plasma cell malignancy with the unique propensity to primarily grow and metastasize to the bone marrow.
Collapse
Affiliation(s)
| | - Yawara Kawano
- Department of Hematology, Kumamoto University Hospital, Kumamoto, Japan
| | - Antonio Sacco
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, P.le Spedali Civili, n.1, 25123, Brescia, Italy
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Angelo Belotti
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Rossella Ribolla
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Marco Chiarini
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Viviana Giustini
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Diego Bertoli
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandra Sottini
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Monica Valotti
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Claudia Ghidini
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Federico Serana
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Malagola
- Adult Bone Marrow Transplantation Unit, ASST Spedali Civili di Brescia, University of Brescia, Brescia, Italy
| | - Luisa Imberti
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Domenico Russo
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Adult Bone Marrow Transplantation Unit, ASST Spedali Civili di Brescia, University of Brescia, Brescia, Italy
| | - Alessandro Montanelli
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Clinical Chemistry Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Giuseppe Rossi
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michaela R Reagan
- Maine Medical Center Research Institute, University of Maine, Scarborough, ME, USA
| | - Patricia Maiso
- Clinical and Translational Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Bruno Paiva
- Clinical and Translational Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Aldo M Roccaro
- Clinical Research Development and Phase I Unit, ASST Spedali Civili di Brescia, P.le Spedali Civili, n.1, 25123, Brescia, Italy.
- CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy.
| |
Collapse
|
69
|
Klein C, Bacac M, Umana P, Fingerle-Rowson G. Combination therapy with the type II anti-CD20 antibody obinutuzumab. Expert Opin Investig Drugs 2017; 26:1145-1162. [DOI: 10.1080/13543784.2017.1373087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Christian Klein
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Marina Bacac
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Pablo Umana
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Zurich, Zurich, Switzerland
| | | |
Collapse
|
70
|
Zhou H, Li Y, Liu B, Shan Y, Li Y, Zhao L, Su Z, Jia L. Downregulation of miR-224 and let-7i contribute to cell survival and chemoresistance in chronic myeloid leukemia cells by regulating ST3GAL IV expression. Gene 2017; 626:106-118. [DOI: 10.1016/j.gene.2017.05.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/30/2017] [Accepted: 05/11/2017] [Indexed: 12/24/2022]
|
71
|
Cai H, Zhou H, Miao Y, Li N, Zhao L, Jia L. MiRNA expression profiles reveal the involvement of miR-26a, miR-548l and miR-34a in hepatocellular carcinoma progression through regulation of ST3GAL5. J Transl Med 2017; 97:530-542. [PMID: 28218742 DOI: 10.1038/labinvest.2017.12] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/24/2016] [Accepted: 01/13/2017] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) have key roles in comprehensive physiological and pathological processes by targeting specific genes through translational repression. Identification of miRNAs related to metastasis enables us to obtain better insight into cancer development. In the current study, we investigated the miRNA expressional profiles in the highly invasive human hepatocellular carcinoma cell line MHCC97-H and MHCC97-L with lower metastatic potential using miRNA microarrays. By quantitative real-time PCR, we confirmed the results of miRNA experiments. Thirteen differentially expressed miRNAs were identified between MHCC97-H and MHCC97-L cells; and the same results were found in clinical samples. Using bioinformatic analysis and luciferase reporter assay, we found that ST3GAL5, a sialyltransferase gene, was the direct target of miR-26a, miR-548l and miR-34a. Engineered expression of miR-26a, miR-548l or miR-34a in MHCC97-H or MHCC97-L cells could significantly change their malignant behaviors and oncogenicity in in vitro and in vivo assays. Manipulated expression of ST3GAL5 also led to the alteration of the metastatic potential of MHCC97-H and MHCC97-L cells, in agreement with the effects of above three miRNAs. Altogether, our data indicate that the levels of these miRNAs may be used as biological markers for evaluating hepatocellular carcinoma progression. miR-26a, miR-548l and miR-34a, acting as tumor suppressors, may exert their effects by regulating ST3GAL5.
Collapse
Affiliation(s)
- Hongjiao Cai
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China.,Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning Province, China
| | - Huimin Zhou
- Department of Microbiology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Yuan Miao
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Nana Li
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Lifen Zhao
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
72
|
E-selectin ligands recognised by HECA452 induce drug resistance in myeloma, which is overcome by the E-selectin antagonist, GMI-1271. Leukemia 2017; 31:2642-2651. [PMID: 28439107 PMCID: PMC5729350 DOI: 10.1038/leu.2017.123] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 01/02/2023]
Abstract
Multiple myeloma (MM) is characterized by the clonal expansion and metastatic spread of malignant plasma cells to multiple sites in the bone marrow (BM). Recently, we implicated the sialyltransferase ST3Gal-6, an enzyme critical to the generation of E-selectin ligands, in MM BM homing and resistance to therapy. Since E-selectin is constitutively expressed in the BM microvasculature, we wished to establish the contribution of E-selectin ligands to MM biology. We report that functional E-selectin ligands are restricted to a minor subpopulation of MM cell lines which, upon expansion, demonstrate specific and robust interaction with recombinant E-selectin in vitro. Moreover, an increase in the mRNA levels of genes involved in the generation of E-selectin ligands was associated with inferior progression-free survival in the CoMMpass study. In vivo, E-selectin ligand-enriched cells induced a more aggressive disease and were completely insensitive to Bortezomib. Importantly, this resistance could be reverted by co-administration of GMI-1271, a specific glycomimetic antagonist of E-selectin. Finally, we report that E-selectin ligand-bearing cells are present in primary MM samples from BM and peripheral blood with a higher proportion seen in relapsed patients. This study provides a rationale for targeting E-selectin receptor/ligand interactions to overcome MM metastasis and chemoresistance.
Collapse
|
73
|
Zatula A, Dikic A, Mulder C, Sharma A, Vågbø CB, Sousa MML, Waage A, Slupphaug G. Proteome alterations associated with transformation of multiple myeloma to secondary plasma cell leukemia. Oncotarget 2017; 8:19427-19442. [PMID: 28038447 PMCID: PMC5386695 DOI: 10.18632/oncotarget.14294] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Plasma cell leukemia is a rare and aggressive plasma cell neoplasm that may either originate de novo (primary PCL) or by leukemic transformation of multiple myeloma (MM) to secondary PCL (sPCL). The prognosis of sPCL is very poor, and currently no standard treatment is available due to lack of prospective clinical studies. In an attempt to elucidate factors contributing to transformation, we have performed super-SILAC quantitative proteome profiling of malignant plasma cells collected from the same patient at both the MM and sPCL stages of the disease. 795 proteins were found to be differentially expressed in the MM and sPCL samples. Gene ontology analysis indicated a metabolic shift towards aerobic glycolysis in sPCL as well as marked down-regulation of enzymes involved in glycan synthesis, potentially mediating altered glycosylation of surface receptors. There was no significant change in overall genomic 5-methylcytosine or 5-hydroxymethylcytosine at the two stages, indicating that epigenetic dysregulation was not a major driver of transformation to sPCL. The present study constitutes the first attempt to provide a comprehensive map of the altered protein expression profile accompanying transformation of MM to sPCL in a single patient, identifying several candidate proteins that can be targeted by currently available small molecule drugs. Our dataset furthermore constitutes a reference dataset for further proteomic analysis of sPCL transformation.
Collapse
Affiliation(s)
- Alexey Zatula
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Aida Dikic
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Celine Mulder
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Present address: University of Utrecht, Utrecht, Holland
| | - Animesh Sharma
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Cathrine B Vågbø
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Mirta M L Sousa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Department of Hematology, Department of Medicine, St. Olav's Hospital, Trondheim, Norway
| | - Geir Slupphaug
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| |
Collapse
|
74
|
Kovacs Z, Simon A, Szabo Z, Nagy Z, Varoczy L, Pal I, Csanky E, Guttman A. Capillary electrophoresis analysis of N-glycosylation changes of serum paraproteins in multiple myeloma. Electrophoresis 2017; 38:2115-2123. [PMID: 28116769 DOI: 10.1002/elps.201700006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 01/17/2017] [Accepted: 01/18/2017] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is an immedicable malignancy of the human plasma cells producing abnormal antibodies (also referred to as paraproteins) leading to kidney problems and hyperviscosity syndrome. In this paper, we report on the N-glycosylation analysis of paraproteins from total human serum as well as the fragment crystallizable region (Fc ) and fragment antigen binding (Fab ) κ/λ light chain fractions of papain digested immunoglobulins from multiple myeloma patients. CE-LIF detection was used for the analysis of the N-glycans after endoglycosidase (PNGase F) mediated sugar release and fluorophore labeling (APTS). While characteristic N-glycosylation pattern differences were found between normal control and untreated, treated and remission stage multiple myeloma patient samples at the global serum level, less distinctive changes were observed at the immunoglobulin level. Principal component analysis adequately differentiated the four groups (control and three patient groups) on the basis of total serum N-glycosylation analysis. 12 N-glycan features showed statistically significant differences (p <0.05) among various stages of the disease in comparison to the control at the serum level, while only six features were identified with similar significance at the immunoglobulin level, including the analysis of the partitioned Fc fragment as well as the Fab κ and Fab λ chains.
Collapse
Affiliation(s)
- Zsuzsanna Kovacs
- Horváth Csaba Memorial Institute for Bioanalytical Research, University of Debrecen, Hungary
| | - Adam Simon
- Horváth Csaba Memorial Institute for Bioanalytical Research, University of Debrecen, Hungary
| | | | - Zsolt Nagy
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Laszlo Varoczy
- Department of Hematology, Institute for Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildiko Pal
- Department of Hematology, Institute for Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Andras Guttman
- Horváth Csaba Memorial Institute for Bioanalytical Research, University of Debrecen, Hungary.,MTA-PE Translational Glycomics Research Group, University of Pannonia, Veszprem, Hungary
| |
Collapse
|
75
|
Reagan MR, Lian JB, Rosen CJ, Stein GS. A perspective on malignancy in the marrow. J Cell Physiol 2017; 232:3218-3220. [PMID: 28206683 DOI: 10.1002/jcp.25860] [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: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 11/07/2022]
Abstract
Malignancies that grow in the bone marrow cause a cascade of devastating consequences and are almost always fatal. For researchers to make significant headway in finding cures and treatments for these tumors and the subsequent devastation of cancer-induced bone disease, novel strategies and creative solutions will have to be considered. Great progress has been made in treating hematologic and sold tumor malignancies that ultimately affect the bone marrow, although it is also obvious that multi-disciplinary teams are required to ultimately win the war on cancers that affect the bone. In this perspective, we review recent advances in the identification of molecular and cellular targets in the bone marrow niche.
Collapse
Affiliation(s)
- Michaela R Reagan
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine
- Tufts University School of Medicine, Boston, Maine
| | - Jane B Lian
- Department of Biochemistry and the University of Vermont Cancer Center, the University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Clifford J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, Maine
- Tufts University School of Medicine, Boston, Maine
| | - Gary S Stein
- Department of Biochemistry and the University of Vermont Cancer Center, the University of Vermont Larner College of Medicine, Burlington, Vermont
| |
Collapse
|
76
|
Sun M, Zhao X, Liang L, Pan X, Lv H, Zhao Y. Sialyltransferase ST3GAL6 mediates the effect of microRNA-26a on cell growth, migration, and invasion in hepatocellular carcinoma through the protein kinase B/mammalian target of rapamycin pathway. Cancer Sci 2017; 108:267-276. [PMID: 27906498 PMCID: PMC5329153 DOI: 10.1111/cas.13128] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/20/2016] [Accepted: 11/22/2016] [Indexed: 12/22/2022] Open
Abstract
Aberrant sialylation profiles on the cell surface have been recognized for their potential diagnostic value in identifying the regulation of tumor properties in several cancers, including hepatocellular carcinoma (HCC). Recently, increasing evidence has suggested that the deregulation of microRNA (miRNA) is a common feature in human cancers. In this study, we found obvious upregulation of sialyltransferase ST3GAL6 both in HCC cell lines and in tissue samples. The altered expression of ST3GAL6 was found to correlate with cell proliferation, migration, and invasion ability in HCC. Further investigation showed that miR-26a negatively regulated ST3GAL6, inducing the suppression of cell proliferation, migration, and invasion in vitro. Moreover, we identified the protein kinase B/mammalian target of rapamycin (Akt/mTOR) pathway as the target of ST3GAL6 based on Western blot analysis. Analysis of a xenograft mouse model showed that miR-26a significantly reduced tumor growth by suppressing activation of the Akt/mTOR pathway by directly targeting ST3GAL6. In conclusion, these data indicate that ST3GAL6 promotes cell growth, migration, and invasion and mediates the effect of miR-26a through the Akt/mTOR signaling pathway in HCC.
Collapse
Affiliation(s)
- Mingming Sun
- Department of General SurgeryThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xuzi Zhao
- School of Basic Medical SciencesHebei Medical UniversityShijiazhuangChina
| | - Leilei Liang
- Department of General SurgeryThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xufeng Pan
- Department of General SurgeryThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Hao Lv
- Department of OrthopedicsThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yongfu Zhao
- Department of General SurgeryThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
77
|
Mittermayr S, Lê GN, Clarke C, Millán Martín S, Larkin AM, O’Gorman P, Bones J. Polyclonal Immunoglobulin G N-Glycosylation in the Pathogenesis of Plasma Cell Disorders. J Proteome Res 2016; 16:748-762. [DOI: 10.1021/acs.jproteome.6b00768] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Stefan Mittermayr
- NIBRT−The
National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock Co., Dublin A94 X099, Ireland
| | - Giao N. Lê
- NIBRT−The
National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock Co., Dublin A94 X099, Ireland
- Department
of Haematology, Mater Misericordiae University Hospital, Dublin D07 R2WY, Ireland
- National
Institute for Cellular Biotechnology, Dublin City University, Dublin D09 NR58, Ireland
| | - Colin Clarke
- NIBRT−The
National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock Co., Dublin A94 X099, Ireland
| | - Silvia Millán Martín
- NIBRT−The
National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock Co., Dublin A94 X099, Ireland
| | - Anne-Marie Larkin
- National
Institute for Cellular Biotechnology, Dublin City University, Dublin D09 NR58, Ireland
| | - Peter O’Gorman
- Department
of Haematology, Mater Misericordiae University Hospital, Dublin D07 R2WY, Ireland
| | - Jonathan Bones
- NIBRT−The
National Institute for Bioprocessing Research and Training, Foster Avenue, Mount Merrion, Blackrock Co., Dublin A94 X099, Ireland
| |
Collapse
|
78
|
Szabo R, Skropeta D. Advancement of Sialyltransferase Inhibitors: Therapeutic Challenges and Opportunities. Med Res Rev 2016; 37:219-270. [DOI: 10.1002/med.21407] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Rémi Szabo
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
- Centre for Medical & Molecular Bioscience; University of Wollongong; Wollongong NSW 2522 Australia
| |
Collapse
|
79
|
Pagnotti GM, Chan ME, Adler BJ, Shroyer KR, Rubin J, Bain SD, Rubin CT. Low intensity vibration mitigates tumor progression and protects bone quantity and quality in a murine model of myeloma. Bone 2016; 90:69-79. [PMID: 27262776 PMCID: PMC4970889 DOI: 10.1016/j.bone.2016.05.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 05/13/2016] [Accepted: 05/31/2016] [Indexed: 01/18/2023]
Abstract
Myeloma facilitates destruction of bone and marrow. Since physical activity encourages musculoskeletal preservation we evaluated whether low-intensity vibration (LIV), a means to deliver mechanical signals, could protect bone and marrow during myeloma progression. Immunocompromised-mice (n=25) were injected with human-myeloma cells, while 8 (AC) were saline-injected. Myeloma-injected mice (LIV; n=13) were subjected to daily-mechanical loading (15min/d; 0.3g @ 90Hz) while 12 (MM) were sham-handled. At 8w, femurs had 86% less trabecular bone volume fraction (BV/TV) in MM than in AC, yet only a 21% decrease in LIV was observed in comparison to AC, reflecting a 76% increase versus MM. Cortical BV was 21% and 15% lower in MM and LIV, respectively, than in AC; LIV showing 30% improvement over MM. Similar outcomes were observed in the axial skeleton, showing a 35% loss in MM with a 27% improved retention of bone in the L5 of LIV-treated mice as compared to MM. Transcortical-perforations in the femur from myeloma-induced osteolysis were 9× higher in MM versus AC, reduced by 57% in LIV. Serum-TRACP5b, 61% greater in MM versus AC, rose by 33% in LIV compared to AC, a 45% reduction in activity when compared to MM. Histomorphometric analyses of femoral trabecular bone demonstrated a 70% elevation in eroded surfaces of MM versus AC, while measures in LIV were 58% below those in MM. 72% of marrow in the femur of MM mice contained tumor, contrasted by a 31% lower burden in LIV. MM mice (42%) presented advanced-stage necrosis of tibial marrow while present in just 8% of LIV. Myeloma infiltration inversely correlated to measures of bone quality, while LIV slowed the systemic, myeloma-associated decline in bone quality and inhibited tumor progression through the hindlimbs.
Collapse
Affiliation(s)
- Gabriel M Pagnotti
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794-5281, United States
| | - M Ete Chan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794-5281, United States
| | - Benjamin J Adler
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794-5281, United States
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-2580, United States
| | - Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, United States
| | - Steven D Bain
- Department of Orthopedics & Sports Medicine, University of Washington, Seattle, WA 98104-2499, United States
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794-5281, United States.
| |
Collapse
|
80
|
Lonial S, Kaufman J, Reece D, Mateos MV, Laubach J, Richardson P. Update on elotuzumab, a novel anti-SLAMF7 monoclonal antibody for the treatment of multiple myeloma. Expert Opin Biol Ther 2016; 16:1291-301. [DOI: 10.1080/14712598.2016.1221920] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jonathan Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Donna Reece
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Canada
| | - Maria-Victoria Mateos
- Haematology Department, Complejo Asistencial Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Jacob Laubach
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
81
|
Zheng Y, Wang Q, Li T, Qian J, Lu Y, Li Y, Bi E, Reu F, Qin Y, Drazba J, Hsi E, Yang J, Cai Z, Yi Q. Role of Myeloma-Derived MIF in Myeloma Cell Adhesion to Bone Marrow and Chemotherapy Response. J Natl Cancer Inst 2016; 108:djw131. [PMID: 27381622 PMCID: PMC5241902 DOI: 10.1093/jnci/djw131] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/15/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) remains an incurable cancer characterized by accumulation of malignant plasma cells in the bone marrow (BM). The mechanism underlying MM homing to BM is poorly elucidated. METHODS The clinical significance of migration inhibitory factor (MIF) expression was examined by analyzing six independent gene expression profile databases of primary MM cells using the Student's t test and Kaplan-Meier test. Enzyme-linked immunosorbent assay was used to examine MIF expression. In vivo bioluminescent imaging was used to determine MM cell localization and treatment efficacy in human MM xenograft mouse models, with three to four mice per group. MM cell attachment to BM stromal cells (BMSCs) was monitored by cell adhesion assay. MIF regulation of the expression of adhesion molecules was determined by chromatin immunoprecipitation (ChIP) assay. Statistical tests were two-sided. RESULTS High levels of MIF were detected in MM BM (MIF level in BM plasma: healthy = 10.72 ± 5.788 ng/mL, n = 5; MM = 1811 ± 248.7 ng/mL, n = 10; P < .001) and associated with poor survival of patients (Kaplan-Meier test for MM OS: 87 MIF(high) patients, 86 MIF(low) patients, P = .02). Knocking down MIF impaired MM cell adhesion to BMSCs in vitro and led to formation of extramedullary tumors in SCID mice. MIF acted through surface receptor CXCR4 and adaptor COPS5 to regulate the expression of adhesion molecules ALCAM, ITGAV, and ITGB5 on MM cells. More importantly, MIF-deficient MM cells were sensitive to chemotherapy in vitro when cocultured with BMSCs and in vivo. MIF inhibitor 4-IPP sensitized MM cells to chemotherapy. CONCLUSIONS MIF is an important player and a novel therapeutic target in MM. Inhibiting MIF activity will sensitize MM cells to chemotherapy.
Collapse
MESH Headings
- Activated-Leukocyte Cell Adhesion Molecule/genetics
- Animals
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antineoplastic Agents, Alkylating/pharmacology
- Apoptosis/drug effects
- Autocrine Communication
- Bone Marrow/metabolism
- Bortezomib/pharmacology
- COP9 Signalosome Complex
- Cell Adhesion/drug effects
- Cell Adhesion/genetics
- Cell Line, Tumor
- Chemotaxis/genetics
- Coculture Techniques
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression
- Gene Knockdown Techniques
- Heterografts
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- Intramolecular Oxidoreductases/antagonists & inhibitors
- Intramolecular Oxidoreductases/genetics
- Intramolecular Oxidoreductases/metabolism
- Macrophage Migration-Inhibitory Factors/antagonists & inhibitors
- Macrophage Migration-Inhibitory Factors/genetics
- Macrophage Migration-Inhibitory Factors/metabolism
- Melphalan/pharmacology
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, SCID
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- Neoplasm Transplantation
- Peptide Hydrolases/metabolism
- Plasma Cells/metabolism
- Pyrimidines/pharmacology
- RNA, Messenger/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
Collapse
Affiliation(s)
- Yuhuan Zheng
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Qiang Wang
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Tianshu Li
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Jianfei Qian
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Yong Lu
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Yi Li
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Enguang Bi
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Frederic Reu
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Yu Qin
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Judy Drazba
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Eric Hsi
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Jing Yang
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Zhen Cai
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Qing Yi
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| |
Collapse
|
82
|
Friedel M, André S, Goldschmidt H, Gabius HJ, Schwartz-Albiez R. Galectin-8 enhances adhesion of multiple myeloma cells to vascular endothelium and is an adverse prognostic factor. Glycobiology 2016; 26:1048-1058. [PMID: 27287437 DOI: 10.1093/glycob/cww066] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/24/2016] [Accepted: 05/29/2016] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma is characterized by abnormal infiltration of malignant plasma cells into bone marrow. Testing the hypothesis that bivalent galectin-8 (Gal-8) may influence homing of myeloma cells to vascular endothelium as a key prerequisite for infiltration, we analyzed the two Gal-8 splice variants (Gal-8S, Gal-8L). They differ in the length of their linker peptide connecting the two lectin domains. Both Gal-8 isoforms bind to cells of the myeloma lines Gal-8+ MOLP-8 and Gal-8- LP-1 in a glycan-inhibitable manner. Both Gal-8 isoforms led to enhanced adhesion of myeloma cells to vascular endothelium under dynamic shear stress conditions, Gal-8L (by more than 40-fold) even stronger than Gal-8S. Additional treatment of endothelial cells with tumour necrosis factor prior to the dynamic shear stress assay entailed an almost 100-fold enhanced adhesion of myeloma cells without addition of Gal-8 variants and a further 1.5-1.7-fold enhancement by addition of Gal-8 variants. We also found that elevated expression of Gal-8 in native multiple myeloma cells is an adverse prognostic factor for overall and event-free survival using patients' gene expression profile data of the total therapy 2 and 3 myeloma studies. Also, elevated concentrations of Gal-8 were detected (45%, 19/42 patients) in sera of multiple myeloma patients compared to those of healthy, age-matched donors. Both experimental and clinical data strongly point to the significance of Gal-8 for multiple myeloma development.
Collapse
Affiliation(s)
- Myriam Friedel
- Clinical Cooperation Unit Applied Tumor Immunity, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| | - Sabine André
- Institut für Physiologische Chemie, Tierärztliche Fakultät, Ludwig-Maximilians-Universität München, 80539 München, Germany
| | - Hartmut Goldschmidt
- Medizinische Klinik V, Universitätsklinikum Heidelberg und Nationales Centrum für Tumorerkrankungen Heidelberg, 69120 Heidelberg, Germany
| | - Hans-Joachim Gabius
- Institut für Physiologische Chemie, Tierärztliche Fakultät, Ludwig-Maximilians-Universität München, 80539 München, Germany
| | - Reinhard Schwartz-Albiez
- Clinical Cooperation Unit Applied Tumor Immunity, Deutsches Krebsforschungszentrum, 69120 Heidelberg, Germany
| |
Collapse
|
83
|
Wu Y, Lan C, Ren D, Chen GY. Induction of Siglec-1 by Endotoxin Tolerance Suppresses the Innate Immune Response by Promoting TGF-β1 Production. J Biol Chem 2016; 291:12370-82. [PMID: 27129263 DOI: 10.1074/jbc.m116.721258] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 12/22/2022] Open
Abstract
Sepsis is one of the leading causes of death worldwide. Although the prevailing theory for the sepsis syndrome is a condition of uncontrolled inflammation in response to infection, sepsis is increasingly being recognized as an immunosuppressive state known as endotoxin tolerance. We found sialylation of cell surface was significantly increased on LPS-induced tolerant cells; knockdown of Neu1 in macrophage cell line RAW 264.7 cells resulted in enhanced LPS-induced tolerance, whereas overexpression of Neu1 or treatment with sialidase abrogated LPS-induced tolerance, as defined by measuring TNF-α levels in the culture supernatants. We also found that the expression of Siglec-1 (a member of sialic acid-binding Ig (I)-like lectin family members, the predominant sialic acid-binding proteins on cell surface) was specifically up-regulated in endotoxin tolerant cells and the induction of Siglec-1 suppresses the innate immune response by promoting TGF-β1 production. The enhanced TGF-β1 production by Siglec-1 was significantly attenuated by spleen tyrosine kinase (Syk) inhibitor. Knockdown of siglec-1 in RAW 264.7 cells resulted in inhibiting the production of TGF-β1 by ubiquitin-dependent degradation of Syk. Mechanistically, Siglec-1 associates with adaptor protein DNAX-activation protein of 12 kDa (DAP12) and transduces a signal to Syk to control the production of TGF-β1 in endotoxin tolerance. Thus, Siglec-1 plays an important role in the development of endotoxin tolerance and targeted manipulation of this process could lead to a new therapeutic opportunity for patients with sepsis.
Collapse
Affiliation(s)
- Yin Wu
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Chao Lan
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Dongren Ren
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| | - Guo-Yun Chen
- From the Children's Foundation Research Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee 38103
| |
Collapse
|
84
|
Natoni A, Macauley MS, O'Dwyer ME. Targeting Selectins and Their Ligands in Cancer. Front Oncol 2016; 6:93. [PMID: 27148485 PMCID: PMC4834419 DOI: 10.3389/fonc.2016.00093] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022] Open
Abstract
Aberrant glycosylation is a hallmark of cancer cells with increased evidence pointing to a role in tumor progression. In particular, aberrant sialylation of glycoproteins and glycolipids has been linked to increased immune cell evasion, drug evasion, drug resistance, tumor invasiveness, and vascular dissemination, leading to metastases. Hypersialylation of cancer cells is largely the result of overexpression of sialyltransferases (STs). Differentially, humans express twenty different STs in a tissue-specific manner, each of which catalyzes the attachment of sialic acids via different glycosidic linkages (α2-3, α2-6, or α2-8) to the underlying glycan chain. One important mechanism whereby overexpression of STs contributes to an enhanced metastatic phenotype is via the generation of selectin ligands. Selectin ligand function requires the expression of sialyl-Lewis X and its structural isomer sialyl-Lewis A, which are synthesized by the combined action of alpha α1-3-fucosyltransferases, α2-3-sialyltransferases, β1-4-galactosyltranferases, and N-acetyl-β-glucosaminyltransferases. The α2-3-sialyltransferases ST3Gal4 and ST3Gal6 are critical to the generation of functional E- and P-selectin ligands and overexpression of these STs have been linked to increased risk of metastatic disease in solid tumors and poor outcome in multiple myeloma. Thus, targeting selectins and their ligands as well as the enzymes involved in their generation, in particular STs, could be beneficial to many cancer patients. Potential strategies include ST inhibition and the use of selectin antagonists, such as glycomimetic drugs and antibodies. Here, we review ongoing efforts to optimize the potency and selectivity of ST inhibitors, including the potential for targeted delivery approaches, as well as evaluate the potential utility of selectin inhibitors, which are now in early clinical development.
Collapse
Affiliation(s)
- Alessandro Natoni
- Biomedical Sciences, National University of Ireland Galway , Galway , Ireland
| | - Matthew S Macauley
- Department of Chemical Physiology, The Scripps Research Institute , La Jolla, CA , USA
| | - Michael E O'Dwyer
- Biomedical Sciences, National University of Ireland Galway, Galway, Ireland; School of Medicine, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
85
|
Lee D, Wang YH, Kalaitzidis D, Ramachandran J, Eda H, Sykes DB, Raje N, Scadden DT. Endogenous transmembrane protein UT2 inhibits pSTAT3 and suppresses hematological malignancy. J Clin Invest 2016; 126:1300-10. [PMID: 26927669 PMCID: PMC4811118 DOI: 10.1172/jci84620] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 01/14/2016] [Indexed: 12/27/2022] Open
Abstract
Regulation of STAT3 activation is critical for normal and malignant hematopoietic cell proliferation. Here, we have reported that the endogenous transmembrane protein upstream-of-mTORC2 (UT2) negatively regulates activation of STAT3. Specifically, we determined that UT2 interacts directly with GP130 and inhibits phosphorylation of STAT3 on tyrosine 705 (STAT3Y705). This reduces cytokine signaling including IL6 that is implicated in multiple myeloma and other hematopoietic malignancies. Modulation of UT2 resulted in inverse effects on animal survival in myeloma models. Samples from multiple myeloma patients also revealed a decreased copy number of UT2 and decreased expression of UT2 in genomic and transcriptomic analyses, respectively. Together, these studies identify a transmembrane protein that functions to negatively regulate cytokine signaling through GP130 and pSTAT3Y705 and is molecularly and mechanistically distinct from the suppressors of cytokine signaling (SOCS) family of genes. Moreover, this work provides evidence that perturbations of this activation-dampening molecule participate in hematologic malignancies and may serve as a key determinant of multiple myeloma pathophysiology. UT2 is a negative regulator shared across STAT3 and mTORC2 signaling cascades, functioning as a tumor suppressor in hematologic malignancies driven by those pathways.
Collapse
Affiliation(s)
- Dongjun Lee
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Ying-Hua Wang
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Demetrios Kalaitzidis
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | | | - Homare Eda
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David B. Sykes
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Noopur Raje
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David T. Scadden
- Center for Regenerative Medicine and
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology and
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
86
|
Fairfield H, Falank C, Avery L, Reagan MR. Multiple myeloma in the marrow: pathogenesis and treatments. Ann N Y Acad Sci 2016; 1364:32-51. [PMID: 27002787 PMCID: PMC4806534 DOI: 10.1111/nyas.13038] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple myeloma (MM) is a B cell malignancy resulting in osteolytic lesions and fractures. In the disease state, bone healing is limited owing to increased osteoclastic and decreased osteoblastic activity, as well as an MM-induced forward-feedback cycle where bone-embedded growth factors further enhance tumor progression as bone is resorbed. Recent work on somatic mutation in MM tumors has provided insight into cytogenetic changes associated with this disease; the initiating driver mutations causing MM are diverse because of the complexity and multitude of mutations inherent in MM tumor cells. This manuscript provides an overview of MM pathogenesis by summarizing cytogenic changes related to oncogenes and tumor suppressors associated with MM, reviewing risk factors, and describing the disease progression from monoclonal gammopathy of undetermined significance to overt MM. It also highlights the importance of the bone marrow microenvironment (BMM) in the establishment and progression of MM, as well as associated MM-induced bone disease, and the relationship of the bone marrow to current and future therapeutics. This review highlights why understanding the basic biology of the healthy and diseased BMM is crucial in the quest for better treatments and work toward a cure for genetically diverse diseases such as MM.
Collapse
Affiliation(s)
| | | | | | - Michaela R Reagan
- Maine Medical Center Research Institute, Scarborough, Maine
- University of Maine, Orono, Maine
| |
Collapse
|
87
|
Abstract
Unprecedented advances in multiple myeloma (MM) therapy during the last 15 years are predominantly based on our increasing understanding of the pathophysiologic role of the bone marrow (BM) microenvironment. Indeed, new treatment paradigms, which incorporate thalidomide, immunomodulatory drugs (IMiDs), and proteasome inhibitors, target the tumor cell as well as its BM microenvironment. Ongoing translational research aims to understand in more detail how disordered BM-niche functions contribute to MM pathogenesis and to identify additional derived targeting agents. One of the most exciting advances in the field of MM treatment is the emergence of immune therapies including elotuzumab, daratumumab, the immune checkpoint inhibitors, Bispecific T-cell engagers (BiTes), and Chimeric antigen receptor (CAR)-T cells. This chapter will review our knowledge on the pathophysiology of the BM microenvironment and discuss derived novel agents that hold promise to further improve outcome in MM.
Collapse
Affiliation(s)
- Michele Moschetta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yawara Kawano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Klaus Podar
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
88
|
Falank C, Fairfield H, Reagan MR. Signaling Interplay between Bone Marrow Adipose Tissue and Multiple Myeloma cells. Front Endocrinol (Lausanne) 2016; 7:67. [PMID: 27379019 PMCID: PMC4911365 DOI: 10.3389/fendo.2016.00067] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 06/03/2016] [Indexed: 01/04/2023] Open
Abstract
In the year 2000, Hanahan and Weinberg (1) defined the six Hallmarks of Cancer as: self-sufficiency in growth signals, evasion of apoptosis, insensitivity to antigrowth mechanisms, tissue invasion and metastasis, limitless replicative potential, and sustained angiogenesis. Eleven years later, two new Hallmarks were added to the list (avoiding immune destruction and reprograming energy metabolism) and two new tumor characteristics (tumor-promoting inflammation and genome instability and mutation) (2). In multiple myeloma (MM), a destructive cancer of the plasma cell that grows predominantly in the bone marrow (BM), it is clear that all these hallmarks and characteristics are in play, contributing to tumor initiation, drug resistance, disease progression, and relapse. Bone marrow adipose tissue (BMAT) is a newly recognized contributor to MM oncogenesis and disease progression, potentially affecting MM cell metabolism, immune action, inflammation, and influences on angiogenesis. In this review, we discuss the confirmed and hypothetical contributions of BMAT to MM development and disease progression. BMAT has been understudied due to technical challenges and a previous lack of appreciation for the endocrine function of this tissue. In this review, we define the dynamic, responsive, metabolically active BM adipocyte. We then describe how BMAT influences MM in terms of: lipids/metabolism, hypoxia/angiogenesis, paracrine or endocrine signaling, and bone disease. We then discuss the connection between BMAT and systemic inflammation and potential treatments to inhibit the feedback loops between BM adipocytes and MM cells that support MM progression. We aim for researchers to use this review to guide and help prioritize their experiments to develop better treatments or a cure for cancers, such as MM, that associate with and may depend on BMAT.
Collapse
Affiliation(s)
- Carolyne Falank
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Heather Fairfield
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Michaela R. Reagan
- Reagan Laboratory, Maine Medical Center Research Institute, Scarborough, ME, USA
- School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- School of Medicine, Tufts University, Boston, MA, USA
- *Correspondence: Michaela R. Reagan,
| |
Collapse
|
89
|
Reagan MR, Rosen CJ. Navigating the bone marrow niche: translational insights and cancer-driven dysfunction. Nat Rev Rheumatol 2015; 12:154-68. [PMID: 26607387 DOI: 10.1038/nrrheum.2015.160] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The bone marrow niche consists of stem and progenitor cells destined to become mature cells such as haematopoietic elements, osteoblasts or adipocytes. Marrow cells, influenced by endocrine, paracrine and autocrine factors, ultimately function as a unit to regulate bone remodelling and haematopoiesis. Current evidence highlights that the bone marrow niche is not merely an anatomic compartment; rather, it integrates the physiology of two distinct organ systems, the skeleton and the marrow. The niche has a hypoxic microenvironment that maintains quiescent haematopoietic stem cells (HSCs) and supports glycolytic metabolism. In response to biochemical cues and under the influence of neural, hormonal, and biochemical factors, marrow stromal elements, such as mesenchymal stromal cells (MSCs), differentiate into mature, functioning cells. However, disruption of the niche can affect cellular differentiation, resulting in disorders ranging from osteoporosis to malignancy. In this Review, we propose that the niche reflects the vitality of two tissues - bone and blood - by providing a unique environment for stem and stromal cells to flourish while simultaneously preventing disproportionate proliferation, malignant transformation or loss of the multipotent progenitors required for healing, functional immunity and growth throughout an organism's lifetime. Through a fuller understanding of the complexity of the niche in physiologic and pathologic states, the successful development of more-effective therapeutic approaches to target the niche and its cellular components for the treatment of rheumatic, endocrine, neoplastic and metabolic diseases becomes achievable.
Collapse
Affiliation(s)
- Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Centre Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | - Clifford J Rosen
- Center for Molecular Medicine, Maine Medical Centre Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| |
Collapse
|
90
|
Pearce OMT, Läubli H. Sialic acids in cancer biology and immunity. Glycobiology 2015; 26:111-28. [DOI: 10.1093/glycob/cwv097] [Citation(s) in RCA: 259] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023] Open
|
91
|
Glycosylation-Based Serum Biomarkers for Cancer Diagnostics and Prognostics. BIOMED RESEARCH INTERNATIONAL 2015; 2015:490531. [PMID: 26509158 PMCID: PMC4609776 DOI: 10.1155/2015/490531] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/28/2015] [Accepted: 05/31/2015] [Indexed: 12/13/2022]
Abstract
Cancer is the second most common cause of death in developed countries with approximately 14 million newly diagnosed individuals and over 6 million cancer-related deaths in 2012. Many cancers are discovered at a more advanced stage but better survival rates are correlated with earlier detection. Current clinically approved cancer biomarkers are most effective when applied to patients with widespread cancer. Single biomarkers with satisfactory sensitivity and specificity have not been identified for the most common cancers and some biomarkers are ineffective for the detection of early stage cancers. Thus, novel biomarkers with better diagnostic and prognostic performance are required. Aberrant protein glycosylation is well known hallmark of cancer and represents a promising source of potential biomarkers. Glycoproteins enter circulation from tissues or blood cells through active secretion or leakage and patient serum is an attractive option as a source for biomarkers from a clinical and diagnostic perspective. A plethora of technical approaches have been developed to address the challenges of glycosylation structure detection and determination. This review summarises currently utilised glycoprotein biomarkers and novel glycosylation-based biomarkers from the serum glycoproteome under investigation as cancer diagnostics and for monitoring and prognostics and includes details of recent high throughput and other emerging glycoanalytical techniques.
Collapse
|
92
|
Chen J, Fang M, Zhao YP, Yi CH, Ji J, Cheng C, Wang MM, Gu X, Sun QS, Chen XL, Gao CF. Serum N-Glycans: A New Diagnostic Biomarker for Light Chain Multiple Myeloma. PLoS One 2015; 10:e0127022. [PMID: 26075387 PMCID: PMC4468189 DOI: 10.1371/journal.pone.0127022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/10/2015] [Indexed: 01/29/2023] Open
Abstract
The aim of this study was to evaluate the diagnostic and differential diagnostic power of serum N-glycans for light chain multiple myeloma (LCMM). A total of 167 cases of subjects, including 42 LCMM, 42 IgG myeloma, 41 IgA myeloma, and 42 healthy controls were recruited in this study. DNA sequencer-assisted fluorophore-assisted capillary electrophoresis (DSA-FACE) was applied to determine the quantitive abundance of serum N-glycans. The core fucosylated, bisecting and sialylated modifications were analyzed in serum of LCMM patients (n=20) and healthy controls (n=20) randomly selected from the same cohort by lectin blot. Moreover, serum sialic acid (SA) level was measured by enzymatic method. We found two N-glycan structures (NG1A2F, Peak3; NA2FB, Peak7) showed the optimum diagnostic efficacy with area under the ROC curve (AUC) 0.939 and 0.940 between LCMM and healthy control. The sensitivity and specificity of Peak3 were 88.1% and 92.9%, while Peak7 were 92.9% and 97.6%, respectively. The abundance of Peak3 could differentiate LCMM from IgG myeloma with AUC 0.899, sensitivity 100% and specificity 76.2%, and Peak7 could be used to differentiate LCMM from IgA myeloma with AUC 0.922, sensitivity 92.9% and specificity 82.9%. Serum SA level was significantly higher in patients with LCMM than that in healthy controls. Moreover, the decreased core fucosylation, bisecting and increased sialylation characters of serum glycoproteins were observed in patients with LCMM. We concluded that serum N-glycan could provide a simple, reliable and non-invasive biomarker for LCMM diagnosis and abnormal glycosylation might imply a new potential therapeutic target in LCMM.
Collapse
Affiliation(s)
- Jie Chen
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Meng Fang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yun-Peng Zhao
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Chang-Hong Yi
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jun Ji
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Cheng Cheng
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Meng-Meng Wang
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xing Gu
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Quan-Sheng Sun
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Ling Chen
- Department of Hematology, Shanghai Zhabei District Central Hospital, Shanghai, China
| | - Chun-Fang Gao
- Department of Laboratory Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
- * E-mail:
| |
Collapse
|
93
|
New therapeutic targets for cancer bone metastasis. Trends Pharmacol Sci 2015; 36:360-73. [PMID: 25962679 DOI: 10.1016/j.tips.2015.04.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 12/18/2022]
Abstract
Bone metastases are dejected consequences of many types of tumors including breast, prostate, lung, kidney, and thyroid cancers. This complicated process begins with the successful tumor cell epithelial-mesenchymal transition, escape from the original site, and penetration into the circulation. The homing of tumor cells to the bone depends on both tumor-intrinsic traits and various molecules supplied by the bone metastatic niche. The colonization and growth of cancer cells in the osseous environment, which awaken their dormancy to form micro- and macro-metastasis, involve an intricate interaction between the circulating tumor cells and local bone cells including osteoclasts, osteoblasts, adipocytes, and macrophages. We discuss the most recent advances in the identification of new molecules and novel mechanisms during each step of bone metastasis that may serve as promising therapeutic targets.
Collapse
|
94
|
Sialic acids: biomarkers in endocrinal cancers. Glycoconj J 2015; 32:79-85. [PMID: 25777812 DOI: 10.1007/s10719-015-9577-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 02/11/2015] [Accepted: 02/18/2015] [Indexed: 12/20/2022]
Abstract
Sialylations are post translational modification of proteins and lipids that play important role in recognition, signaling, immunological response and cell-cell interaction. Improper sialylations due to altered sialyl transferases, sialidases, gene structure and expression, sialic acid metabolism however lead to diseases and thus sialic acids form an important biomarker in disease. In the endocrinal biology such improper sialylations including altered expression of sialylated moieties have been shown to be associated with disorders. Cancer still remains to be the major cause of global death and the cancer of the endocrine organs suffer from the dearth of appropriate markers for disease prediction at the early stage and monitoring. This review is aimed at evaluating the role of sialic acids as markers in endocrinal disorders with special reference to cancer of the endocrine organs. The current study is summarized under the following headings of altered sialylations in endocrinal cancer of the (i) ovary (ii) pancreas (iii) thyroid (iv) adrenal and (v) pituitary gland. Studies in expression of sialic acid in testis cancer are limited. The future scope of this review remains in the targeting of endocrinal cancer by targeting altered sialylation which is a common expression associated with endocrinal cancer.
Collapse
|
95
|
Glavey SV, Huynh D, Reagan MR, Manier S, Moschetta M, Kawano Y, Roccaro AM, Ghobrial IM, Joshi L, O'Dwyer ME. The cancer glycome: carbohydrates as mediators of metastasis. Blood Rev 2015; 29:269-79. [PMID: 25636501 DOI: 10.1016/j.blre.2015.01.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/06/2015] [Accepted: 01/16/2015] [Indexed: 12/30/2022]
Abstract
Glycosylation is a frequent post-translational modification which results in the addition of carbohydrate determinants, "glycans", to cell surface proteins and lipids. These glycan structures form the "glycome" and play an integral role in cell-cell and cell-matrix interactions through modulation of adhesion and cell trafficking. Glycosylation is increasingly recognized as a modulator of the malignant phenotype of cancer cells, where the interaction between cells and the tumor micro-environment is altered to facilitate processes such as drug resistance and metastasis. Changes in glycosylation of cell surface adhesion molecules such as selectin ligands, integrins and mucins have been implicated in the pathogenesis of several solid and hematological malignancies, often with prognostic implications. In this review we focus on the functional significance of alterations in cancer cell glycosylation, in terms of cell adhesion, trafficking and the metastatic cascade and provide insights into the prognostic and therapeutic implications of recent findings in this fast-evolving niche.
Collapse
Affiliation(s)
- Siobhan V Glavey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Glycoscience Research Group, National University of Ireland, Galway, Ireland.
| | - Daisy Huynh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Michaela R Reagan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Salomon Manier
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Michele Moschetta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Yawara Kawano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Aldo M Roccaro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Lokesh Joshi
- Glycoscience Research Group, National University of Ireland, Galway, Ireland.
| | - Michael E O'Dwyer
- Glycoscience Research Group, National University of Ireland, Galway, Ireland; Department of Hematology National University of Ireland, Galway and Galway University Hospital, Ireland.
| |
Collapse
|