1
|
Zoabi M, Orbuch E, Komemi O, Jarchowsky-Dolberg O, Brin YS, Tartakover-Matalon S, Pasmanik-Chor M, Lishner M, Drucker L. Reduced elastin in multiple myeloma niche promotes cell proliferation. Exp Cell Res 2025; 444:114395. [PMID: 39725193 DOI: 10.1016/j.yexcr.2024.114395] [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: 07/23/2024] [Revised: 11/14/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
Multiple myeloma (MM) malignant plasma cells accumulate in the bone marrow (BM) where their interactions with the microenvironment promote disease progression and drug resistance. Previously, we have shown that bone marrow mesenchymal stem cells (BM-MSCs) (MM and normal donors- ND) derived extracellular matrix (ECM) affected MM cell lines differentially with a pro-MM effect attributed to MM-MSCs' ECM. Here we studied the composition of BM-MSC's ECM (ND versus MM) with focus on elastin (ELN). Isolated BM-MSCs' ECM mass spectrometry (proteomics) demonstrated distinct differences in proteins repertoire in a source dependent manner (MM or ND-MSCs) with ELN being the most significantly decreased protein in MM-MSCs ECM. To study this observation, we cultured MM cell lines (MM1S, RPMI-8226) and BM-MSCs with/without ELN and assayed the cells' phenotype. We demonstrated that supplementing ELN to MM cell lines reduced live cell counts and increased cell adhesion. ELN also decreased MM-MSCs' proliferation but did not affect ND-MSCs. Importantly, ELN addition to MM-MSC ECM abrogated its pro-MM effect on MM cells' proliferation. These novel findings underscore a suppressive role for ELN in MM and suggest it may hold potential diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Mozayan Zoabi
- Oncogenetics Laboratory, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Elina Orbuch
- Oncogenetics Laboratory, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Oded Komemi
- Oncogenetics Laboratory, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Osnat Jarchowsky-Dolberg
- Internal Medicine Department A, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Yaron Shraga Brin
- Orthopedics Department A, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel.
| | - Shelly Tartakover-Matalon
- Autoimmunity Laboratory, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, G.S.W. Faculty of Life Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Michael Lishner
- Oncogenetics Laboratory, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Hematology Unit, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| | - Liat Drucker
- Oncogenetics Laboratory, Meir Medical Center, Tchernichovsky St 59, Kfar Saba, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, PO Box 39040, Tel Aviv, Tel Aviv, Israel.
| |
Collapse
|
2
|
Tang W, Xu J, Xu C. Noncoding RNAs in the crosstalk between multiple myeloma cells and bone marrow microenvironment. Cancer Lett 2023; 556:216081. [PMID: 36739065 DOI: 10.1016/j.canlet.2023.216081] [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: 08/31/2022] [Revised: 01/18/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy; however, it remains incurable, and the underlying pathogenesis and mechanisms of drug resistance remain unclear. It is widely recognized that the bone marrow microenvironment plays a crucial role in regulating the immune response, inducing drug resistance, and promoting tumor proliferation and invasion in MM, and thus serves as a potential therapeutic target. Among the various signaling loops between myeloma cells and components of the microenvironment, noncoding RNAs are emerging as crucial regulators of intercellular communication within the microenvironment. Noncoding RNAs, such as microRNAs, long noncoding RNAs, circular RNAs, and PIWI-interacting RNAs, have been associated with numerous biological processes involved in myeloma cell growth, survival, migration, invasion, and drug resistance. This review summarizes recent advances in the regulatory mechanisms of noncoding RNAs involved in the interaction between the MM bone marrow microenvironment and discusses the therapeutic potential of noncoding RNAs in MM.
Collapse
Affiliation(s)
- Wenjiao Tang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Juan Xu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Caigang Xu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Zhao Q, Liu X, Yu C, Xiao Y. Macrophages and Bone Marrow-Derived Mesenchymal Stem Cells Work in Concert to Promote Fracture Healing: A Brief Review. DNA Cell Biol 2022; 41:276-284. [PMID: 35196145 DOI: 10.1089/dna.2021.0869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cell (BMSC)-based and macrophage-based cell therapy are regarded as promising strategies to promote fracture healing because of incredible osteogenic potential of BMSCs and typical immunomodulatory function of macrophages. Apart from their respective key roles, accumulative evidence has also demonstrated the importance of cross talk between these two cell types in fracture healing process. This review takes a deep insight into the recent research progress of the synergic performance of BMSCs and macrophages by discussing not only the cells own functions but also the relevant impact factors and mechanisms (ambient microenvironment stimulus, miRNAs, etc). The aim of this review is to provide some valuable cues and technique support for the macrophage- and BMSC-related research, which will be helpful to propel BMSC/macrophage-based combined cell therapy for bone fracture treatment.
Collapse
Affiliation(s)
- Qing Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Xinran Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Chuanying Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Dabbah M, Lishner M, Jarchowsky-Dolberg O, Tartakover-Matalon S, Brin YS, Pasmanik-Chor M, Neumann A, Drucker L. Ribosomal proteins as distinct "passengers" of microvesicles: new semantics in myeloma and mesenchymal stem cells' communication. Transl Res 2021; 236:117-132. [PMID: 33887527 DOI: 10.1016/j.trsl.2021.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 01/10/2023]
Abstract
Aberrant mesenchymal stem cells (MSCs) in multiple myeloma (MM) bone marrows (BM) promote disease progression and drug resistance. Here, we assayed the protein cargo transported from MM-MSCs to MM cells via microvesicles (MVs) with focus on ribosomal proteins (RPs) and assessment of their influence on translation initiation and design of MM phenotype. Proteomics analysis (mass spectrometry) demonstrated increased levels and repertoire of RPs in MM-MSCs MVs compared to normal donors (ND) counterparts (n = 3-8; P = 9.96E - 08). We limited the RPs load in MM-MSCs MVs (starvation, RSK and XPO1 inhibitions), reapplied the modified MVs to MM cell lines (U266, MM1S), and demonstrated that the RPs are essential to the proliferative effect of MM-MSCs MVs on MM cells (n = 3; P < 0.05). We also observed that inhibition with KPT-185 (XPO1 inhibitor) displayed the most extensive effect on RPs delivery into the MVs (↓80%; P = 3.12E - 05). Using flow cytometry we assessed the expression of select RPs (n = 10) in BM-MSCs cell populations (ND and MM; n ≥ 6 each). This demonstrated a heterogeneous expression of RPs in MM-MSCs with distinct subgroups, a phenomenon absent from ND-MSCs samples. These findings bring to light a new mechanism in which the tumor microenvironment participates in cancer promotion. MVs-mediated horizontal transfer of RPs between niche MSCs and myeloma cells is a systemic way to bestow pro-cancer advantages. This capacity also differentiates normal MSCs from the MM-modified MSCs and may mark their reprogramming. Future studies will be aimed at assessing the clinical and therapeutic potential of the increased RPs levels in MM-MSCs MVs.
Collapse
Affiliation(s)
- Mahmoud Dabbah
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Lishner
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Resaerch Institute, Meir Medical Center, Kfar saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Jarchowsky-Dolberg
- Hematology Unit, Meir Medical Center, Kfar saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shelly Tartakover-Matalon
- Autoimmunity laboratory, Meir Medical Center, Kfar saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaron S Brin
- Orthopedics Department, Meir Medical Center, Kfar Saba, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, G.S.W. Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Avivit Neumann
- Oncology Department, Rambam Medical Center, Haifa, Israel
| | - Liat Drucker
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Sackler faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
Abstract
Mesenchymal stem cells (MSCs) represent a promising source of cell-based therapies for treatment of a wide variety of injuries and diseases. Their tropism and migration to the damaged sites, which are elicited by cytokines secreted from tissues around pathology, are the prerequisite for tissue repair and regeneration. Better understanding of the elicited-migration of MSCs and discovering conditions that elevate their migration ability, will help to increase their homing to pathologies and improve therapeutic efficacy. It is increasingly recognized that microRNAs are important regulators of cell migration. Here we summarize current understanding of the microRNA-regulated migration of MSCs.
Collapse
|
6
|
Spelat R, Ferro F, Contessotto P, Warren NJ, Marsico G, Armes SP, Pandit A. A worm gel-based 3D model to elucidate the paracrine interaction between multiple myeloma and mesenchymal stem cells. Mater Today Bio 2020; 5:100040. [PMID: 32211606 PMCID: PMC7083757 DOI: 10.1016/j.mtbio.2019.100040] [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: 09/13/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 02/04/2023] Open
Abstract
Multiple myeloma (MM) is a malignancy of terminally-differentiated plasma cells that develops mainly inside the bone marrow (BM) microenvironment. It is well known that autocrine and paracrine signals are responsible for the progression of this disease but the precise mechanism and contributions from single cell remain largely unknown. Mesenchymal stem cells (MSC) are an important cellular component of the BM: they support MM growth by increasing its survival and chemo-resistance, but little is known about the paracrine signaling pathways. Three-dimensional (3D) models of MM-MSC paracrine interactions are much more biologically-relevant than simple 2D models and are considered essential for detailed studies of MM pathogenesis. Herein we present a novel 3D co-culture model designed to mimic the paracrine interaction between MSC and MM cells. MSC were embedded within a previously characterized thermoresponsive block copolymer worm gel that can induce stasis in human pluripotent stem cells (hPSC) and then co-cultured with MM cells. Transcriptional phenotyping of co-cultured cells indicated the dysregulation of genes that code for known disease-relevant factors, and also revealed IL-6 and IL-10 as upstream regulators. Importantly, we have identified a synergistic paracrine signaling pathway between IL-6 and IL-10 that plays a critical role in sustaining MM cell proliferation. Our findings indicate that this 3D co-culture system is a useful model to investigate the paracrine interaction between MM cells and the BM microenvironment in vitro. This approach has revealed a new mechanism that promotes the proliferation of MM cells and suggested a new therapeutic target.
Collapse
Affiliation(s)
- Renza Spelat
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway-H91 TK33, Ireland
| | - Federico Ferro
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway-H91 TK33, Ireland
| | - Paolo Contessotto
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway-H91 TK33, Ireland
| | - Nicholas J Warren
- Department of Chemistry, University of Sheffield, Sheffield, South Yorkshire S3 7HF, United Kingdom
| | - Grazia Marsico
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway-H91 TK33, Ireland
| | - Steven P Armes
- Department of Chemistry, University of Sheffield, Sheffield, South Yorkshire S3 7HF, United Kingdom
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway-H91 TK33, Ireland
| |
Collapse
|
7
|
Niche origin of mesenchymal stem cells derived microvesicles determines opposing effects on NSCLC: Primary versus metastatic. Cell Signal 2019; 65:109456. [PMID: 31672605 DOI: 10.1016/j.cellsig.2019.109456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/16/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023]
Abstract
Novel therapeutic approaches that address the malignant cells in their stroma microenvironment are urgently needed in lung cancer. The stroma resident mesenchymal stem cells (MSCs) interact with cancer cells in diverse ways including microvesicles (MVs) that transfer proteins and RNA species thereby modulating recipient cells' phenotype. Previously, we have demonstrated that MSCs' secretome from the primary non-small cell lung cancer (NSCLC) niche (lung) and metastatic niche (bone marrow (BM)) demonstrate opposite effects on NSCLC cells in a translation initiation (TI) dependent manner. Here, we examined the effect of MVs secreted from BM-MSCs' or lung-MSCs (healthy, NSCLC) to NSCLC phenotype. Briefly, NSCLC cell lines treated with Lung or BM-MSCs' MVs were assayed for viability (WST-1), cell count/death (trypan), migration (scratch), TI status and MAPKs activation (immunoblotting). Corresponding to previous published trends, Lung-MSCs' MVs promoted NSCLC cells' assayed traits whereas, BM-MSCs' MVs suppressed them. Activation of MAPKs and autophagy was registered in lung-MSCs MVs treated NSCLC cell lines only. Furthermore, lung-MSCs' MVs' treated NSCLC cells demonstrated an early (5min) activation of MAPKs and TI factors (peIF4E/peIF4GI) not evident in BM-MSCs MVs treated cells. These observations depict a role for MSCs'-MVs in NSCLC phenotype design and display distinct differences between the primary and metastatic niches that correspond to disease progression. In conclusion, the systemic nature of MVs marks them as attractive therapeutic markers/targets and we propose that identification of specific cargoes/signals that differentiate between MSCs MVs of primary and metastatic niches may introduce fresh therapeutic approaches.
Collapse
|
8
|
Dabbah M, Jarchowsky-Dolberg O, Attar-Schneider O, Tartakover Matalon S, Pasmanik-Chor M, Drucker L, Lishner M. Multiple myeloma BM-MSCs increase the tumorigenicity of MM cells via transfer of VLA4-enriched microvesicles. Carcinogenesis 2019; 41:100-110. [DOI: 10.1093/carcin/bgz169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/16/2019] [Accepted: 10/02/2019] [Indexed: 11/14/2022] Open
Abstract
Abstract
Multiple myeloma (MM) cells accumulate in the bone marrow (BM) where their interactions impede disease therapy. We have shown that microvesicles (MVs) derived from BM mesenchymal stem cells (MSCs) of MM patients promote the malignant traits via modulation of translation initiation (TI), whereas MVs from normal donors (ND) do not. Here, we observed that this phenomenon is contingent on a MVs’ protein constituent, and determined correlations between the MVs from the tumor microenvironment, for example, MM BM-MSCs and patients’ clinical characteristics. BM-MSCs’ MVs (ND/MM) proteomes were assayed (mass spectrometry) and compared. Elevated integrin CD49d (X80) and CD29 (X2) was determined in MM-MSCs’ MVs and correlated with patients’ staging and treatment response (free light chain, BM plasma cells count, stage, response to treatment). BM-MSCs’ MVs uptake into MM cell lines was assayed (flow cytometry) with/without integrin inhibitors (RGD, natalizumab, and anti-CD29 monoclonal antibody) and recipient cells were analyzed for cell count, migration, MAPKs, TI, and drug response (doxorubicin, Velcade). Their inhibition, particularly together, attenuated the uptake of MM-MSCs MVs (but not ND-MSCs MVs) into MM cells and reduced MM cells’ signaling, phenotype, and increased drug response. This study exposed a critical novel role for CD49d/CD29 on MM-MSCs MVs and presented a discriminate method to inhibit cancer promoting action of MM-MSCs MVs while retaining the anticancer function of ND-MSCs-MVs. Moreover, these findings demonstrate yet again the intricacy of the microenvironment involvement in the malignant process and highlight new therapeutic avenues to be explored.
Collapse
Affiliation(s)
- Mahmoud Dabbah
- Oncogenetic Laboratory, Kfar Saba
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | - Liat Drucker
- Oncogenetic Laboratory, Kfar Saba
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Lishner
- Oncogenetic Laboratory, Kfar Saba
- Hematology Unit, Kfar Saba
- Research Institute, Meir Medical Center, Kfar Saba
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Qi J, Shi LY, Wu Y, Shen XJ, Yuan J, Jin CJ, Cong H, Ju SQ. Epigenetic silencing of miR-335 induces migration by targeting insulin-like growth factor-1 receptor in multiple myeloma. Leuk Lymphoma 2019; 60:3188-3198. [PMID: 31190579 DOI: 10.1080/10428194.2019.1627534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a common hematological malignancy and remains incurable. MiRNA-335 is a classic tumor suppressor, yet its expression pattern and biological role in MM is unclear. The aim of the present study was to determine the expression pattern, biological role, and mechanism of miR-335 in MM. In this study, we found that miR-335 expression was decreased in MM. The promoter of miR-335 was also hypermethylated in MM. It was found that over-expression of miR-335 or 5-azacytidine treatment suppressed migration of MM cells and down-regulated the expression of IGF-1R. MiR-335 thus acts as a metastatic suppressor by targeting IGF-1R in MM. Moreover, aberrant promoter hyper-methylation is critical for miR-335 silencing in MM. We also found that miR-335 assisted in predicting both the prognosis and progression of disease in MM patients. Observations might offer a new complementary diagnostic and therapeutic target in MM.
Collapse
Affiliation(s)
- Jing Qi
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Lin-Ying Shi
- Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Yin Wu
- Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Xian-Juan Shen
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jie Yuan
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Chun-Jing Jin
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Hui Cong
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Shao-Qing Ju
- Center of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
10
|
Ibraheem A, Attar-Schneider O, Dabbah M, Dolberg Jarchowsky O, Tartakover Matalon S, Lishner M, Drucker L. BM-MSCs-derived ECM modifies multiple myeloma phenotype and drug response in a source-dependent manner. Transl Res 2019; 207:83-95. [PMID: 30738861 DOI: 10.1016/j.trsl.2019.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/17/2018] [Accepted: 01/14/2019] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) malignant plasma cells accumulate in the bone marrow (BM) where their interaction with the microenvironment promotes disease progression and drug resistance. Previously, we have shown that MM cells cocultured with BM-mesenchymal stem cells (MSCs) comodulated cells' phenotype in a MAPKs/translation initiation (TI)-dependent manner. Dissection of the coculture model showed that BM-MSCs secretomes and microvesicles (MVs) participate in this crosstalk. Here, we addressed the role of the BM-MSCs extracellular matrix (ECM). MM cell lines cultured on decellularized ECM of normal donors' (ND) or MM patients' BM-MSCs were assayed for phenotype (viability, cell count, death, proliferation, migration, and invasion), microRNAs (MIR125a-3p, MIR199a-3p) and targets, MAPKs, TI epithelial-to-mesenchymal transition (EMT), CXCR4, and autophagy. Drug (doxorubicin, velcade) response of MM cells cultured on ND/MM-MSCs' ECM with/without adhered MVs was also evaluated. ECM evoked opposite responses according to its origin: MM cells cultured on ND-MSCs' ECM demonstrated a rapid and continued decrease in MAPK/TI activation (↓10%-25%, P < 0.05) (15-24 hours) followed by diminished viability, cell count, proliferation, migration, and invasion (16-72 hours) (↓10%-50%, P < 0.05). In contrast, MM cells cultured on MM-MSCs' ECM displayed activated MAPK/TI, proliferation, EMT, and CXCR4 (↑15%-250%, P < 0.05). Corresponding changes in microRNAs relevant to the MM cells' altered phenotype were also determined. The hierarchy and interdependence of MAPKs/TI/autophagy/phenotype cascade were demonstrated. Finally, we showed that the ECM cooperates with MVs to modulate MM cells drug response. These data demonstrate the contribution of BM-MSCs' ECM to MM niche design and underscore the clinical potential of identifying targetable signals.
Collapse
Affiliation(s)
- Amjd Ibraheem
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Mahmoud Dabbah
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Dolberg Jarchowsky
- Hematology Clinique, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shelly Tartakover Matalon
- Gastroentroloy Laboratory, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Lishner
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Hematology Clinique, Meir Medical Center, Kfar Saba, Israel; Research Authority, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liat Drucker
- Oncogenetic Laboratory, Meir Medical Center, Kfar Saba, Israel; Research Authority, Meir Medical Center, Kfar Saba, Israel.
| |
Collapse
|
11
|
Attar-Schneider O, Drucker L, Gottfried M. The effect of mesenchymal stem cells' secretome on lung cancer progression is contingent on their origin: primary or metastatic niche. J Transl Med 2018; 98:1549-1561. [PMID: 30089856 DOI: 10.1038/s41374-018-0110-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/02/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
The fatality of non-small-cell lung cancer (NSCLC) and the role of the cancer microenvironment in its resistance to therapy are long recognized. Accumulating data allocate a significant role for mesenchymal stem cells (MSCs) in the malignant environment. Previously, we have demonstrated that MSCs from NSCLC metastatic bone marrow (BM) niche deleteriously affected NSCLC cells. Here, we have decided to examine the effect of MSCs from the primary niche of the lung (healthy or adjacent to tumor) on NSCLC phenotype. We cultured NSCLC cell lines with healthy/NSCLC lung-MSCs conditioned media (secretome) and showed elevation in cells' MAPKs and translation initiation signals, proliferation, viability, death, and migration. We also established enhanced autophagy and epithelial to mesenchymal transition processes. Moreover, we observed that MSCs from tumor adjacent sites (pathological niche) exhibited a more profound effect than MSCs from healthy lung tissue. Our findings underscore the capacity of the lung-MSCs to modulate NSCLC phenotype. Interestingly, both tumor adjacent (pathological) and distant lung-MSCs (healthy) promoted the NSCLC's TI, proliferation, migration, and epithelial to mesenchymal transition, yet the pathological MSCs displayed a greater affect. In conclusion, by comparing the effects of normal lung-MSCs, NSCLC adjacent MSCs, and BM-MSCs, we have established that the primary and metastatic niches display opposite and critical effects that promote the cancerous systemic state. Specifically, the primary site MSCs promote the expansion of the malignant clone and its dispersion, whereas the metastatic site MSCs facilitates the cells re-seeding. We suggest that sabotaging the cross-talk between MSCs and NSCLC affords effective means to inhibit lung cancer progression and will require different targeting strategies in accordance with niche/disease stage.
Collapse
Affiliation(s)
- Oshrat Attar-Schneider
- Lung Cancer Research, Lung Cancer Unit, Meir Medical Center, Kfar Saba, 44281, Israel. .,Oncogenetic Laboratories, Lung Cancer Unit, Meir Medical Center, Kfar Saba, 44281, Israel. .,Department of Oncology, Lung Cancer Unit, Meir Medical Center, Kfar Saba, 44281, Israel.
| | - Liat Drucker
- Oncogenetic Laboratories, Lung Cancer Unit, Meir Medical Center, Kfar Saba, 44281, Israel.,Sackler Faculty of Medicine, Tel Aviv University Ramat Aviv, Tel Aviv, 69978, Israel
| | - Maya Gottfried
- Lung Cancer Research, Lung Cancer Unit, Meir Medical Center, Kfar Saba, 44281, Israel.,Department of Oncology, Lung Cancer Unit, Meir Medical Center, Kfar Saba, 44281, Israel.,Sackler Faculty of Medicine, Tel Aviv University Ramat Aviv, Tel Aviv, 69978, Israel
| |
Collapse
|
12
|
Mehdi SJ, Johnson SK, Epstein J, Zangari M, Qu P, Hoering A, van Rhee F, Schinke C, Thanendrarajan S, Barlogie B, Davies FE, Morgan GJ, Yaccoby S. Mesenchymal stem cells gene signature in high-risk myeloma bone marrow linked to suppression of distinct IGFBP2-expressing small adipocytes. Br J Haematol 2018; 184:578-593. [PMID: 30408155 DOI: 10.1111/bjh.15669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
Recent studies suggest that multiple myeloma (MM) induces proliferation and expansion of bone marrow (BM) mesenchymal stem cells (MSCs), but others showed that MM cells induce MSC senescence. To clarify the interaction between MM and MSCs, we exploited our established MSC gene signature to identify gene expression changes in myeloma MSCs and associated functional differences. Single MSCs from patients with MM had changes in expression of genes associated with cellular proliferation and senescence and a higher proportion of senescent cells and lower proliferative potential than those from age-matched healthy donors. Single MSCs from both sources heterogeneously express MSC genes associated with adipogenesis and osteoblastogenesis. We identified the gene encoding insulin-like growth factor-binding protein 2 (IGFBP2), an MSC gene commonly altered in high risk MM, as under-expressed. Morphologically, IGFBP2+ cells are underrepresented in MM BM compared to smouldering MM. Strong IGFBP2 and adiponectin co-expression was detected in a subset of small adipocytes. Co-culturing normal MSCs with myeloma cells suppressed MSC differentiation to adipocytes and osteoblasts, and reduced expression of IGFBP2 and adiponectin. Recombinant IGFBP2 blocked IGF1-mediated myeloma cell growth. Our data demonstrate that myeloma MSCs are less proliferative and that IGFBP2+ small adipocytes are a distinct mesenchymal cell population suppressed by myeloma.
Collapse
Affiliation(s)
- Syed J Mehdi
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sarah K Johnson
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Joshua Epstein
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maurizio Zangari
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Pingping Qu
- Cancer Research and Biostatistics, Seattle, WA, USA
| | | | - Frits van Rhee
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Carolina Schinke
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Bart Barlogie
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Faith E Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shmuel Yaccoby
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
13
|
Yang H, Chennamaneni LR, Ho MWT, Ang SH, Tan ESW, Jeyaraj DA, Yeap YS, Liu B, Ong EH, Joy JK, Wee JLK, Kwek P, Retna P, Dinie N, Nguyen TTH, Tai SJ, Manoharan V, Pendharkar V, Low CB, Chew YS, Vuddagiri S, Sangthongpitag K, Choong ML, Lee MA, Kannan S, Verma CS, Poulsen A, Lim S, Chuah C, Ong TS, Hill J, Matter A, Nacro K. Optimization of Selective Mitogen-Activated Protein Kinase Interacting Kinases 1 and 2 Inhibitors for the Treatment of Blast Crisis Leukemia. J Med Chem 2018; 61:4348-4369. [PMID: 29683667 DOI: 10.1021/acs.jmedchem.7b01714] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by bcr-abl1, a constitutively active tyrosine kinase fusion gene responsible for an abnormal proliferation of leukemic stem cells (LSCs). Inhibition of BCR-ABL1 kinase activity offers long-term relief to CML patients. However, for a proportion of them, BCR-ABL1 inhibition will become ineffective at treating the disease, and CML will progress to blast crisis (BC) CML with poor prognosis. BC-CML is often associated with excessive phosphorylated eukaryotic translation initiation factor 4E (eIF4E), which renders LSCs capable of proliferating via self-renewal, oblivious to BCR-ABL1 inhibition. In vivo, eIF4E is exclusively phosphorylated on Ser209 by MNK1/2. Consequently, a selective inhibitor of MNK1/2 should reduce the level of phosphorylated eIF4E and re-sensitize LSCs to BCR-ABL1 inhibition, thus hindering the proliferation of BC LSCs. We report herein the structure-activity relationships and pharmacokinetic properties of a selective MNK1/2 inhibitor clinical candidate, ETC-206, which in combination with dasatinib prevents BC-CML LSC self-renewal in vitro and enhances dasatinib antitumor activity in vivo.
Collapse
Affiliation(s)
- Haiyan Yang
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Lohitha Rao Chennamaneni
- Organic Chemistry, Institute of Chemical and Engineering Sciences (ICES), A*STAR , 8 Biomedical Grove, Neuros, #07-01 , 138665 Singapore
| | - Melvyn Wai Tuck Ho
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Shi Hua Ang
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Eldwin Sum Wai Tan
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | | | - Yoon Sheng Yeap
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Boping Liu
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Esther Hq Ong
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Joma Kanikadu Joy
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - John Liang Kuan Wee
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Perlyn Kwek
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Priya Retna
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Nurul Dinie
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Thuy Thi Hanh Nguyen
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Shi Jing Tai
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Vithya Manoharan
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Vishal Pendharkar
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Choon Bing Low
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Yun Shan Chew
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Susmitha Vuddagiri
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Kanda Sangthongpitag
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Meng Ling Choong
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - May Ann Lee
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | | | - Chandra S Verma
- Bioinformatics Institute (BII) , A*STAR , 30 Biopolis Street, #07-01 Matrix , 138671 Singapore.,School of Biological Sciences , Nanyang Technological University , 60 Nanyang Drive , 637551 Singapore.,Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , 117543 Singapore
| | - Anders Poulsen
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Sharon Lim
- Duke-NUS Medical School , 8 College Road , 169857 Singapore
| | - Charles Chuah
- Duke-NUS Medical School , 8 College Road , 169857 Singapore
| | - Tiong Sin Ong
- Duke-NUS Medical School , 8 College Road , 169857 Singapore.,Department of Medicine , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Jeffrey Hill
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Alex Matter
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Kassoum Nacro
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| |
Collapse
|
14
|
Dabbah M, Attar-Schneider O, Tartakover Matalon S, Shefler I, Jarchwsky Dolberg O, Lishner M, Drucker L. Microvesicles derived from normal and multiple myeloma bone marrow mesenchymal stem cells differentially modulate myeloma cells' phenotype and translation initiation. Carcinogenesis 2017; 38:708-716. [PMID: 28838065 DOI: 10.1093/carcin/bgx045] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) cells' interaction with the bone marrow (BM) microenvironment critically hinders disease therapy. Previously, we showed that MM co-culture with BM-mesenchymal stem cells (MSCs) caused co-modulation of translation initiation (TI) and cell phenotype and implicated secreted components, specifically microvesicles (MVs). Here, we studied the role of the BM-MSCs [normal donors (ND) and MM] secreted MVs in design of MM cells' phenotype, TI and signaling. BM-MSCs' MVs collected from BM-MSCs (MM/ND) cultures were applied to MM cell lines. After MVs uptake confirmation, the MM cells were assayed for viability, cell count and death, proliferation, migration, invasion, autophagy, TI status (factors, regulators, targets) and MAPKs activation. The interdependence of MAPKs, TI and autophagy was determined (inhibitors). ND-MSCs MVs' treated MM cells demonstrated a rapid (5 min) activation of MAPKs followed by a persistent decrease (1-24 h), while MM-MSCs MVs' treated cells demonstrated a rapid and continued (5 min-24 h) activation of MAPKs and TI (↑25-200%, P < 0.05). Within 24 h, BM-MSCs MVs were internalized by MM cells evoking opposite responses according to MVs origin. ND-MSCs' MVs decreased viability, proliferation, migration and TI (↓15-80%; P < 0.05), whereas MM-MSCs' MVs increased them (↑10-250%, P < 0.05). Inhibition of MAPKs in MM-MSCs MVs treated MM cells decreased TI and inhibition of autophagy elevated cell death. These data demonstrate that BM-MSCs MVs have a fundamental effect on MM cells phenotype in accordance with normal or pathological source implemented via TI modulation. Future studies will aim to elucidate the involvement of MVs-MM receptor ligand interactions and cargo transfer in our model.
Collapse
Affiliation(s)
- Mahmoud Dabbah
- Oncogenetic, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Shelly Tartakover Matalon
- Oncogenetic, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Michael Lishner
- Oncogenetic, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Internal Medicine A, Meir Medical Center, Kfar Saba, Israel
| | - Liat Drucker
- Oncogenetic, Meir Medical Center, Kfar Saba, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|