1
|
Sharafi Monfared M, Nazmi S, Parhizkar F, Jafari D. Soluble B7 and TNF family in colorectal cancer: Serum level, prognostic and treatment value. Hum Immunol 2025; 86:111232. [PMID: 39793378 DOI: 10.1016/j.humimm.2025.111232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Soluble immune checkpoints (sIC) are crucial factors in the immune system. They regulate immune responses by transforming intercellular signals via binding to their membrane-bound receptor or ligand. Moreover, soluble ICs are vital in immune regulation, cancer development, and prognosis. They can be identified and measured in various tumor microenvironments. Recently, sICs have become increasingly important in clinically assessing malignancies like colorectal cancer (CRC) patients. This review explores the evolving role of the soluble B7 family and soluble tumor necrosis factor (TNF) superfamily members in predicting disease progression, treatment response, and overall patient outcomes in CRC. We comprehensively analyze the diagnostic and prognostic potential of soluble immune checkpoints in CRC. Understanding the role of these soluble immune checkpoints in CRC management and their potential as targets for precision medicine approaches can be critical for improving outcomes for patients with colorectal cancer.
Collapse
Affiliation(s)
- Mohanna Sharafi Monfared
- Student's Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Sina Nazmi
- Student's Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran; School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Forough Parhizkar
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Davood Jafari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
2
|
Wang C, Su NW, Hsu K, Kao CW, Chang MC, Chang YF, Lim KH, Chiang YH, Chang YC, Sung MT, Wu HH, Chen CG. The implication of serum HLA-G in angiogenesis of multiple myeloma. Mol Med 2024; 30:86. [PMID: 38877399 PMCID: PMC11177474 DOI: 10.1186/s10020-024-00860-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Despite the advances of therapies, multiple myeloma (MM) remains an incurable hematological cancer that most patients experience relapse. Tumor angiogenesis is strongly correlated with cancer relapse. Human leukocyte antigen G (HLA-G) has been known as a molecule to suppress angiogenesis. We aimed to investigate whether soluble HLA-G (sHLA-G) was involved in the relapse of MM. METHODS We first investigated the dynamics of serum sHLA-G, vascular endothelial growth factor (VEGF) and interleukin 6 (IL-6) in 57 successfully treated MM patients undergoing remission and relapse. The interactions among these angiogenesis-related targets (sHLA-G, VEGF and IL-6) were examined in vitro. Their expression at different oxygen concentrations was investigated using a xenograft animal model by intra-bone marrow and skin grafts with myeloma cells. RESULTS We found that HLA-G protein degradation augmented angiogenesis. Soluble HLA-G directly inhibited vasculature formation in vitro. Mechanistically, HLA-G expression was regulated by hypoxia-inducible factor-1α (HIF-1α) in MM cells under hypoxia. We thus developed two mouse models of myeloma xenografts in intra-bone marrow (BM) and underneath the skin, and found a strong correlation between HLA-G and HIF-1α expressions in hypoxic BM, but not in oxygenated tissues. Yet when stimulated with IL-6, both HLA-G and HIF-1α could be targeted to ubiquitin-mediated degradation via PARKIN. CONCLUSION These results highlight the importance of sHLA-G in angiogenesis at different phases of multiple myeloma. The experimental evidence that sHLA-G as an angiogenesis suppressor in MM may be useful for future development of novel therapies to prevent relapse.
Collapse
Affiliation(s)
- Chi Wang
- Department of Laboratory Medicine, MacKay Memorial Hospital, New Taipei, 25160, Taiwan
| | - Nai-Wen Su
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Nursing, and Management, MacKay Junior College of Medicine, New Taipei, 25245, Taiwan
| | - Kate Hsu
- Nursing, and Management, MacKay Junior College of Medicine, New Taipei, 25245, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, 25245, Taiwan
- Department of Medical Research, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Chen-Wei Kao
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Ming-Chih Chang
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Yi-Fang Chang
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
| | - Ken-Hong Lim
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Yi-Hao Chiang
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Yu-Cheng Chang
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, 25245, Taiwan
| | - Meng-Ta Sung
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan
| | - Hsueh-Hsia Wu
- Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, 110, Taiwan
| | - Caleb G Chen
- Department of Hematology, MacKay Memorial Hospital, Taipei, 10449, Taiwan.
- Nursing, and Management, MacKay Junior College of Medicine, New Taipei, 25245, Taiwan.
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, New Taipei City, 25160, Taiwan.
- Institute of Molecular Medicine, National Tsing-Hua University, Hsin-Chu, Taiwan.
| |
Collapse
|
3
|
Thakur A, Rana M, Mishra A, Kaur C, Pan CH, Nepali K. Recent advances and future directions on small molecule VEGFR inhibitors in oncological conditions. Eur J Med Chem 2024; 272:116472. [PMID: 38728867 DOI: 10.1016/j.ejmech.2024.116472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
"A journey of mixed emotions" is a quote that best describes the progress chart of vascular endothelial growth factor receptor (VEGFR) inhibitors as cancer therapeutics in the last decade. Exhilarated with the Food and Drug Administration (FDA) approvals of numerous VEGFR inhibitors coupled with the annoyance of encountering the complications associated with their use, drug discovery enthusiasts are on their toes with an unswerving determination to enhance the rate of translation of VEGFR inhibitors from preclinical to clinical stage. The recently crafted armory of VEGFR inhibitors is a testament to their growing dominance over other antiangiogenic therapies for cancer treatment. This review perspicuously underscores the earnest attempts of the researchers to extract the antiproliferative potential of VEGFR inhibitors through the design of mechanistically diverse structural assemblages. Moreover, this review encompasses sections on structural/molecular properties and physiological functions of VEGFR, FDA-approved VEGFR inhibitors, and hurdles restricting the activity range/clinical applicability of VEGFR targeting antitumor agents. In addition, tactics to overcome the limitations of VEGFR inhibitors are discussed. A clear-cut viewpoint transmitted through this compilation can provide practical directions to push the cart of VEGFR inhibitors to advanced-stage clinical investigations in diverse malignancies.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Chun-Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
4
|
Yan H, Gao S, Xu A, Zuo L, Zhang J, Zhao Y, Cheng Q, Yin X, Sun C, Hu Y. MALAT1 regulates network of microRNA-15a/16-VEGFA to promote tumorigenesis and angiogenesis in multiple myeloma. Carcinogenesis 2023; 44:760-772. [PMID: 37549238 DOI: 10.1093/carcin/bgad053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/09/2023] Open
Abstract
MALAT1 is one of the most hopeful members implicated in angiogenesis in a variety of non-malignant diseases. In multiple myeloma (MM), MALAT1 is recognized as the most highly expressed long non-coding RNA. However, the functional roles of MALAT1 in angiogenesis and the responsible mechanisms have not yet been explored. Herein, we discovered a novel regulatory network dependent on MALAT1 in relation to MM tumorigenesis and angiogenesis. We observed that MALAT1 was upregulated in MM and significantly associated with poor overall survival. MALAT1 knockdown suppressed MM cell proliferation and promoted apoptosis, while restricting endothelial cells angiogenesis. Moreover, MALAT1 directly targeted microRNA-15a/16, and microRNA-15a/16 suppression partly reverted the effects of MALAT1 deletion on MM cells in vitro as well as tumor growth and angiogenesis in vivo. In addition, further study indicated that MALAT1 functioned as a competing endogenous RNA for microRNA-15a/16 to regulate vascular endothelial growth factor A (VEGFA) expression. Our results suggest that MALAT1 plays an important role in the regulatory axis of microRNA-15a/16-VEGFA to promote tumorigenicity and angiogenesis in MM. Consequently, MALAT1 could serve as a novel promising biomarker and a potential antiangiogenic target against MM.
Collapse
Affiliation(s)
- Han Yan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Su Gao
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liping Zuo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiasi Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuhong Zhao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianwen Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuejiao Yin
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Mielnik M, Szudy-Szczyrek A, Homa-Mlak I, Mlak R, Podgajna-Mielnik M, Gorący A, Małecka-Massalska T, Hus M. The Clinical Relevance of Selected Cytokines in Newly Diagnosed Multiple Myeloma Patients. Biomedicines 2023; 11:3012. [PMID: 38002012 PMCID: PMC10669681 DOI: 10.3390/biomedicines11113012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/29/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematological neoplasm. Cytokines, chemokines, and their receptors, induced by the microenvironment of MM, participate in tumor growth, the attraction of leukocytes, cell homing, and bone destruction. This study aimed to assess the correlation between the pretreatment serum concentrations of interleukin-6 (IL-6), interleukin-8 (IL-8), angiogenic chemokine monocyte chemoattractant protein-1 (MCP-1), and vascular endothelial growth factor (VEGF) and the clinical outcomes and survival of patients newly diagnosed with MM. The study group consisted of 82 individuals. The IL-8 concentration was significantly positively correlated with the age of onset (p = 0.007), the International Staging System (ISS) stage (p = 0.03), the Eastern Cooperative Oncology Group (ECOG) performance status (p < 0.001), the degree of anemia before treatment (p < 0.0001), the degree of kidney disease (p < 0.001), and VEGF (p = 0.0364). Chemotherapy responders had significantly lower concentrations of IL-8 (p < 0.001), IL-6 (p < 0.001), and VEGF (p = 0.04) compared with non-responders. Patients with treatment-induced polyneuropathy had significantly higher levels of IL-8 (p = 0.033). Patients with a high level of IL-6 had a 2-fold higher risk of progression-free survival (PFS) reduction (17 vs. 35 months; HR = 1.89; p = 0.0078), and a more than 2.5-fold higher risk of overall survival (OS) reduction (28 vs. 78 months; HR = 2.62; p < 0.001). High levels of IL-6, IL-8, and VEGF demonstrated significant predictive values for some clinical conditions or outcomes of newly diagnosed MM patients. Patients with an early response to chemotherapy had a significantly lower concentration of these cytokines. A high pretreatment IL-6 concentration was an independent negative prognostic marker for newly diagnosed MM patients.
Collapse
Affiliation(s)
- Michał Mielnik
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| | - Aneta Szudy-Szczyrek
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| | - Iwona Homa-Mlak
- Department of Human Physiology, Medical University of Lublin, 20-080 Lublin, Poland; (I.H.-M.)
| | - Radosław Mlak
- Department of Laboratory Diagnostics, Medical University of Lublin, Doktora Witolda Chodźki 1 Str., 20-093 Lublin, Poland;
| | - Martyna Podgajna-Mielnik
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| | - Aneta Gorący
- Department of Hematology and Bone Marrow Transplantation, Saint Jan of Dukla Oncology Centre of the Lublin Region, Doktora Kazimierza Jaczewskiego 7 Str., 20-090 Lublin, Poland
| | | | - Marek Hus
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
6
|
Urdeitx P, Mousavi SJ, Avril S, Doweidar MH. Computational modeling of multiple myeloma interactions with resident bone marrow cells. Comput Biol Med 2023; 153:106458. [PMID: 36599211 DOI: 10.1016/j.compbiomed.2022.106458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022]
Abstract
The interaction of multiple myeloma with bone marrow resident cells plays a key role in tumor progression and the development of drug resistance. The tumor cell response involves contact-mediated and paracrine interactions. The heterogeneity of myeloma cells and bone marrow cells makes it difficult to reproduce this environment in in-vitro experiments. The use of in-silico established tools can help to understand these complex problems. In this article, we present a computational model based on the finite element method to define the interactions of multiple myeloma cells with resident bone marrow cells. This model includes cell migration, which is controlled by stress-strain equilibrium, and cell processes such as proliferation, differentiation, and apoptosis. A series of computational experiments were performed to validate the proposed model. Cell proliferation by the growth factor IGF-1 is studied for different concentrations ranging from 0-10 ng/mL. Cell motility is studied for different concentrations of VEGF and fibronectin in the range of 0-100 ng/mL. Finally, cells were simulated under a combination of IGF-1 and VEGF stimuli whose concentrations are considered to be dependent on the cancer-associated fibroblasts in the extracellular matrix. Results show a good agreement with previous in-vitro results. Multiple myeloma growth and migration are shown to correlate linearly to the IGF-1 stimuli. These stimuli are coupled with the mechanical environment, which also improves cell growth. Moreover, cell migration depends on the fiber and VEGF concentration in the extracellular matrix. Finally, our computational model shows myeloma cells trigger mesenchymal stem cells to differentiate into cancer-associated fibroblasts, in a dose-dependent manner.
Collapse
Affiliation(s)
- Pau Urdeitx
- School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, 50018, Spain; Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, 50018, Spain
| | - S Jamaleddin Mousavi
- Mines Saint-Étienne, University of Lyon, University of Jean Monnet, INSERM, Saint-Etienne, 42023, France
| | - Stephane Avril
- Mines Saint-Étienne, University of Lyon, University of Jean Monnet, INSERM, Saint-Etienne, 42023, France; Institute for Mechanics of Materials and Structures, TU Wien-Vienna University of Technology, Vienna, 1040, Austria
| | - Mohamed H Doweidar
- School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, 50018, Spain; Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, 50018, Spain.
| |
Collapse
|
7
|
Monoclonal Gammopathies and the Bone Marrow Microenvironment: From Bench to Bedside and Then Back Again. Hematol Rep 2023; 15:23-49. [PMID: 36648882 PMCID: PMC9844382 DOI: 10.3390/hematolrep15010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/11/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematologic malignancy characterized by a multistep evolutionary pathway, with an initial phase called monoclonal gammopathy of undetermined significance (MGUS), potentially evolving into the symptomatic disease, often preceded by an intermediate phase called "smoldering" MM (sMM). From a biological point of view, genomic alterations (translocations/deletions/mutations) are already present at the MGUS phase, thus rendering their role in disease evolution questionable. On the other hand, we currently know that changes in the bone marrow microenvironment (TME) could play a key role in MM evolution through a progressive shift towards a pro-inflammatory and immunosuppressive shape, which may drive cancer progression as well as clonal plasma cells migration, proliferation, survival, and drug resistance. Along this line, the major advancement in MM patients' survival has been achieved by the introduction of microenvironment-oriented drugs (including immunomodulatory drugs and monoclonal antibodies). In this review, we summarized the role of the different components of the TME in MM evolution from MGUS as well as potential novel therapeutic targets/opportunities.
Collapse
|
8
|
Aksoy O, Lind J, Sunder-Plaßmann V, Vallet S, Podar K. Bone marrow microenvironment- induced regulation of Bcl-2 family members in multiple myeloma (MM): Therapeutic implications. Cytokine 2023; 161:156062. [PMID: 36332463 DOI: 10.1016/j.cyto.2022.156062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/19/2022] [Accepted: 09/30/2022] [Indexed: 11/23/2022]
Abstract
In Multiple Myeloma (MM) the finely tuned homeostasis of the bone marrow (BM) microenvironment is disrupted. Evasion of programmed cell death (apoptosis) represents a hallmark of cancer. Besides genetic aberrations, the supportive and protective MM BM milieu, which is constituted by cytokines and growth factors, intercellular and cell: extracellular matrix (ECM) interactions and exosomes, in particular, plays a key role in the abundance of pro-survival members of the Bcl-2 family (i.e., Mcl-1, Bcl-2, and Bcl-xL) in tumor cells. Moreover, microenvironmental cues have also an impact on stability- regulating post-translational modifications of anti-apoptotic proteins including de/phosphorylation, polyubiquitination; on their intracellular binding affinities, and localization. Advances of our molecular knowledge on the escape of cancer cells from apoptosis have informed the development of a new class of small molecules that mimic the action of BH3-only proteins. Indeed, approaches to directly target anti-apoptotic Bcl-2 family members are among today's most promising therapeutic strategies and BH3-mimetics (i.e., venetoclax) are currently revolutionizing not only the treatment of CLL and AML, but also hold great therapeutic promise in MM. Furthermore, approaches that activate apoptotic pathways indirectly via modification of the tumor microenvironment have already entered clinical practice. The present review article will summarize our up-to-date knowledge on molecular mechanisms by which the MM BM microenvironment, cytokines, and growth factors in particular, mediates tumor cell evasion from apoptosis. Moreover, it will discuss some of the most promising science- derived therapeutic strategies to overcome Bcl-2- mediated tumor cell survival in order to further improve MM patient outcome.
Collapse
Affiliation(s)
- Osman Aksoy
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Judith Lind
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Vincent Sunder-Plaßmann
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Sonia Vallet
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria; Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
| | - Klaus Podar
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria; Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria.
| |
Collapse
|
9
|
Li S, Co CM, Izuagbe S, Hong Y, Liao J, Borrelli J, Tang L. Biomolecules-releasing click chemistry-based bioadhesives for repairing acetabular labrum tears. J Orthop Res 2022; 40:2646-2655. [PMID: 35112388 DOI: 10.1002/jor.25290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 12/07/2021] [Accepted: 01/30/2022] [Indexed: 02/04/2023]
Abstract
Currently, there are no effective clinical or experimental treatments to fully restore the function of the torn acetabular labrum. To fill the gap, here, we report the finding of progenitor cells in labral tissue, which can be recruited and stimulated to repair torn acetabular labral tissue. This study aimed to develop a biomolecule releasing bioadhesive which can speed up labral tissue healing by eliciting autologous labral progenitor cellular responses. A click chemistry-based bioadhesive, capable of releasing biomolecules, was synthesized to exert ~3× adhesion strength compared with fibrin glue. Via the release of platelet-derived growth factor (PDGF), the adhesive was shown to actively recruit and stimulate the proliferation of labral progenitor cells to the tear sites and within the adhesive. Finally, the ability of this biomolecules-releasing adhesive designed to promote labral tissue regeneration was evaluated using discarded human acetabular labrum tissue compared with surgical suture ex vivo. Histological analysis shows that PDGF-releasing bioadhesive yielded significantly more labrum cell responses and extracellular matrix protein (proteoglycan and collagen) production at the tear tissue site than surgical suture controls. The results confirm that the new PDGF-releasing bioadhesive can activate the responses of autologous labral progenitor cells to significantly improve labral tissue regeneration. Clinical significance: These PDGF-releasing bioadhesives may serve as a new and effective tool for repairing and regenerating acetabular labrum tears.
Collapse
Affiliation(s)
- Shuxin Li
- Department of Research & Development, Progenitec Inc., Arlington, Texas, USA
| | - Cynthia M Co
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Samira Izuagbe
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Joseph Borrelli
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Liping Tang
- Department of Research & Development, Progenitec Inc., Arlington, Texas, USA.,Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
10
|
Melaccio A, Reale A, Saltarella I, Desantis V, Lamanuzzi A, Cicco S, Frassanito MA, Vacca A, Ria R. Pathways of Angiogenic and Inflammatory Cytokines in Multiple Myeloma: Role in Plasma Cell Clonal Expansion and Drug Resistance. J Clin Med 2022; 11:jcm11216491. [PMID: 36362718 PMCID: PMC9658666 DOI: 10.3390/jcm11216491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, and despite the introduction of innovative therapies, remains an incurable disease. Identifying early and minimally or non-invasive biomarkers for predicting clinical outcomes and therapeutic responses is an active field of investigation. Malignant plasma cells (PCs) reside in the bone marrow (BM) microenvironment (BMME) which comprises cells (e.g., tumour, immune, stromal cells), components of the extracellular matrix (ECM) and vesicular and non-vesicular (soluble) molecules, all factors that support PCs’ survival and proliferation. The interaction between PCs and BM stromal cells (BMSCs), a hallmark of MM progression, is based not only on intercellular interactions but also on autocrine and paracrine circuits mediated by soluble or vesicular components. In fact, PCs and BMSCs secrete various cytokines, including angiogenic cytokines, essential for the formation of specialized niches called “osteoblastic and vascular niches”, thus supporting neovascularization and bone disease, vital processes that modulate the pathophysiological PCs–BMME interactions, and ultimately promoting disease progression. Here, we aim to discuss the roles of cytokines and growth factors in pathogenetic pathways in MM and as prognostic and predictive biomarkers. We also discuss the potential of targeted drugs that simultaneously block PCs’ proliferation and survival, PCs–BMSCs interactions and BMSCs activity, which may represent the future goal of MM therapy.
Collapse
Affiliation(s)
- Assunta Melaccio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| | - Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne 3004, Australia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- General Pathology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| |
Collapse
|
11
|
Krhovska P, Pika T, Proskova J, Balcarkova J, Zapletalova J, Bacovsky J, Minarik J. Bone metabolism parameters and their relation to cytogenetics in multiple myeloma. Eur J Haematol Suppl 2022; 109:75-82. [PMID: 35306691 DOI: 10.1111/ejh.13771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Our aim was to correlate serum levels of selected markers of bone metabolism and bone marrow microenvironment to cytogenetic changes in patients with multiple myeloma (MM). METHODS We assed cytogenetic changes in 308 patients and correlated them with the following levels of bone marrow metabolism: thymidine kinase (TK), β2-microglobulin (b-2-m), Dickkopf-1 protein (DKK-1), C-terminal telopeptide collagen-I (ICTP), N-terminal propeptide of type I procollagen (PINP), receptor for interleukin 6 (rIL-6), vascular cell adhesive molecule-1 (VCAM), soluble intercellular adhesion molecule-1, osteoprotegerin (OPG), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), syndecan-1 (SYN-1) and Fas antigen. RESULT Individuals with delRB1 had lower levels of OPG (M = 7.39 vs. 5.46 pmol/L, p = .025) and VEGF (M = 304 vs. 196 pg/ml; p = .036). t(14;16) was associated with higher β2m levels (M = 7.59 vs. 4.13 mg/L; p = .022) and lower DKK-1 levels (M = 4465 ng/L vs. 12,593). The presence of 1q21 gain was associated with higher levels of TK (M = 100.0 vs. 11.0 IU/L, p = .026) and lower levels of PINP (M = 49.3 vs. 67.4 mg/L, p = .030). CONCLUSIONS Our analysis has shown, some cytogenetic changes, especially delRB1, t(14;16) and 1q21gain, which affect the components of the cytokine network in multiple myeloma.
Collapse
Affiliation(s)
- Petra Krhovska
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Tomas Pika
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jitka Proskova
- Department of Clinical Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jana Balcarkova
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jana Zapletalova
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jaroslav Bacovsky
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Jiri Minarik
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| |
Collapse
|
12
|
Mi J, Xu J, Zhou J, Zhao W, Chen Z, Melenhorst JJ, Chen S. CAR T-cell immunotherapy: a powerful weapon for fighting hematological B-cell malignancies. Front Med 2021; 15:783-804. [DOI: 10.1007/s11684-021-0904-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
|
13
|
Allegra A, Di Gioacchino M, Tonacci A, Petrarca C, Musolino C, Gangemi S. Multiple Myeloma Cell-Derived Exosomes: Implications on Tumorigenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cells 2021; 10:2865. [PMID: 34831088 PMCID: PMC8616233 DOI: 10.3390/cells10112865] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a hematological disease that is still not curable. The bone marrow milieu, with cellular and non-cellular elements, participate in the creation of a pro-tumoral environment enhancing growth and survival of MM plasma cells. Exosomes are vesicles oscillating in dimension between 50 nm and 100 nm in size that can be released by various cells and contribute to the pathogenesis and progression of MM. Exosomes enclose proteins, cytokines, lipids, microRNAs, long noncoding RNAs, and circular RNAs able to regulate interactions between MM plasma cells and adjacent cells. Through exosomes, mesenchymal stem cells confer chemoresistance to MM cells, while myeloma cells promote angiogenesis, influence immune response, cause bone lesions, and have an impact on the outcome of MM patients. In this review, we analyze the role played by exosomes in the progression of monoclonal gammopathies and the effects on the proliferation of neoplastic plasma cells, and discuss the possible employment of exosomes as potential targets for the treatment of MM patients.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
| | - Alessandro Tonacci
- National Research Council of Italy (IFC-CNR), Clinical Physiology Institute, 56124 Pisa, Italy;
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- National Research Council of Italy (IFC-CNR), Clinical Physiology Institute, 56124 Pisa, Italy;
- Department of Medicine and Science of Ageing, G. D’Annunzio University, 66100 Chieti, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
14
|
Li T, Chen J, Zeng Z. Pathophysiological role of calcium channels and transporters in the multiple myeloma. Cell Commun Signal 2021; 19:99. [PMID: 34579758 PMCID: PMC8477534 DOI: 10.1186/s12964-021-00781-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/28/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a common malignant tumor of plasma cells. Despite several treatment approaches in the past two decades, MM remains an aggressive and incurable disease in dire need of new treatment strategies. Approximately 70–80% of patients with MM have myeloma bone disease (MBD), often accompanied by pathological fractures and hypercalcemia, which seriously affect the prognosis of the patients. Calcium channels and transporters can mediate Ca2+ balance inside and outside of the membrane, indicating that they may be closely related to the prognosis of MM. Therefore, this review focuses on the roles of some critical calcium channels and transporters in MM prognosis, which located in the plasma membrane, endoplasmic reticulum and mitochondria. The goal of this review is to facilitate the identification of new targets for the treatment and prognosis of MM.![]() Video Abstract
Collapse
Affiliation(s)
- Tingting Li
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, Fujian, 350005, People's Republic of China.,Fujian Key Laboratory of Laboratory Medicine, Fuzhou, People's Republic of China
| | - Junmin Chen
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, Fujian, 350005, People's Republic of China. .,Fujian Key Laboratory of Laboratory Medicine, Fuzhou, People's Republic of China.
| | - Zhiyong Zeng
- Department of Hematology, The First Affiliated Hospital of Fujian Medical University, 20 Chazhong Road, Fuzhou, Fujian, 350005, People's Republic of China. .,Fujian Key Laboratory of Laboratory Medicine, Fuzhou, People's Republic of China.
| |
Collapse
|
15
|
Sarwar A, Hashim L, Faisal MS, Haider MZ, Ahmed Z, Ahmed TF, Shahzad M, Ansar I, Ali S, Aslam MM, Anwer F. Advances in viral oncolytics for treatment of multiple myeloma - a focused review. Expert Rev Hematol 2021; 14:1071-1083. [PMID: 34428997 DOI: 10.1080/17474086.2021.1972802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Oncolytic viruses are genetically engineered viruses that target myeloma-affected cells by detecting specific cell surface receptors (CD46, CD138), causing cell death by activating the signaling pathway to induce apoptosis or by immune-mediated cellular destruction. AREAS COVERED This article summarizes oncolytic virotherapy advancements such as the therapeutic use of viruses by targeting cell surface proteins of myeloma cells as well as the carriers to deliver viruses to the target tissues safely. The major classes of viruses that have been studied for this include measles, myxoma, adenovirus, reovirus, vaccinia, vesicular-stomatitis virus, coxsackie, and others. The measles virus acts as oncolytic viral therapy by binding to the CD46 receptors on the myeloma cells to utilize its surface H protein. These H-protein and CD46 interactions lead to cellular syncytia formation resulting in cellular apoptosis. Vesicular-stomatitis virus acts by downregulation of anti-apoptotic factors (Mcl-2, BCL-2). Based upon the published literature searches till December 2020, we have summarized the data supporting the advances in viral oncolytic for the treatment of MM. EXPERT OPINION Oncolytic virotherapy is an experimental approach in multiple myeloma (MM); many issues need to be addressed for safe viral delivery to the target tissue.
Collapse
Affiliation(s)
- Ayesha Sarwar
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Muhammad Salman Faisal
- Department of Internal Medicine, Division of Hematology, The Ohio State University Columbus Oh, USA
| | | | - Zahoor Ahmed
- Department of Internal Medicine, King Edward Medical University, Lahore, Pakistan
| | - Tehniat Faraz Ahmed
- Department of Biochemistry, Dow University of Health Sciences, Karachi, Pakistan
| | - Moazzam Shahzad
- Department of Internal Medicine, St Mary's Medical Center, Huntington, WV, USA
| | - Iqraa Ansar
- Department of Internal medicine, Riverside Methodist hospital, Columbus OH
| | - Sundas Ali
- Department of Internal medicine, Rawalpindi Medical University, Rawalpindi, Pakistan
| | | | - Faiz Anwer
- Department of Hematology and Oncology, Taussig Cancer Center, Cleveland Clinic, Ohio, USA
| |
Collapse
|
16
|
Zahan T, Das PK, Akter SF, Habib R, Rahman MH, Karim MR, Islam F. Therapy Resistance in Cancers: Phenotypic, Metabolic, Epigenetic and Tumour Microenvironmental Perspectives. Anticancer Agents Med Chem 2021; 20:2190-2206. [PMID: 32748758 DOI: 10.2174/1871520620999200730161829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/02/2020] [Accepted: 05/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chemoresistance is a vital problem in cancer therapy where cancer cells develop mechanisms to encounter the effect of chemotherapeutics, resulting in cancer recurrence. In addition, chemotherapy- resistant leads to the formation of a more aggressive form of cancer cells, which, in turn, contributes to the poor survival of patients with cancer. OBJECTIVE In this review, we aimed to provide an overview of how the therapy resistance property evolves in cancer cells, contributing factors and their role in cancer chemoresistance, and exemplified the problems of some available therapies. METHODS The published literature on various electronic databases including, Pubmed, Scopus, Google scholar containing keywords cancer therapy resistance, phenotypic, metabolic and epigenetic factors, were vigorously searched, retrieved and analyzed. RESULTS Cancer cells have developed a range of cellular processes, including uncontrolled activation of Epithelial- Mesenchymal Transition (EMT), metabolic reprogramming and epigenetic alterations. These cellular processes play significant roles in the generation of therapy resistance. Furthermore, the microenvironment where cancer cells evolve effectively contributes to the process of chemoresistance. In tumour microenvironment immune cells, Mesenchymal Stem Cells (MSCs), endothelial cells and cancer-associated fibroblasts (CAFs) contribute to the maintenance of therapy-resistant phenotype via the secretion of factors that promote resistance to chemotherapy. CONCLUSION To conclude, as these factors hinder successful cancer therapies, therapeutic resistance property of cancer cells is a subject of intense research, which in turn could open a new horizon to aim for developing efficient therapies.
Collapse
Affiliation(s)
- Tasnim Zahan
- Molecular Mechanisms of Disease, Radboud University, Nijmegen, The Netherlands
| | - Plabon K Das
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Syeda F Akter
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Rowshanul Habib
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md Habibur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md Rezaul Karim
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi-6205, Bangladesh,Institute for Glycomics, Griffith University, Queensland, Australia
| |
Collapse
|
17
|
Teramachi J, Tenshin H, Hiasa M, Oda A, Bat-Erdene A, Harada T, Nakamura S, Ashtar M, Shimizu S, Iwasa M, Sogabe K, Oura M, Fujii S, Kagawa K, Miki H, Endo I, Haneji T, Matsumoto T, Abe M. TAK1 is a pivotal therapeutic target for tumor progression and bone destruction in myeloma. Haematologica 2021; 106:1401-1413. [PMID: 32273474 PMCID: PMC8094086 DOI: 10.3324/haematol.2019.234476] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Indexed: 12/31/2022] Open
Abstract
Along with tumor progression, the bone marrow microenvironment is skewed in multiple myeloma (MM), which underlies the unique pathophysiology of MM and confers aggressiveness and drug resistance in MM cells. TGF-b-activated kinase-1 (TAK1) mediates a wide range of intracellular signaling pathways. We demonstrate here that TAK1 is constitutively overexpressed and phosphorylated in MM cells, and that TAK1 inhibition suppresses the activation of NF-κB, p38MAPK, ERK and STAT3 in order to decrease the expression of critical mediators for MM growth and survival, including PIM2, MYC, Mcl- 1, IRF4, and Sp1, along with a substantial reduction in the angiogenic factor VEGF in MM cells. Intriguingly, TAK1 phosphorylation was also induced along with upregulation of vascular cell adhesion molecule-1 (VCAM-1) in bone marrow stromal cells (BMSC) in cocultures with MM cells, which facilitated MM cell-BMSC adhesion while inducing IL-6 production and receptor activator of nuclear factor κ-B ligand (RANKL) expression by BMSC. TAK1 inhibition effectively impaired MM cell adhesion to BMSC to disrupt the support of MM cell growth and survival by BMSC. Furthermore, TAK1 inhibition suppressed osteoclastogenesis enhanced by RANKL in cocultures of bone marrow cells with MM cells, and restored osteoblastic differentiation suppressed by MM cells or inhibitory factors for osteoblastogenesis overproduced in MM. Finally, treatment with the TAK1 inhibitor LLZ1640-2 markedly suppressed MM tumor growth and prevented bone destruction and loss in mouse MM models. Therefore, TAK1 inhibition may be a promising therapeutic option targeting not only MM cells but also the skewed bone marrow microenvironment in MM.
Collapse
Affiliation(s)
- Jumpei Teramachi
- Dept. of Histology-Oral Histology and Dept. of Hematology, Tokushima University,Tokushima, Japan
| | - Hirofumi Tenshin
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - Masahiro Hiasa
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - Asuka Oda
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Ariunzaya Bat-Erdene
- Dept of Hematology, Tokushima University and University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Takeshi Harada
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Shingen Nakamura
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Mohannad Ashtar
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - So Shimizu
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - Masami Iwasa
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Kimiko Sogabe
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Masahiro Oura
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Shiro Fujii
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Kumiko Kagawa
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Hirokazu Miki
- Division of Transfusion Medicine and Cell Therapy, Tokushima University Hospital, Tokushima, Japan
| | - Itsuro Endo
- Department of Chronomedicine, Tokushima University, Tokushima, Japan
| | - Tatsuji Haneji
- Department of Histology and Oral Histology, Tokushima University, Tokushima, Japan
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masahiro Abe
- Department of Hematology, Tokushima University, Tokushima, Japan
| |
Collapse
|
18
|
Signaling Pathway Mediating Myeloma Cell Growth and Survival. Cancers (Basel) 2021; 13:cancers13020216. [PMID: 33435632 PMCID: PMC7827005 DOI: 10.3390/cancers13020216] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The bone marrow (BM) microenvironment plays a crucial role in pathogenesis of multiple myeloma (MM), and delineation of the intracellular signaling pathways activated in the BM microenvironment in MM cells is essential to develop novel therapeutic strategies to improve patient outcome. Abstract The multiple myeloma (MM) bone marrow (BM) microenvironment consists of different types of accessory cells. Both soluble factors (i.e., cytokines) secreted from these cells and adhesion of MM cells to these cells play crucial roles in activation of intracellular signaling pathways mediating MM cell growth, survival, migration, and drug resistance. Importantly, there is crosstalk between the signaling pathways, increasing the complexity of signal transduction networks in MM cells in the BM microenvironment, highlighting the requirement for combination treatment strategies to blocking multiple signaling pathways.
Collapse
|
19
|
Ntellas P, Mavroeidis L, Gkoura S, Gazouli I, Amylidi AL, Papadaki A, Zarkavelis G, Mauri D, Karpathiou G, Kolettas E, Batistatou A, Pentheroudakis G. Old Player-New Tricks: Non Angiogenic Effects of the VEGF/VEGFR Pathway in Cancer. Cancers (Basel) 2020; 12:E3145. [PMID: 33121034 PMCID: PMC7692709 DOI: 10.3390/cancers12113145] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis has long been considered to facilitate and sustain cancer growth, making the introduction of anti-angiogenic agents that disrupt the vascular endothelial growth factor/receptor (VEGF/VEGFR) pathway an important milestone at the beginning of the 21st century. Originally research on VEGF signaling focused on its survival and mitogenic effects towards endothelial cells, with moderate so far success of anti-angiogenic therapy. However, VEGF can have multiple effects on additional cell types including immune and tumor cells, by directly influencing and promoting tumor cell survival, proliferation and invasion and contributing to an immunosuppressive microenvironment. In this review, we summarize the effects of the VEGF/VEGFR pathway on non-endothelial cells and the resulting implications of anti-angiogenic agents that include direct inhibition of tumor cell growth and immunostimulatory functions. Finally, we present how previously unappreciated studies on VEGF biology, that have demonstrated immunomodulatory properties and tumor regression by disrupting the VEGF/VEGFR pathway, now provide the scientific basis for new combinational treatments of immunotherapy with anti-angiogenic agents.
Collapse
Affiliation(s)
- Panagiotis Ntellas
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Leonidas Mavroeidis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Stefania Gkoura
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Ioanna Gazouli
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Anna-Lea Amylidi
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Alexandra Papadaki
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Davide Mauri
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Georgia Karpathiou
- Department of Pathology, University Hospital of St-Etienne, 42055 Saint Etienne, France;
| | - Evangelos Kolettas
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Biomedical Research Division, Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology, 45115 Ioannina, Greece
| | - Anna Batistatou
- Department of Pathology, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - George Pentheroudakis
- Department of Medical Oncology, University Hospital of Ioannina, 45500 Ioannina, Greece; (P.N.); (L.M.); (S.G.); (I.G.); (A.-L.A.); (A.P.); (G.Z.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| |
Collapse
|
20
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
21
|
Liu L, Ye Q, Liu L, Bihl JC, Chen Y, Liu J, Cheng Q. C6-ceramide treatment inhibits the proangiogenic activity of multiple myeloma exosomes via the miR-29b/Akt pathway. J Transl Med 2020; 18:298. [PMID: 32746845 PMCID: PMC7398185 DOI: 10.1186/s12967-020-02468-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Background The increased bone marrow angiogenesis is involved in the progression of multiple myeloma (MM) with the underlying mechanism poorly understood. Cancer-released exosomes could play an important role in the pathological angiogenesis through exosomal microRNAs (miRs) delivery. It is reported that miR-29b played an important role in regulating the tumor angiogenesis. Methods In this study, we explored the role of C6-ceramide (C6-cer, a Ceramide pathway activator) in the angiogenic effect of MM exosomes and its potential mechanism. MM cells (OPM2 and RPMI-8226) treated with C6-cer were studied for its effects on the endothelial cell (EC) functions. Results Our results showed that exosomes released from MM cells treated by C6-cer (ExoC6-cer) significantly inhibited the proliferation, migration and tube formation of ECs. For mechanism studies, we found that the level of miR-29b was increased in ECs treated by ExoC6-cer, while mRNA and protein expressions of Akt3, PI3K and VEGFA were decreased in ECs, indicating the involvement of Akt pathway. Furthermore, downregulation of miR-29b by inhibitor administration could prevent the ExoC6-cer-induced cell proliferation, migration and angiogenesis of ECs, accompanied with the increased expressions of Akt3, PI3K and VEGFA. Conclusions Collectively, our data suggest that ExoC6-cer-mediated miR-29b expression participates in the progression of MM through suppressing the proliferation, migration and angiogenesis of ECs by targeting Akt signal pathway.
Collapse
Affiliation(s)
- Liping Liu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Qinmao Ye
- Department of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Langni Liu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, 45435, USA
| | - Ji Chen Bihl
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, 45435, USA
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, 45435, USA
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Qian Cheng
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, 45435, USA. .,Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
22
|
Palta A, Kaur M, Tahlan A, Dimri K. Evaluation of Angiogenesis in Multiple Myeloma by VEGF Immunoexpression and Microvessel Density. J Lab Physicians 2020; 12:38-43. [PMID: 32792792 PMCID: PMC7419170 DOI: 10.1055/s-0040-1714933] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background
Multiple myeloma (MM) is a plasma cell disorder characterized by monoclonal proliferation of plasma cells in bone marrow. Plasmablastic MM is a morphologic subset of MM, containing ≥2% plasmablasts of all plasma cells.
Methods
The study included 30 consecutively diagnosed patients of MM (6 plasmablastic, 24 nonplasmablastic) over a span of 2 years. Angiogenesis in MM was assessed by analysis of vascular endothelial growth factor (VEGF) immunoexpression by plasma cells and microvessel density (MVD) using anti-CD34 antibody. CD34 and VEGF immunohistochemical staining was performed in all the 30 cases. Angiogenesis was studied in relation to plasmablastic morphology and clinical profile to determine if any correlation exists between these.
Results
The mean VEGF expression of 80.83 ± 7.36 in plasmablastic myeloma cases was significantly higher compared with a mean VEGF of 53.54 ±17.09 in nonplasmablastic cases. Most of the cases (66.6%) of plasmablastic myeloma exhibited strong (3+) VEGF expression. The difference in mean VEGF expression between plasmablastic and nonplasmablastic cases was found to be statistically significant (
p
= 0.001). The mean MVD in plasmablastic cases was 44.8 ± 3.69, while in the nonplasmablastic category, the mean MVD was 23.7 ± 5.14, difference being statistically significant (
p
< 0.05). Also, a positive correlation was found between VEGF expression and MVD.
Conclusion
A moderate/strong VEGF intensity and higher MVD were found in cases of plasmablastic MM, suggesting that a more aggressive histological disease may be associated with increased production of VEGF. This finding might be helpful to identify a subset of patients with adverse prognosis and to provide antiangiogenic therapy to improve their survival. However, studies comprising larger number of patients are required to bring out a statistical significance to further substantiate these findings.
Collapse
Affiliation(s)
- Anshu Palta
- Department of Pathology, Government Medical College and Hospital, Chandigarh, India
| | - Manveen Kaur
- Department of Pathology, Government Medical College and Hospital, Chandigarh, India
| | - Anita Tahlan
- Department of Pathology, Government Medical College and Hospital, Chandigarh, India
| | - Kislay Dimri
- Department of Radiation Oncology, Government Medical College and Hospital, Chandigarh, India
| |
Collapse
|
23
|
Cytokine-Mediated Dysregulation of Signaling Pathways in the Pathogenesis of Multiple Myeloma. Int J Mol Sci 2020; 21:ijms21145002. [PMID: 32679860 PMCID: PMC7403981 DOI: 10.3390/ijms21145002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic disorder of B lymphocytes characterized by the accumulation of malignant plasma cells (PCs) in the bone marrow. The altered plasma cells overproduce abnormal monoclonal immunoglobulins and also stimulate osteoclasts. The host’s immune system and microenvironment are of paramount importance in the growth of PCs and, thus, in the pathogenesis of the disease. The interaction of MM cells with the bone marrow (BM) microenvironment through soluble factors and cell adhesion molecules causes pathogenesis of the disease through activation of multiple signaling pathways, including NF-κβ, PI3K/AKT and JAK/STAT. These activated pathways play a critical role in the inhibition of apoptosis, sustained proliferation, survival and migration of MM cells. Besides, these pathways also participate in developing resistance against the chemotherapeutic drugs in MM. The imbalance between inflammatory and anti-inflammatory cytokines in MM leads to an increased level of pro-inflammatory cytokines, which in turn play a significant role in dysregulation of signaling pathways and proliferation of MM cells; however, the association appears to be inadequate and needs more research. In this review, we are highlighting the recent findings on the roles of various cytokines and growth factors in the pathogenesis of MM and the potential therapeutic utility of aberrantly activated signaling pathways to manage the MM disease.
Collapse
|
24
|
Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e752-e768. [PMID: 32651110 DOI: 10.1016/j.clml.2020.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of the plasma cells within the bone marrow (BM). Studies have shown that the cellular and noncellular components of the BM milieu, such as cytokines and exosomes, play an integral role in MM pathogenesis and progression by mediating drug resistance and inducing MM proliferation. Moreover, the BM microenvironment of patients with MM facilitates cancer tolerance and immune evasion through the expansion of regulatory immune cells, inhibition of antitumor effector cells, and disruption of the antigen presentation machinery. These are of special relevance, especially in the current era of cancer immunotherapy. An improved understanding of the supportive role of the MM BM microenvironment will allow for the development of future therapies targeting MM in the context of the BM milieu to elicit deeper and more durable responses. In the present review, we have discussed our current understanding of the role of the BM microenvironment in MM progression and resistance to therapy and discuss novel potential approaches to alter its pro-MM function.
Collapse
|
25
|
Ria R, Melaccio A, Racanelli V, Vacca A. Anti-VEGF Drugs in the Treatment of Multiple Myeloma Patients. J Clin Med 2020; 9:E1765. [PMID: 32517267 PMCID: PMC7355441 DOI: 10.3390/jcm9061765] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
The interaction between the bone marrow microenvironment and plasma cells plays an essential role in multiple myeloma progression and drug resistance. The vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) pathway in vascular endothelial cells activates and promotes angiogenesis. Moreover, VEGF activates and promotes vasculogenesis and vasculogenic mimicry when it interacts with VEGF receptors expressed in precursor cells and inflammatory cells, respectively. In myeloma bone marrow, VEGF and VEGF receptor expression are upregulated and hyperactive in the stromal and tumor cells. It has been demonstrated that several antiangiogenic agents can effectively target VEGF-related pathways in the preclinical phase. However, they are not successful in treating multiple myeloma, probably due to the vicarious action of other cytokines and signaling pathways. Thus, the simultaneous blocking of multiple cytokine pathways, including the VEGF/VEGFR pathway, may represent a valid strategy to treat multiple myeloma. This review aims to summarize recent advances in understanding the role of the VEGF/VEGFR pathway in multiple myeloma, and mainly focuses on the transcription pathway and on strategies that target this pathway.
Collapse
Affiliation(s)
- Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (V.R.); (A.V.)
| | | | | | | |
Collapse
|
26
|
Bai W, Wang H, Bai H. Identification of Candidate Genes and Therapeutic Agents for Light Chain Amyloidosis Based on Bioinformatics Approach. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 12:387-396. [PMID: 32099441 PMCID: PMC6997413 DOI: 10.2147/pgpm.s228574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022]
Abstract
Objective Systemic amyloid light chain (AL) amyloidosis is a rare plasma cell disease. However, the regulatory mechanisms of AL amyloidosis have not been thoroughly uncovered, identification of candidate genes and therapeutic agents for this disease is crucial to provide novel insights into exploring the regulatory mechanisms underlying AL amyloidosis. Methods The gene expression profile of GSE73040, including 9 specimens from AL amyloidosis patients and 5 specimens from normal control, was downloaded from GEO datasets. Differentially expressed genes (DEGs) were sorted with regard to AL amyloidosis versus normal control group using Limma package. The gene enrichment analyses including GO and KEGG pathway were performed using DAVID website subsequently. Furthermore, the protein–protein interaction (PPI) network for DEGs was constructed by Cytoscape software and STRING database. DEGs were mapped to the connectivity map datasets to identify potential molecular agents of AL amyloidosis. Results A total of 1464 DEGs (727 up-regulated, 737 down-regulated) were identified in AL amyloidosis samples versus control samples, these dysregulated genes were associated with the dysfunction of ribosome biogenesis and immune response. PPI network and module analysis uncovered that several crucial genes were defined as candidate genes, including ITGAM, ITGB2, ITGAX, IMP3 and FBL. More importantly, we identified the small molecular agents (AT-9283, Ritonavir and PKC beta-inhibitor) as the potential drugs for AL amyloidosis. Conclusion Using bioinformatics approach, we have identified candidate genes and pathways in AL amyloidosis, which can extend our understanding of the cause and molecular mechanisms, and these crucial genes and pathways could act as biomarkers and therapeutic targets for AL amyloidosis.
Collapse
Affiliation(s)
- Wenxiang Bai
- Comprehensive Cancer Center, Xiangshui People's Hospital, Xiangshui 224600, People's Republic of China.,Department of Respiratory Medicine, Xiangshui People's Hospital, Xiangshui, 224600, People's Republic of China
| | - Honghua Wang
- Comprehensive Cancer Center, Xiangshui People's Hospital, Xiangshui 224600, People's Republic of China
| | - Hua Bai
- Comprehensive Cancer Center, Xiangshui People's Hospital, Xiangshui 224600, People's Republic of China.,Department of Hematology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People's Republic of China
| |
Collapse
|
27
|
Pinto V, Bergantim R, Caires HR, Seca H, Guimarães JE, Vasconcelos MH. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers (Basel) 2020; 12:E407. [PMID: 32050631 PMCID: PMC7072128 DOI: 10.3390/cancers12020407] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is the second most common blood cancer. Treatments for MM include corticosteroids, alkylating agents, anthracyclines, proteasome inhibitors, immunomodulatory drugs, histone deacetylase inhibitors and monoclonal antibodies. Survival outcomes have improved substantially due to the introduction of many of these drugs allied with their rational use. Nonetheless, MM patients successively relapse after one or more treatment regimens or become refractory, mostly due to drug resistance. This review focuses on the main drugs used in MM treatment and on causes of drug resistance, including cytogenetic, genetic and epigenetic alterations, abnormal drug transport and metabolism, dysregulation of apoptosis, autophagy activation and other intracellular signaling pathways, the presence of cancer stem cells, and the tumor microenvironment. Furthermore, we highlight the areas that need to be further clarified in an attempt to identify novel therapeutic targets to counteract drug resistance in MM patients.
Collapse
Affiliation(s)
- Vanessa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- FCTUC–Faculty of Science and Technology of the University of Coimbra, 3030-790 Coimbra, Portugal
| | - Rui Bergantim
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hugo R. Caires
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Hugo Seca
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - M. Helena Vasconcelos
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP-Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
28
|
Targeting the ubiquitin-proteasome pathway to overcome anti-cancer drug resistance. Drug Resist Updat 2020; 48:100663. [DOI: 10.1016/j.drup.2019.100663] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/01/2019] [Accepted: 11/03/2019] [Indexed: 02/07/2023]
|
29
|
Sławińska-Brych A, Zdzisińska B, Czerwonka A, Mizerska-Kowalska M, Dmoszyńska-Graniczka M, Stepulak A, Gagoś M. Xanthohumol exhibits anti-myeloma activity in vitro through inhibition of cell proliferation, induction of apoptosis via the ERK and JNK-dependent mechanism, and suppression of sIL-6R and VEGF production. Biochim Biophys Acta Gen Subj 2019; 1863:129408. [PMID: 31386885 DOI: 10.1016/j.bbagen.2019.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 07/13/2019] [Accepted: 08/01/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Xanthohumol (XN, a hop-derived prenylflavonoid) was found to exert anticancer effects on various cancer types. However, the mechanisms by which XN affects the survival of multiple myeloma cells (MM) are little known. Therefore, our study was undertaken to address this issue. METHODS Anti-proliferative activity of XN towards two phenotypically distinct MM cell lines U266 and RPMI8226 was evaluated with the MTT and BrdU assays. Cytotoxicity was determined with the LDH method, whereas apoptosis was assessed by flow cytometry and fluorescence staining. The expression of cell cycle- and apoptosis-related proteins and the activation status of signaling pathways were estimated by immunoblotting and ELISA assays. RESULTS XN reduced the viability of RPMI8226 cells more potently than in U266 cells. It blocked cell cycle progression through downregulation of cyclin D1 and increased p21 expression. The marked apoptosis induction in the XN-treated RPMI8226 cells was related to initiation of mitochondrial and extrinsic pathways, as indicated by the altered p53, Bax, and Bcl-2 protein expression, cleavage of procaspase 8 and 9, and elevated caspase-3 activity. The apoptotic process was probably mediated via ROS overproduction and MAPK (ERK and JNK) activation as N-acetylcysteine, or specific inhibitors of these kinases prevented the XN-induced caspase-3 activity and, hence, apoptosis. Moreover, XN decreased sIL-6R and VEGF production in the studied cells. CONCLUSIONS ERK and JNK signaling pathways are involved in XN-induced cytotoxicity against MM cells. GENERAL SIGNIFICANCE The advanced understanding of the molecular mechanisms of XN action can be useful in developing therapeutic strategies to treat multiple myeloma.
Collapse
Affiliation(s)
- Adrianna Sławińska-Brych
- Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland.
| | - Barbara Zdzisińska
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Arkadiusz Czerwonka
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Magdalena Mizerska-Kowalska
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| | | | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| | - Mariusz Gagoś
- Department of Cell Biology, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland
| |
Collapse
|
30
|
Allegra A, Innao V, Allegra AG, Pugliese M, Di Salvo E, Ventura-Spagnolo E, Musolino C, Gangemi S. Lymphocyte Subsets and Inflammatory Cytokines of Monoclonal Gammopathy of Undetermined Significance and Multiple Myeloma. Int J Mol Sci 2019; 20:ijms20112822. [PMID: 31185596 PMCID: PMC6600674 DOI: 10.3390/ijms20112822] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/29/2019] [Accepted: 06/08/2019] [Indexed: 01/10/2023] Open
Abstract
Almost all multiple myeloma (MM) cases have been demonstrated to be linked to earlier monoclonal gammopathy of undetermined significance (MGUS). Nevertheless, there are no identified characteristics in the diagnosis of MGUS that have been helpful in differentiating subjects whose cancer may progress to a malignant situation. Regarding malignancy, the role of lymphocyte subsets and cytokines at the beginning of neoplastic diseases is now incontestable. In this review, we have concentrated our attention on the equilibrium between the diverse lymphocyte subsets and the cytokine system and summarized the current state of knowledge, providing an overview of the condition of the entire system in MGUS and MM. In an age where the therapy of neoplastic monoclonal gammopathies largely relies on drugs capable of acting on the immune system (immunomodulants, immunological checkpoint inhibitors, CAR-T), detailed knowledge of the the differences existing in benign and neoplastic forms of gammopathy is the main foundation for the adequate and optimal use of new drugs.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Vanessa Innao
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Andrea Gaetano Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Marta Pugliese
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Eleonora Di Salvo
- National Research Council of Italy (CNR)-Institute of Applied Science and Intelligent System (ISASI), 98164 Messina, Italy.
| | - Elvira Ventura-Spagnolo
- Legal Medicine Section, Department for Health Promotion and Mother-Child Care, University of Palermo, 90127 Palermo, Italy.
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125 Messina, Italy.
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital "G. Martino", Via Consolare Valeria SNC, 98125 Messina, Italy.
| |
Collapse
|
31
|
Huang FI, Wu YW, Sung TY, Liou JP, Lin MH, Pan SL, Yang CR. MPT0G413, A Novel HDAC6-Selective Inhibitor, and Bortezomib Synergistically Exert Anti-tumor Activity in Multiple Myeloma Cells. Front Oncol 2019; 9:249. [PMID: 31024851 PMCID: PMC6465934 DOI: 10.3389/fonc.2019.00249] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/19/2019] [Indexed: 12/16/2022] Open
Abstract
In multiple myeloma (MM), homeostasis is largely maintained by misfolded protein clearance via the proteasomal and aggresomal pathways. Histone deacetylase 6 (HDAC6) binds polyubiquitinated proteins and dynein motors and transports this protein cargo to the aggresome for further degradation. Accordingly, a combination of an HDAC6 inhibitor and bortezomib (BTZ) could increase ubiquitinated protein accumulation, leading to further apoptosis. Here we evaluated the anti-MM activity of MPT0G413, a novel specific HDAC6 inhibitor, using in vitro and in vivo models. MPT0G413 treatment more significantly inhibited cell growth in MM cells than in normal bone marrow cells. Furthermore, the combination of MPT0G413 and BTZ enhanced polyubiquitinated protein accumulation and synergistically reduced MM viability, increased caspase-3, caspase-8, caspase-9 levels, and cleaved poly (ADP) ribosome polymerase and also inhibited adherence of MM cells to bone marrow stromal cells (BMSC) and reduced VEGF and IL-6 levels and cell growth in a co-culture system. The combination treatment disturbed the bone marrow microenvironment and induced synergic, caspase-dependent apoptosis. Xenograft tumor growth significantly decreased in combination-treated SCID mice. In conclusion, MPT0G413 and BTZ synergistically inhibit MM viability, providing a framework for the clinical evaluation of combined therapies for MM.
Collapse
Affiliation(s)
- Fang-I Huang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Wen Wu
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Ting-Yi Sung
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiang Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shiow-Lin Pan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
32
|
Caffeic acid phenethyl ester exerts apoptotic and oxidative stress on human multiple myeloma cells. Invest New Drugs 2018; 37:837-848. [DOI: 10.1007/s10637-018-0701-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/14/2018] [Indexed: 01/02/2023]
|
33
|
Chen XY, Sun RX, Zhang WY, Liu T, Zheng YH, Wu Y. [Molecular mechanisms and relationship of M2-polarized macrophages with early response in multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 38:480-486. [PMID: 28655090 PMCID: PMC7342963 DOI: 10.3760/cma.j.issn.0253-2727.2017.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
目的 探讨替代活化型巨噬细胞(M2 MΦ)和多发性骨髓瘤(MM)早期治疗反应的关系及其在发病机制中的可能作用。 方法 采用免疫组化法标记240例MM患者骨髓标本中的MΦ;建立体外M2 MΦ诱导培养体系,构建Transwell共培养模型与RPMI 8226和U266细胞共培养,CCK-8法检测M2 MΦ对细胞增殖的影响,流式细胞术检测对地塞米松(1 µ mol/L)诱导骨髓瘤细胞凋亡的影响,ELISA法检测对TNF-α和IL-6表达的影响,real time PCR法检测对趋化因子、血管内皮生长因子(VEGF)及其受体表达的影响。 结果 ①依据骨髓组织M2 MΦ浸润程度将患者分为高浸润组(92例)和低浸润组(148例),高浸润组患者早期治疗有效率明显低于低浸润组,差异有统计学意义(23.9%对73.0%,χ2=60.31,P<0.001)。②培养24、36 h,共培养组细胞增殖能力较对照组显著上升:M2 MΦ+RPMI 8226细胞组与对照组比较,P值分别为0.005、0.020;M2 MΦ+U266细胞与对照组比较,P值分别为0.030、0.020。③地塞米松诱导后,共培养组与对照组比较,RPMI 8226细胞凋亡率下降(29.0%对71.0%,t=4.97,P=0.008),U266细胞凋亡率也下降(24.9%对67.7%,t=6.99,P=0.002)。④共培养48 h后,与对照组比较,加入M2 MΦ后可促进RPMI 8226和U266细胞分泌IL-6、TNF-α,促进表达CCL2、CCL3、CCR2、CCR5、VEGFA、VEGFR-1和VEGFR-2。 结论 MM患者骨髓组织M2 MΦ浸润程度和早期治疗反应相关。M2 MΦ通过促进骨髓瘤细胞分泌系列炎症因子、趋化因子和相关受体的表达,从而促进骨髓瘤细胞增殖以及保护骨髓瘤细胞免于凋亡。
Collapse
Affiliation(s)
- X Y Chen
- Department of Hematology and Research Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
34
|
Burwick N, Sharma S. Glucocorticoids in multiple myeloma: past, present, and future. Ann Hematol 2018; 98:19-28. [PMID: 30073393 DOI: 10.1007/s00277-018-3465-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Glucocorticoids are a backbone of treatment for multiple myeloma in both the upfront and relapsed/refractory setting. While glucocorticoids have single agent activity in multiple myeloma, in the modern era, they are paired with novel agents to induce high clinical response rates. On the other hand, toxicities of steroid therapy limit high dose delivery and impact patient quality of life. We provide a history of steroid use in multiple myeloma with the aim to understand how steroids have emerged and persisted in the treatment of multiple myeloma. We review mechanisms of glucocorticoid sensitivity and resistance and highlight potential future directions to evaluate steroid responsiveness. Further research in this area will aid in optimizing steroid utilization and help determine when glucocorticoid therapy may no longer benefit patients.
Collapse
Affiliation(s)
- Nicholas Burwick
- VA Puget Sound Health Care System, Seattle, WA, USA. .,Department of Medicine, University of Washington, 1705 NE Pacific St, M/S 358280, Seattle, WA, 98195, USA.
| | | |
Collapse
|
35
|
He Y, Li Z, Chen Z, Yu X, Ji Z, Wang J, Qian Y, Li L. Effects of VEGF-ANG-1-PLA nano-sustained release microspheres on proliferation and differentiation of ADSCs. Cell Biol Int 2018; 42:1060-1068. [PMID: 29745446 DOI: 10.1002/cbin.10986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/07/2018] [Indexed: 12/22/2022]
Abstract
The improvement of fat graft viability might depend on the presence of multipotent resident adipose derived stem cells (ADSCs) which is the important component of stromal vascular fraction (SVF). Vascular endothelial growth factor (VEGF) and angiogenin-1 (Ang-1) are responsible for neovascularization. However, their half-life is too short to produce a biological effect. We thus investigated whether VEGF-ANG-1-polylactic acid (PLA) microspheres could enhance the angiogenic properties of ADSCs. PLA microspheres containing VEGF and ANG-1 were prepared by in vitro ultrasonic emulsification and characterized according to their encapsulation efficiency (EE), drug-loading rate (DL), particle size, and drug release. The systemic toxicity of empty loaded nanospheres (NPs) and the ability of these microspheres to promote the proliferation and differentiation of ADSCs were evaluated. The EE and DL were above 86% and 0.0288%, respectively [corrected].The drug release was completed after 20 days. Systemic toxicity was verified in ADSCs that received the unloaded NPs. It was observed that ADSCs treated with VEGF-ANG-1-PLA microspheres had an increase in the proliferation and the number of CD31 positive cells. ADSCs proliferation and differentiation toward endothelial cells (ECs) could be enhanced by the addition of VEGF-ANG-1-PLA nano-sustained release microspheres.
Collapse
Affiliation(s)
- Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Zihao Li
- Hangzhou Medical College, Hangzhou, Zhejiang, 310053, China
| | - Zhuojie Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Xiaofang Yu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Ziwan Ji
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Jingping Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Yao Qian
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
36
|
The orally available multikinase inhibitor regorafenib (BAY 73-4506) in multiple myeloma. Ann Hematol 2018; 97:839-849. [DOI: 10.1007/s00277-018-3237-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023]
|
37
|
Gao M, Kong Y, Yang G, Gao L, Shi J. Multiple myeloma cancer stem cells. Oncotarget 2018; 7:35466-77. [PMID: 27007154 PMCID: PMC5085244 DOI: 10.18632/oncotarget.8154] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/02/2016] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) remains incurable despite much progress that has been made in the treatment of the disease. MM cancer stem cell (MMSC), a rare subpopulation of MM cells with the capacity for self-renewal and drug resistance, is considered to lead to disease relapse. Several markers such as side population (SP) and ALDH1+ have been used to identify MMSCs. However, ideally and more precisely, the identification of the MMSCs should rely on MMSCs phenotype. Unfortunately the MMSC phenotype has not been properly defined yet. Drug resistance is the most important property of MMSCs and contributes to disease relapse, but the mechanisms of drug resistance have not been fully understood. The major signaling pathways involved in the regulation of self-renewal and differentiation of MMSCs include Hedgehog (Hh), Wingless (Wnt), Notch and PI3K/Akt/mTOR. However, the precise role of these signaling pathways needs to be clarified. It has been reported that the microRNA profile of MMSCs is remarkably different than that of non-MMSCs. Therefore, the search for targeting MMSCs has also been focused on microRNAs. Complex and mutual interactions between the MMSC and the surrounding bone marrow (BM) microenvironment sustain self-renewal and survival of MMSC. However, the required molecules for the interaction of the MMSC and the surrounding BM microenvironment need to be further identified. In this review, we summarize the current state of knowledge of MMSCs regarding their phenotype, mechanisms of drug resistance, signaling pathways that regulate MMSCs self-renewal and differentiation, abnormal microRNAs expression, and their interactions with the BM microenvironment.
Collapse
Affiliation(s)
- Minjie Gao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanyuan Kong
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guang Yang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Gao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Inflammatory and Anti-Inflammatory Equilibrium, Proliferative and Antiproliferative Balance: The Role of Cytokines in Multiple Myeloma. Mediators Inflamm 2017; 2017:1852517. [PMID: 29089667 PMCID: PMC5635476 DOI: 10.1155/2017/1852517] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/11/2017] [Indexed: 12/23/2022] Open
Abstract
Multiple myeloma (MM) is typically exemplified by a desynchronized cytokine system with increased levels of inflammatory cytokines. We focused on the contrast between inflammatory and anti-inflammatory systems by assessing the role of cytokines and their influence on MM. The aim of this review is to summarize the available information to date concerning this equilibrium to provide an overview of the research exploring the roles of serum cytokines in MM. However, the association between MM and inflammatory cytokines appears to be inadequate, and other functions, such as pro-proliferative or antiproliferative effects, can assume the role of cytokines in the genesis and progression of MM. It is possible that inflammation, when guided by cancer-specific Th1 cells, may inhibit tumour onset and progression. In a Th1 microenvironment, proinflammatory cytokines (e.g., IL-6 and IL-1) may contribute to tumour eradication by attracting leucocytes from the circulation and by increasing CD4 + T cell activity. Hence, caution should be used when considering therapies that target factors with pro- or anti-inflammatory activity. Drugs that may reduce the tumour-suppressive Th1-driven inflammatory immune response should be avoided. A better understanding of the relationship between inflammation and myeloma will ensure more effective therapeutic interventions.
Collapse
|
39
|
Baek YY, Lee DK, Kim J, Kim JH, Park W, Kim T, Han S, Jeoung D, You JC, Lee H, Won MH, Ha KS, Kwon YG, Kim YM. Arg-Leu-Tyr-Glu tetrapeptide inhibits tumor progression by suppressing angiogenesis and vascular permeability via VEGF receptor-2 antagonism. Oncotarget 2017; 8:11763-11777. [PMID: 28052029 PMCID: PMC5355302 DOI: 10.18632/oncotarget.14343] [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: 12/21/2015] [Accepted: 12/03/2016] [Indexed: 02/06/2023] Open
Abstract
The tetrapeptide Arg-Leu-Tyr-Glu (RLYE) is known to inhibit vascular endothelial growth factor-A (VEGF-A)-induced angiogenesis in vitro. Herein, we examined its underlying mechanism and antitumor activity associated with vascular remodeling. RLYE inhibited VEGF-A-induced angiogenesis in a mouse model and suppressed VEGF-A-induced angiogenic signal cascades in human endothelial cells. However, RLYE showed no inhibitory effect on VEGF-A-induced proliferation and migration of multiple myeloma cells expressing VEGF receptor (VEGFR)-1, but not VEGFR-2. In addition, RLYE showed no inhibitory effect on angiogenic activities induced by VEGF-B, basic fibroblast growth factor, epithermal growth factor, sphingosine-1-phosphate, and placental growth factor. RLYE bound specifically to VEGFR-2 at the VEGF-A binding site, thereby blocking VEGF-A-VEGFR-2 binding and VEGF-A-induced VEGFR-2 internalization. The RLYE peptide inhibited tumor growth and metastasis via suppression of tumor angiogenesis in tumor-bearing mice. Moreover, RLYE showed a synergistic effect of the cytotoxic agent irinotecan on tumor cell apoptosis and tumor progression via tumor vessel normalization due to stabilization of VE-cadherin-mediated adherens junction, improvement of pericyte coverage, and inhibition of vascular leakage in tumors. Our results suggest that RLYE can be used as an antiangiogenic and tumor blood vessel remodeling agent for inhibition of tumor growth and metastasis by antagonizing VEGFR-2, with the synergistic anti-cancer effect via enhancement of drug delivery and therapeutic efficacy.
Collapse
Affiliation(s)
- Yi-Yong Baek
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Dong-Keon Lee
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Joohwan Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Ji-Hee Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Wonjin Park
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Taesam Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Sanghwa Han
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Ji Chang You
- Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | - Hansoo Lee
- Department of and Life Sciences, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-752, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-702, South Korea
| |
Collapse
|
40
|
Ntellas P, Perivoliotis K, Dadouli K, Koukoulis GK, Ioannou M. Microvessel Density as a Surrogate Prognostic Marker in Patients with Multiple Myeloma: A Meta-Analysis. Acta Haematol 2017; 138:77-84. [PMID: 28796988 DOI: 10.1159/000478085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIMS Bone marrow (BM) angiogenesis is considered a hallmark of multiple myeloma (MM) development and progression, and can be quantified with the use of microvessel density (MVD). The purpose of this study is to provide a review and a meta-analysis of the current literature regarding the prognostic value of MVD in the overall survival (OS) of MM patients. METHODS MEDLINE was screened for studies evaluating the OS of MM patients with regard to their MVD count in BM trephine. The pooled hazard ratio (HR) and its associated 95% confidence interval (CI) among MM patients with a high and low MVD count was the primary end point. Secondary outcomes included odds ratios (OR) for 12-, 36-, and 60-month survival. RESULTS Ten eligible trials were identified for the analysis of the primary end point and 9 for the secondary end points. Pooled HR for OS was 1.85 (95% CI: 1.25-2.73, p = 0.002). The pooled OR of survival were 1.59 (95% CI: 1.02-2.46, p = 0.04) at 12 months, 2.90 (95% CI: 1.68-5.03, p = 0.0001) at 36 months, and 3.42 (95% CI: 2.41-4.85, p < 0.00001) at 60 months, in favor of the low MVD group. CONCLUSION This meta-analysis provides persuasive evidence that MVD has significant impact on the clinical outcome of MM patients.
Collapse
Affiliation(s)
- Panagiotis Ntellas
- Department of Pathology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | | | | | | |
Collapse
|
41
|
Liu DC, Zheng X, Zho Y, Yi WR, Li ZH, Hu X, Yu AX. HIF-1α inhibits IDH-1 expression in osteosarcoma. Oncol Rep 2017; 38:336-342. [DOI: 10.3892/or.2017.5655] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/16/2016] [Indexed: 11/05/2022] Open
|
42
|
Kostjukova MN, Tupitsyn NN, Karamysheva AF. Vascular endothelial growth factor receptor 3 is a novel marker differing CD138-positive and CD138-negative multiple myeloma cells. Br J Haematol 2017; 181:840-843. [PMID: 28508400 DOI: 10.1111/bjh.14706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/23/2017] [Indexed: 11/27/2022]
Affiliation(s)
- Maria N Kostjukova
- Laboratory of Haematopoiesis Immunology, Institute of Clinical Oncology, N.N. Blokhin Russian Cancer Research Centre, Moscow, Russia
| | - Nikolai N Tupitsyn
- Laboratory of Haematopoiesis Immunology, Institute of Clinical Oncology, N.N. Blokhin Russian Cancer Research Centre, Moscow, Russia
| | - Aida F Karamysheva
- Laboratory of Tumour Cell Genetics, Institute of Carcinogenesis, N.N. Blokhin Russian Cancer Research Centre, Moscow, Russia
| |
Collapse
|
43
|
Xu Y, Zhang X, Liu H, Zhao P, Chen Y, Luo Y, Zhang Z, Wang X. Mesenchymal stromal cells enhance the suppressive effects ofmyeloid-derived suppressor cells of multiple myeloma. Leuk Lymphoma 2017; 58:2668-2676. [PMID: 28317413 DOI: 10.1080/10428194.2017.1298753] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Both mesenchymal stromal cells (MSCs) and myeloid-derived suppressor cells (MDSCs) have immunosuppressive properties, and their presence may confer a worse prognosis upon cancer patients. However, whether MSCs can enhance the immunosuppressive effects of MDSCs in MM remains unknown. We evaluated the influence of MSCs on MDSCs growth, apoptosis, and functions. Our results show that MSCs promote proliferation and inhibit apoptosis in MDSCs. Additionally, MSCs enhance the ability of MDSCs by inhibiting T-cell proliferation and IFN-γ production. Furthermore, both the mRNA and protein levels of Arg1 and NOS2 were upregulated in MDSCs. These results suggest that MSCs may exert immunomodulatory effects on MDSCs by upregulating Arg1 and NOS2.
Collapse
Affiliation(s)
- Yinhui Xu
- a Thoracic Surgery , The Second Affiliated Hospital of Guilin Medical University , Guilin, Guangxi , China
| | - Xiaoying Zhang
- b Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Cancer Hospital of Tianjin, Tianjin Medical University , Tianjin , China
| | - Hongbo Liu
- c Department of Laboratory Medicine , The Second Affiliated Hospital of Guilin Medical University , Guilin, Guangxi , China
| | - Pan Zhao
- b Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Cancer Hospital of Tianjin, Tianjin Medical University , Tianjin , China
| | - Yafang Chen
- b Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Cancer Hospital of Tianjin, Tianjin Medical University , Tianjin , China
| | - Yimin Luo
- d Department of Cancer Research , University of South China , Hengyang , China
| | - Zhiqing Zhang
- e Department of Neurology , the Fourth Central Hospital , Tianjin , China
| | - Xiaofang Wang
- b Department of Hematology, Key Laboratory of Cancer Prevention and Therapy , Cancer Hospital of Tianjin, Tianjin Medical University , Tianjin , China
| |
Collapse
|
44
|
de Waal EGM, Glaudemans AWJM, Schröder CP, Vellenga E, Slart RHJA. Nuclear medicine imaging of multiple myeloma, particularly in the relapsed setting. Eur J Nucl Med Mol Imaging 2016; 44:332-341. [PMID: 27900520 PMCID: PMC5215256 DOI: 10.1007/s00259-016-3576-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/18/2016] [Indexed: 02/04/2023]
Abstract
Multiple myeloma (MM) is characterized by a monoclonal plasma cell population in the bone marrow. Lytic lesions occur in up to 90 % of patients. For many years, whole-body X-ray (WBX) was the method of choice for detecting skeleton abnormalities. However, the value of WBX in relapsing disease is limited because lesions persist post-treatment, which restricts the capacity to distinguish between old, inactive skeletal lesions and new, active ones. Therefore, alternative techniques are necessary to visualize disease activity. Modern imaging techniques such as magnetic resonance imaging, positron emission tomography and computed tomography offer superior detection of myeloma bone disease and extramedullary manifestations. In particular, the properties of nuclear imaging enable the identification of disease activity by directly targeting the specific cellular properties of malignant plasma cells. In this review, an overview is provided of the effectiveness of radiopharmaceuticals that target metabolism, surface receptors and angiogenesis. The available literature data for commonly used nuclear imaging tracers, the promising first results of new tracers, and our pilot work indicate that a number of these radiopharmaceutical applications can be used effectively for staging and response monitoring of relapsing MM patients. Moreover, some tracers can potentially be used for radio immunotherapy.
Collapse
Affiliation(s)
- Esther G M de Waal
- Department of Hematology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands.
| | - Andor W J M Glaudemans
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Carolien P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Edo Vellenga
- Department of Hematology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Riemer H J A Slart
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Biomedical Photonic Imaging, University of Twente, Enschede, The Netherlands
| |
Collapse
|
45
|
Xu X, Liu J, Shen C, Ding L, Zhong F, Ouyang Y, Wang Y, He S. The role of ubiquitin-specific protease 14 (USP14) in cell adhesion-mediated drug resistance (CAM-DR) of multiple myeloma cells. Eur J Haematol 2016; 98:4-12. [DOI: 10.1111/ejh.12729] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Xiaohong Xu
- Department of Pathology; Affiliated Cancer Hospital of Nantong University; Nantong Jiangsu Province China
| | - Jing Liu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong University; Nantong Jiangsu Province China
| | - Chaoyan Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong University; Nantong Jiangsu Province China
| | - Linlin Ding
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong University; Nantong Jiangsu Province China
| | - Fei Zhong
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong University; Nantong Jiangsu Province China
| | - Yu Ouyang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong University; Nantong Jiangsu Province China
| | - Yuchan Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong University; Nantong Jiangsu Province China
| | - Song He
- Department of Pathology; Affiliated Cancer Hospital of Nantong University; Nantong Jiangsu Province China
| |
Collapse
|
46
|
Li Y, Zhang B, Li W, Wang L, Yan Z, Li H, Yao Y, Yao R, Xu K, Li Z. MiR-15a/16 regulates the growth of myeloma cells, angiogenesis and antitumor immunity by inhibiting Bcl-2, VEGF-A and IL-17 expression in multiple myeloma. Leuk Res 2016; 49:73-9. [PMID: 27596960 DOI: 10.1016/j.leukres.2016.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/21/2016] [Accepted: 08/26/2016] [Indexed: 01/13/2023]
Abstract
miRNAs have been reported to be involved in the pathogenesis of many cancers. In this article, we investigated the role and the mechanisms of miR-15a/16 in the pathogenesis of multiple myeloma (MM). We found that miR-15a/16 was down-regulated in bone marrow-derived mononuclear cells (BM-MNCs) of newly diagnosed patients with MM and the downregulation of miR-15a/16 was correlated with International Staging System (ISS) stage. We then demonstrated miR-15a/16 inhibited myeloma cells proliferation, and increased apoptosis rate of U266 cells by suppressing the expression of anti-apoptosis protein Bcl-2. We also found miR-15a/16 could decrease VEGF-A and IL-17 levels in the supernatant of myeloma cells. These results indicate that miR-15a/16 may function as a tumor suppressor in MM through multiple regulatory mechanisms and they may be potential targets for the therapy of MM.
Collapse
Affiliation(s)
- Yanjie Li
- Laboratory Center of Diagnostics, Xuzhou Medical University
| | - Bingyun Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Wenjing Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Lijin Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Zhiling Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Hujun Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Yao Yao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Ruosi Yao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University; Institute of Hematology, Xuzhou Medical University.
| |
Collapse
|
47
|
Citrullination of histone H3 drives IL-6 production by bone marrow mesenchymal stem cells in MGUS and multiple myeloma. Leukemia 2016; 31:373-381. [PMID: 27400413 PMCID: PMC5292682 DOI: 10.1038/leu.2016.187] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/24/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM), an incurable plasma cell malignancy, requires localisation within the bone marrow. This microenvironment facilitates crucial interactions between the cancer cells and stromal cell types that permit the tumour to survive and proliferate. There is increasing evidence that the bone marrow mesenchymal stem cell (BMMSC) is stably altered in patients with MM-a phenotype also postulated to exist in patients with monoclonal gammopathy of undetermined significance (MGUS) a benign condition that precedes MM. In this study, we describe a mechanism by which increased expression of peptidyl arginine deiminase 2 (PADI2) by BMMSCs in patients with MGUS and MM directly alters malignant plasma cell phenotype. We identify PADI2 as one of the most highly upregulated transcripts in BMMSCs from both MGUS and MM patients, and that through its enzymatic deimination of histone H3 arginine 26, PADI2 activity directly induces the upregulation of interleukin-6 expression. This leads to the acquisition of resistance to the chemotherapeutic agent, bortezomib, by malignant plasma cells. We therefore describe a novel mechanism by which BMMSC dysfunction in patients with MGUS and MM directly leads to pro-malignancy signalling through the citrullination of histone H3R26.
Collapse
|
48
|
Liu J, Wang Y, He S, Xu X, Huang Y, Tang J, Wu Y, Miao X, He Y, Wang Q, Liang L, Cheng C. Expression of vaccinia-related kinase 1 (VRK1) accelerates cell proliferation but overcomes cell adhesion mediated drug resistance (CAM-DR) in multiple myeloma. Hematology 2016; 21:603-612. [PMID: 27319807 PMCID: PMC9491125 DOI: 10.1080/10245332.2016.1147678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Objective: Vaccinia-related kinase 1 (VRK1) has been reported to participate in the development of a variety of tumors. However, the role of VRK1 in multiple myeloma (MM) has not been investigated. The present study was undertaken to determine the expression and biologic function of VRK1 in human MM. Methods: First, we constructed a model of cell adhesion in MM, the mRNA and protein level of VRK1 in suspension and adhesion model was analyzed by RT-PCR and western blot. Then, flow cytometry assay and western blot were used to investigate the mechanism of VRK1 in the proliferation of MM cells. In vitro, following using shRNA interfering VRK1 expression, we performed adhesion assay and cell viability assay to determine the effect of VRK1 on adhesive rate and drug sensitivity. Results: VRK1 was lowly expressed in adherent MM cells and highly expressed in suspended cells. In addition, VRK1 was positively correlated with the proliferation of MM cells by regulating the expression of cell cycle-related protein, such as cyclinD1, CDK2 and p27kip1. Furthermore, VRK1 could reverse cell adhesion mediated drug resistance (CAM-DR) by down-regulating the ability of cell adhesion. Conclusion and discussion: Our data supports a role for VRK1 in MM cell proliferation, adhesion, and drug resistance, and it may pave the way for a novel therapeutic approach for CAM-DR in MM.
Collapse
Affiliation(s)
- Jing Liu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yuchan Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Song He
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xiaohong Xu
- Department of Hematology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yuejiao Huang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jie Tang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yaxun Wu
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xiaobing Miao
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yunhua He
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Qiru Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Li Liang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Chun Cheng
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| |
Collapse
|
49
|
Babarović E, Valković T, Budisavljević I, Balen I, Štifter S, Duletić-Načinović A, Lučin K, Jonjić N. The expression of osteopontin and vascular endothelial growth factor in correlation with angiogenesis in monoclonal gammopathy of undetermined significance and multiple myeloma. Pathol Res Pract 2016; 212:509-16. [DOI: 10.1016/j.prp.2015.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 10/24/2015] [Accepted: 11/23/2015] [Indexed: 01/24/2023]
|
50
|
Krishnan SR, Jaiswal R, Brown RD, Luk F, Bebawy M. Multiple myeloma and persistence of drug resistance in the age of novel drugs (Review). Int J Oncol 2016; 49:33-50. [PMID: 27175906 DOI: 10.3892/ijo.2016.3516] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/17/2015] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is a mature B cell neoplasm that results in multi-organ failure. The median age of onset, diverse clinical manifestations, heterogeneous survival rate, clonal evolution, intrinsic and acquired drug resistance have impact on the therapeutic management of the disease. Specifically, the emergence of multidrug resistance (MDR) during the course of treatment contributes significantly to treatment failure. The introduction of the immunomodulatory agents and proteasome inhibitors has seen an increase in overall patient survival, however, for the majority of patients, relapse remains inevitable with evidence that these agents, like the conventional chemotherapeutics are also subject to the development of MDR. Clinical management of patients with MM is currently compromised by lack of a suitable procedure to monitor the development of clinical drug resistance in individual patients. The current MM prognostic measures fail to pick the clonotypic tumor cells overexpressing drug efflux pumps, and invasive biopsy is insufficient in detecting sporadic tumors in the skeletal system. This review summarizes the challenges associated with treating the complex disease spectrum of myeloma, with an emphasis on the role of deleterious multidrug resistant clones orchestrating relapse.
Collapse
Affiliation(s)
- Sabna Rajeev Krishnan
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ritu Jaiswal
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Ross D Brown
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Frederick Luk
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| | - Mary Bebawy
- Graduate School of Health, Discipline of Pharmacy, University of Technology, Sydney, NSW 2007, Australia
| |
Collapse
|