1
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
2
|
Zhai Y, Cheng Y, Yuan Y, Meng X, Li Y, Wang Y, Ren T, Li S, Sun H. Increased thrombospondin-1 levels contribute to epileptic susceptibility in neonatal hyperthermia without seizures via altered synaptogenesis. Cell Death Discov 2024; 10:73. [PMID: 38346981 PMCID: PMC10861539 DOI: 10.1038/s41420-024-01837-3] [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: 06/03/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Childhood febrile seizures (FS) represent one of the most common types of seizures and may lead to severe neurological damage and an increased risk of epilepsy. However, most children with fevers do not show clinical manifestations of convulsions, and the consequences of hyperthermia without seizures remain elusive. This study focused on hyperthermia not reaching the individual's seizure threshold (sub-FS stimulus). Changes in thrombospondin-1 (TSP-1) levels, synapses, seizure susceptibility, and seizure severity in subsequent FS were investigated in rats exposed to sub-FS stimuli. Pharmacological and genetic interventions were used to explore the role of TSP-1 in sub-FS-induced effects. We found that after sub-FS stimuli, the levels of TSP-1 and synapses, especially excitatory synapses, were concomitantly increased, with increased epilepsy and FS susceptibility. Moreover, more severe neuronal damage was found in subsequent FS. These changes were temperature dependent. Reducing TSP-1 levels by genetic intervention or inhibiting the activation of transforming growth factor-β1 (TGF-β1) by Leu-Ser-Lys-Leu (LSKL) led to lower synapse/excitatory synapse levels, decreased epileptic susceptibility, and attenuated neuronal injury after FS stimuli. Our study confirmed that even without seizures, hyperthermia may promote synaptogenesis, increase epileptic and FS susceptibility, and lead to more severe neuronal damage by subsequent FS. Inhibition of the TSP-1/TGF-β1 pathway may be a new therapeutic target to prevent detrimental sub-FS sequelae.
Collapse
Affiliation(s)
- Yujie Zhai
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yao Cheng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yi Yuan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xianfeng Meng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yang Li
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yan Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Tianpu Ren
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Shucui Li
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
3
|
Yi Z, Ma T, Liu J, Tie W, Li Y, Bai J, Li L, Zhang L. LGR4 promotes tumorigenesis by activating TGF-β1/Smad signaling pathway in multiple myeloma. Cell Signal 2023; 110:110814. [PMID: 37473901 DOI: 10.1016/j.cellsig.2023.110814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/09/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Multiple myeloma (MM) is a common hematologic malignancy that remains incurable. Although accumulating evidence suggests that the leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4) plays a biological function in a variety of cancers, its biological function and molecular mechanisms in MM are unclear. In the present study, we found that LGR4 was significantly upregulated in MM tissues and cells. In vitro and in vivo experiments showed that knockdown of LGR4 significantly inhibited proliferation of MM cells, promoted apoptosis and arrested cell cycle in G1. Overexpression showed the opposite effect. Mechanistic studies revealed that LGR4 could interact with TGF-β1 and regulate TGF-β1 expression, thereby activating the TGF-β1/Smad signaling pathway and promoting MM progression. LGR4 may be a potential new target for MM diagnosis and treatment.
Collapse
Affiliation(s)
- Zhigang Yi
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China; Department of Pediatric Orthopedics and Pediatrics, Lanzhou University Second Hospital, Lanzhou, China
| | - Tao Ma
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China; Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia Liu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenting Tie
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanhong Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jun Bai
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China.
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
4
|
Zhang J, Du J, Jin Z, Qian J, Xu J. A novel immunogenic cell death signature for the prediction of prognosis and therapies in glioma. PeerJ 2023; 11:e15615. [PMID: 37456890 PMCID: PMC10348309 DOI: 10.7717/peerj.15615] [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: 04/26/2023] [Accepted: 06/01/2023] [Indexed: 07/18/2023] Open
Abstract
Glioma is a primary cranial malignancy with high recurrence rate, poor prognosis and high mortality. However, the roles of immunogenic cell death (ICD) in glioma remain unclear. Twenty ICD genes were analyzed to be differentially expressed between glioma tissues and non-tumor tissues in 371 glioma patients from The Cancer Genome Atlas (TCGA). Patients were classified into three subgroups via unsupervised clustering. Interestingly, the features of cell-infiltrating from three clusters were matched with three immune phenotypes. An applied scoring system was built depending on the expression of hub ICD-related genes. Notably, the ICD-related score was linked with immune checkpoints and the prognosis of glioma patients. In addition, the applied risk model could be used for the prediction of the effect of chemotherapy and immunotherapy for glioma patients. Furthermore, MYD88 was identified to play key roles in the risk model for glioma patients. MYD88 was specifically expressed in malignant cells and validated to correlate with cell proliferation and invasion. Ligand-receptor pairs are determined as novel communications indicating between immunocytes and malignant cells. Therefore, our research established an ICD-related score to investigate the potential effect to chemotherapy and immunotherapy for glioma patients and indicated that MYD88 was a key role in this risk model.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Oncology, People’s Hospital of Chizhou, Chizhou, China
| | - Jin Du
- Department of Neurosurgery, People’s Hospital of Chizhou, Chizhou, China
| | - Zhihai Jin
- Department of Orthopedics, Handan First Hospital, Handan, China
| | - Jiang Qian
- Department of Oncology, People’s Hospital of Chizhou, Chizhou, China
| | - Jinfa Xu
- Department of Oncology, People’s Hospital of Chizhou, Chizhou, China
| |
Collapse
|
5
|
Matamala Montoya M, van Slobbe GJJ, Chang JC, Zaal EA, Berkers CR. Metabolic changes underlying drug resistance in the multiple myeloma tumor microenvironment. Front Oncol 2023; 13:1155621. [PMID: 37091139 PMCID: PMC10117897 DOI: 10.3389/fonc.2023.1155621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells in the bone marrow (BM). MM remains an incurable disease, with the majority of patients experiencing multiple relapses from different drugs. The MM tumor microenvironment (TME) and in particular bone-marrow stromal cells (BMSCs) play a crucial role in the development of drug resistance. Metabolic reprogramming is emerging as a hallmark of cancer that can potentially be exploited for cancer treatment. Recent studies show that metabolism is further adjusted in MM cells during the development of drug resistance. However, little is known about the role of BMSCs in inducing metabolic changes that are associated with drug resistance. In this Perspective, we summarize current knowledge concerning the metabolic reprogramming of MM, with a focus on those changes associated with drug resistance to the proteasome inhibitor Bortezomib (BTZ). In addition, we present proof-of-concept fluxomics (glucose isotope-tracing) and Seahorse data to show that co-culture of MM cells with BMSCs skews the metabolic phenotype of MM cells towards a drug-resistant phenotype, with increased oxidative phosphorylation (OXPHOS), serine synthesis pathway (SSP), TCA cycle and glutathione (GSH) synthesis. Given the crucial role of BMSCs in conveying drug resistance, insights into the metabolic interaction between MM and BMSCs may ultimately aid in the identification of novel metabolic targets that can be exploited for therapy.
Collapse
Affiliation(s)
- María Matamala Montoya
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Gijs J. J. van Slobbe
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jung-Chin Chang
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Esther A. Zaal
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Celia R. Berkers, ; Esther A. Zaal,
| | - Celia R. Berkers
- Division Cell Biology, Metabolism & Cancer, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Celia R. Berkers, ; Esther A. Zaal,
| |
Collapse
|
6
|
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
|
7
|
Matsumura K, Hayashi H, Uemura N, Ogata Y, Zhao L, Sato H, Shiraishi Y, Kuroki H, Kitamura F, Kaida T, Higashi T, Nakagawa S, Mima K, Imai K, Yamashita YI, Baba H. Thrombospondin-1 overexpression stimulates loss of Smad4 and accelerates malignant behavior via TGF-β signal activation in pancreatic ductal adenocarcinoma. Transl Oncol 2022; 26:101533. [PMID: 36115074 PMCID: PMC9483797 DOI: 10.1016/j.tranon.2022.101533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is characterized by abundant stroma and cancer-associated fibroblasts (CAFs) provide a favorable tumor microenvironment. Smad4 is known as tumor suppressor in several types of cancers including PDAC, and loss of Smad4 triggers accelerated cell invasiveness and metastatic potential. The thrombospondin-1 (TSP-1) can act as a major activator of latent transforming growth factor-β (TGF-β) in vivo. However, the roles of TSP-1 and the mediator of Smad4 loss and TGF-β signal activation during PDAC progression have not yet been addressed. The aim is to elucidate the biological role of TSP-1 in PDAC progression. METHODS AND RESULTS High substrate stiffness stimulated TSP-1 expression in CAFs, and TSP-1 knockdown inhibited cell proliferation with suppressed profibrogenic and activated stroma-related gene expressions in CAFs. Paracrine TSP-1 treatment for PDAC cells promoted cell proliferation and epithelial mesenchymal transition (EMT) with activated TGF-β signals such as phosphorylated Akt and Smad2/3 expressions. Surprisingly, knockdown of DPC4 (Smad4 gene) induced TSP-1 overexpression with TGF-β signal activation in PDAC cells. Interestingly, TSP-1 overexpression also induced downregulation of Smad4 expression and enhanced cell proliferation in vitro and in vivo. Treatment with LSKL peptide, which antagonizes TSP-1-mediated latent TGF-β activation, attenuated cell proliferation, migration and chemoresistance with enhanced apoptosis in PDAC cells. CONCLUSIONS TSP-1 derived from CAFs stimulates loss of Smad4 expression in cancer cells and accelerates malignant behavior by TGF-β signal activation in PDAC. TSP-1 could be a novel therapeutic target, not only for CAFs in stiff stroma, but also for cancer cells in the PDAC microenvironment.
Collapse
Affiliation(s)
- Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yoko Ogata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Liu Zhao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yuta Shiraishi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hideyuki Kuroki
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Fumimasa Kitamura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Takayoshi Kaida
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Takaaki Higashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan.
| |
Collapse
|
8
|
Beider K, Voevoda-Dimenshtein V, Zoabi A, Rosenberg E, Magen H, Ostrovsky O, Shimoni A, Weiss L, Abraham M, Peled A, Nagler A. CXCL13 chemokine is a novel player in multiple myeloma osteolytic microenvironment, M2 macrophage polarization, and tumor progression. J Hematol Oncol 2022; 15:144. [PMID: 36217194 PMCID: PMC9549634 DOI: 10.1186/s13045-022-01366-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/02/2022] [Indexed: 12/04/2022] Open
Abstract
Background We assessed the mechanism by which multiple myeloma (MM) shapes the bone marrow (BM) microenvironment and affects MΦ polarization. Methods In vivo xenograft model of BM-disseminated human myeloma, as well as analysis of MM cell lines, stromal components, and primary samples from patients with MM, was utilized. Results Analysis of the BM from MM-bearing mice inoculated with human CXCR4-expressing RPMI8226 cells revealed a significant increase in M2 MΦ cell numbers (p < 0.01). CXCL13 was one of the most profoundly increased factors upon MM growth with increased levels in the blood of MM-bearing animals. Myeloid cells were the main source of the increased murine CXCL13 detected in MM-infiltrated BM. MM cell lines induced CXCL13 and concurrent expression of M2 markers (MERTK, CD206, CD163) in co-cultured human MΦ in vitro. Interaction with MΦ reciprocally induced CXCL13 expression in MM cell lines. Mechanistically, TGFβ signaling was involved in CXCL13 induction in MM cells, while BTK signaling was implicated in MM-stimulated increase of CXCL13 in MΦ. Recombinant CXCL13 increased RANKL expression and induced TRAP+ osteoclast (OC) formation in vitro, while CXCL13 neutralization blocked these activities. Moreover, mice inoculated with CXCL13-silenced MM cells developed significantly lower BM disease. Reduced tumor load correlated with decreased numbers of M2 MΦ in BM, decreased bone disease, and lower expression of OC-associated genes. Finally, higher levels of CXCL13 were detected in the blood and BM samples of MM patients in comparison with healthy individuals. Conclusions Altogether, our findings suggest that bidirectional interactions of MΦ with MM tumor cells result in M2 MΦ polarization, CXCL13 induction, and subsequent OC activation, enhancing their ability to support bone resorption and MM progression. CXCL13 may thus serve as a potential novel target in MM. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01366-5.
Collapse
Affiliation(s)
- Katia Beider
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | | | - Ali Zoabi
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Evgenia Rosenberg
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Hila Magen
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Olga Ostrovsky
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Avichai Shimoni
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Arnon Nagler
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel.
| |
Collapse
|
9
|
Yi Z, Ma T, Liu J, Tie W, Li Y, Bai J, Li L, Zhang L. The yin–yang effects of immunity: From monoclonal gammopathy of undetermined significance to multiple myeloma. Front Immunol 2022; 13:925266. [PMID: 35958625 PMCID: PMC9357873 DOI: 10.3389/fimmu.2022.925266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/30/2022] [Indexed: 01/10/2023] Open
Abstract
Multiple myeloma (MM) is the third most common malignant neoplasm of the hematological system. It often develops from monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) precursor states. In this process, the immune microenvironment interacts with the MM cells to exert yin and yang effects, promoting tumor progression on the one hand and inhibiting it on the other. Despite significant therapeutic advances, MM remains incurable, and the main reason for this may be related to the complex and variable immune microenvironment. Therefore, it is crucial to investigate the dynamic relationship between the immune microenvironment and tumors, to elucidate the molecular mechanisms of different factors in the microenvironment, and to develop novel therapeutic agents targeting the immune microenvironment of MM. In this paper, we review the latest research progress and describe the dual influences of the immune microenvironment on the development and progression of MM from the perspective of immune cells and molecules.
Collapse
Affiliation(s)
- Zhigang Yi
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- Department of Pediatric Orthopedics and Pediatrics Lanzhou University Second Hospital, Lanzhou, China
| | - Tao Ma
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia Liu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenting Tie
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanhong Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jun Bai
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| |
Collapse
|
10
|
Timmins MA, Ringshausen I. Transforming Growth Factor-Beta Orchestrates Tumour and Bystander Cells in B-Cell Non-Hodgkin Lymphoma. Cancers (Basel) 2022; 14:1772. [PMID: 35406544 PMCID: PMC8996985 DOI: 10.3390/cancers14071772] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Transforming growth factor-beta (TGFB) is a critical regulator of normal haematopoiesis. Dysregulation of the TGFB pathway is associated with numerous haematological malignancies including myelofibrosis, acute myeloid leukaemia, and lymphoid disorders. TGFB has classically been seen as a negative regulator of proliferation in haematopoiesis whilst stimulating differentiation and apoptosis, as required to maintain homeostasis. Tumours frequently develop intrinsic resistant mechanisms to homeostatic TGFB signalling to antagonise its tumour-suppressive functions. Furthermore, elevated levels of TGFB enhance pathogenesis through modulation of the immune system and tumour microenvironment. Here, we review recent advances in the understanding of TGFB signalling in B-cell malignancies with a focus on the tumour microenvironment. Malignant B-cells harbour subtype-specific alterations in TGFB signalling elements including downregulation of surface receptors, modulation of SMAD signalling proteins, as well as genetic and epigenetic aberrations. Microenvironmental TGFB generates a protumoural niche reprogramming stromal, natural killer (NK), and T-cells. Increasingly, evidence points to complex bi-directional cross-talk between cells of the microenvironment and malignant B-cells. A greater understanding of intercellular communication and the context-specific nature of TGFB signalling may provide further insight into disease pathogenesis and future therapeutic strategies.
Collapse
Affiliation(s)
- Matthew A. Timmins
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospital, Cambridge CB2 0AH, UK
| | - Ingo Ringshausen
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospital, Cambridge CB2 0AH, UK
| |
Collapse
|
11
|
Swamydas M, Murphy EV, Ignatz-Hoover JJ, Malek E, Driscoll JJ. Deciphering mechanisms of immune escape to inform immunotherapeutic strategies in multiple myeloma. J Hematol Oncol 2022; 15:17. [PMID: 35172851 PMCID: PMC8848665 DOI: 10.1186/s13045-022-01234-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/03/2022] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma is an incurable cancer characterized by the uncontrolled growth of malignant plasma cells nurtured within a permissive bone marrow microenvironment. While patients mount numerous adaptive immune responses directed against their disease, emerging data demonstrate that tumor intrinsic and extrinsic mechanisms allow myeloma cells to subvert host immunosurveillance and resist current therapeutic strategies. Myeloma downregulates antigens recognized by cellular immunity and modulates the bone marrow microenvironment to promote uncontrolled tumor proliferation, apoptotic resistance, and further hamper anti-tumor immunity. Additional resistance often develops after an initial clinical response to small molecules, immune-targeting antibodies, immune checkpoint blockade or cellular immunotherapy. Profound quantitative and qualitative dysfunction of numerous immune effector cell types that confer anti-myeloma immunity further supports myelomagenesis, disease progression and the emergence of drug resistance. Identification of tumor intrinsic and extrinsic resistance mechanisms may direct the design of rationally-designed drug combinations that prevent or overcome drug resistance to improve patient survival. Here, we summarize various mechanisms of immune escape as a means to inform novel strategies that may restore and improve host anti-myeloma immunity.
Collapse
Affiliation(s)
| | - Elena V Murphy
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH, USA
| | - James J Ignatz-Hoover
- Seidman Cancer Center, University Hospitals, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Hematopoietic and Immune Cancer Biology Program, Cleveland, OH, USA
| | - Ehsan Malek
- Seidman Cancer Center, University Hospitals, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Hematopoietic and Immune Cancer Biology Program, Cleveland, OH, USA
| | - James J Driscoll
- Seidman Cancer Center, University Hospitals, Cleveland, OH, USA. .,Case Comprehensive Cancer Center, Hematopoietic and Immune Cancer Biology Program, Cleveland, OH, USA.
| |
Collapse
|
12
|
Reina-Ortiz C, Giraldos D, Azaceta G, Palomera L, Marzo I, Naval J, Villalba M, Anel A. Harnessing the Potential of NK Cell-Based Immunotherapies against Multiple Myeloma. Cells 2022; 11:cells11030392. [PMID: 35159200 PMCID: PMC8834301 DOI: 10.3390/cells11030392] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cell-based therapies have emerged as promising anticancer treatments due to their potency as cytolytic effectors and synergy with concurrent treatments. Multiple myeloma (MM) is an aggressive B-cell malignancy that, despite development of novel therapeutic agents, remains incurable with a high rate of relapse. In MM, the inhospitable tumor microenvironment prevents host NK cells from exerting their cytolytic function. The development of NK cell immunotherapy works to overcome this altered immune landscape and can be classified in two major groups based on the origin of the cell: autologous or allogeneic. In this review, we compare the treatments in each group, such as autologous chimeric antigen receptor (CAR) NKs and allogeneic off-the-shelf NK cell infusions, and their combinatorial effect with existing MM therapies including monoclonal antibodies and proteasome inhibitors. We also discuss their placement in clinical treatment regimens based on the immune profile of each patient. Through this examination, we would like to discover precisely when each NK cell-based treatment will produce the maximum benefit to the MM patient.
Collapse
Affiliation(s)
- Chantal Reina-Ortiz
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
- Correspondence: (C.R.-O.); (A.A.)
| | - David Giraldos
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Gemma Azaceta
- Hematology Department, Lozano Blesa Hospital, 50009 Zaragoza, Spain; (G.A.); (L.P.)
| | - Luis Palomera
- Hematology Department, Lozano Blesa Hospital, 50009 Zaragoza, Spain; (G.A.); (L.P.)
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
| | - Martín Villalba
- Institut of Regenerative Medicine and Biotherapy, University of Montpellier, INSERM, CNRS, University Hospital Center Montpellier, 34000 Montpellier, France;
- Institut Sainte-Catherine, 84918 Avignon, France
| | - Alberto Anel
- Apoptosis, Immunity & Cancer Group, Department Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (D.G.); (I.M.); (J.N.)
- Correspondence: (C.R.-O.); (A.A.)
| |
Collapse
|
13
|
Zhang C, Xu X, Trotter TN, Gowda PS, Lu Y, Suto MJ, Javed A, Murphy-Ullrich JE, Li J, Yang Y. Runx2 deficiency in osteoblasts promotes myeloma resistance to bortezomib by increasing TSP-1-dependent TGF-β1 activation and suppressing immunity in bone marrow. Mol Cancer Ther 2021; 21:347-358. [PMID: 34907087 DOI: 10.1158/1535-7163.mct-21-0310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/25/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a plasma cell malignancy that thrives in the bone marrow (BM). The proteasome inhibitor bortezomib (BTZ) is one of the most effective front-line chemotherapeutic drugs for MM; however, 15-20% of high-risk patients do not respond to or become resistant to this drug and the mechanisms of chemoresistance remain unclear. We previously demonstrated that MM cells inhibit Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OBs), and that this OB-Runx2 deficiency induces a cytokine-rich and immunosuppressive microenvironment in the BM. In the current study, we assessed the impact of OB-Runx2 deficiency on the outcome of BTZ treatment using OB-Runx2+/+ and OB-Runx2-/- mouse models of MM. In vitro and in vivo experiments revealed that OB-Runx2 deficiency induces MM cell resistance to BTZ via the upregulation of immunosuppressive myeloid-derived suppressor cells (MDSCs), downregulation of cytotoxic T cells, and activation of TGF-β1 in the BM. In MM tumor-bearing OB-Runx2-/- mice, treatment with SRI31277, an antagonist of thrombospondin-1 (TSP-1)-mediated TGF-β1 activation, reversed the BM immunosuppression and significantly reduced tumor burden. Furthermore, treatment with SRI31277 combined with BTZ alleviated MM cell resistance to BTZ-induced apoptosis caused by OB-Runx2 deficiency in co-cultured cells and produced a synergistic effect on tumor burden in OB-Runx2-/- mice. Depletion of MDSCs by 5-fluorouracil or gemcitabine similarly reversed the immunosuppressive effects and BTZ resistance induced by OB-Runx2 deficiency in tumor-bearing mice, indicating the importance of the immune environment for drug resistance and suggesting new strategies to overcome BTZ resistance in the treatment of MM.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Hematology, First Affiliated Hospital of Sun Yat-sen University
| | - Xiaoxuan Xu
- Department of Hematology, Guangzhou First People's Hospital, the Second Affiliated Hospital of South China University of Technology
| | | | | | - Yun Lu
- Radiology, University of Alabama at Birmingham
| | | | - Amjad Javed
- 3Comprehensive Cancer Center and the Center for Metabolic Bone Disease, University of Alabama at Birmingham
| | - Joanne E Murphy-Ullrich
- Pathology, Cell Developmental and Integrative Biology, and Ophthalmology, University of Alabama at Birmingham
| | - Juan Li
- First Affiliated Hospital of Sun Yat-sen University
| | - Yang Yang
- Pathology, University of Alabama at Birmingham
| |
Collapse
|
14
|
Andrews RE, Brown JE, Lawson MA, Chantry AD. Myeloma Bone Disease: The Osteoblast in the Spotlight. J Clin Med 2021; 10:jcm10173973. [PMID: 34501423 PMCID: PMC8432062 DOI: 10.3390/jcm10173973] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/17/2022] Open
Abstract
Lytic bone disease remains a life-altering complication of multiple myeloma, with up to 90% of sufferers experiencing skeletal events at some point in their cancer journey. This tumour-induced bone disease is driven by an upregulation of bone resorption (via increased osteoclast (OC) activity) and a downregulation of bone formation (via reduced osteoblast (OB) activity), leading to phenotypic osteolysis. Treatments are limited, and currently exclusively target OCs. Despite existing bone targeting therapies, patients successfully achieving remission from their cancer can still be left with chronic pain, poor mobility, and reduced quality of life as a result of bone disease. As such, the field is desperately in need of new and improved bone-modulating therapeutic agents. One such option is the use of bone anabolics, drugs that are gaining traction in the osteoporosis field following successful clinical trials. The prospect of using these therapies in relation to myeloma is an attractive option, as they aim to stimulate OBs, as opposed to existing therapeutics that do little to orchestrate new bone formation. The preclinical application of bone anabolics in myeloma mouse models has demonstrated positive outcomes for bone repair and fracture resistance. Here, we review the role of the OB in the pathophysiology of myeloma-induced bone disease and explore whether novel OB targeted therapies could improve outcomes for patients.
Collapse
Affiliation(s)
- Rebecca E. Andrews
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
- Correspondence:
| | - Janet E. Brown
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| | - Michelle A. Lawson
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
| | - Andrew D. Chantry
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield S10 2RX, UK; (J.E.B.); (M.A.L.); (A.D.C.)
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| |
Collapse
|
15
|
Fan X, Huang J, Xu C, Bao M, Xia W, Zhu C. Differential expression of microRNAs in human endometrium after implantation of an intrauterine contraceptive device containing copper. Mol Hum Reprod 2021; 27:6357049. [PMID: 34427668 DOI: 10.1093/molehr/gaab052] [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: 05/07/2021] [Revised: 08/02/2021] [Indexed: 11/13/2022] Open
Abstract
Intrauterine devices containing copper placement will release a large amount of Cu2+ into the uterine fluid, leading to local endometrial damage and inflammation, which is considered to be one of the causes of abnormal uterine bleeding. Studies have shown that the metabolism and function of metal ions are related to the regulation of microRNA. The aims of this study were to investigate changes in endometrial microRNA levels after implantation of an intrauterine device containing copper and to preliminarily explore the signalling pathways involved in abnormal uterine bleeding. The subjects were fertile women, aged 25-35, without major obstetrics and gynaecology diseases. Human endometrial tissues were collected before implantation or removal of the intrauterine device containing copper. High-throughput microRNA sequencing was performed on human endometrial tissues, and real-time quantitative PCR, western blotting and immunohistochemistry were used to detect the expression of relevant genes. MicroRNA sequencing results showed that 72 miRNAs were differentially expressed in the endometrial tissue after the insertion of the intrauterine device containing copper. Implantation of an intrauterine device containing copper implantation can up-regulate the expression of miR-144-3p in endometrial tissue, and therefore, decreases the mRNA and protein expression levels of genes related to endometrial injury and tissue repair, including the MT/NF-κB/MMP damage pathway and the THBS-1/TGF-β/SMAD3 repair pathway. In this study, the molecular mechanisms of abnormal uterine bleeding due to an intrauterine device containing copper were preliminarily investigated. The information will be beneficial for the clinical treatment of abnormal uterine bleeding caused by intrauterine device.
Collapse
Affiliation(s)
- Xiaorong Fan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Huang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengcheng Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Bao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xia
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changhong Zhu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Mukkamalla SKR, Malipeddi D. Myeloma Bone Disease: A Comprehensive Review. Int J Mol Sci 2021; 22:6208. [PMID: 34201396 PMCID: PMC8227693 DOI: 10.3390/ijms22126208] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 05/17/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a neoplastic clonal proliferation of plasma cells in the bone marrow microenvironment, characterized by overproduction of heavy- and light-chain monoclonal proteins (M-protein). These proteins are mainly found in the serum and/or urine. Reduction in normal gammaglobulins (immunoparesis) leads to an increased risk of infection. The primary site of origin is the bone marrow for nearly all patients affected by MM with disseminated marrow involvement in most cases. MM is known to involve bones and result in myeloma bone disease. Osteolytic lesions are seen in 80% of patients with MM which are complicated frequently by skeletal-related events (SRE) such as hypercalcemia, bone pain, pathological fractures, vertebral collapse, and spinal cord compression. These deteriorate the patient's quality of life and affect the overall survival of the patient. The underlying pathogenesis of myeloma bone disease involves uncoupling of the bone remodeling processes. Interaction of myeloma cells with the bone marrow microenvironment promotes the release of many biochemical markers including osteoclast activating factors and osteoblast inhibitory factors. Elevated levels of osteoclast activating factors such as RANK/RANKL/OPG, MIP-1-α., TNF-α, IL-3, IL-6, and IL-11 increase bone resorption by osteoclast stimulation, differentiation, and maturation, whereas osteoblast inhibitory factors such as the Wnt/DKK1 pathway, secreted frizzle related protein-2, and runt-related transcription factor 2 inhibit osteoblast differentiation and formation leading to decreased bone formation. These biochemical factors also help in development and utilization of appropriate anti-myeloma treatments in myeloma patients. This review article summarizes the pathophysiology and the recent developments of abnormal bone remodeling in MM, while reviewing various approved and potential treatments for myeloma bone disease.
Collapse
Affiliation(s)
| | - Dhatri Malipeddi
- Internal Medicine, Canton Medical Education Foundation/NEOMED, Canton, OH 44710, USA;
| |
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: 21] [Impact Index Per Article: 7.0] [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
|
Kaur S, Bronson SM, Pal-Nath D, Miller TW, Soto-Pantoja DR, Roberts DD. Functions of Thrombospondin-1 in the Tumor Microenvironment. Int J Mol Sci 2021; 22:4570. [PMID: 33925464 PMCID: PMC8123789 DOI: 10.3390/ijms22094570] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
The identification of thrombospondin-1 as an angiogenesis inhibitor in 1990 prompted interest in its role in cancer biology and potential as a therapeutic target. Decreased thrombospondin-1 mRNA and protein expression are associated with progression in several cancers, while expression by nonmalignant cells in the tumor microenvironment and circulating levels in cancer patients can be elevated. THBS1 is not a tumor suppressor gene, but the regulation of its expression in malignant cells by oncogenes and tumor suppressor genes mediates some of their effects on carcinogenesis, tumor progression, and metastasis. In addition to regulating angiogenesis and perfusion of the tumor vasculature, thrombospondin-1 limits antitumor immunity by CD47-dependent regulation of innate and adaptive immune cells. Conversely, thrombospondin-1 is a component of particles released by immune cells that mediate tumor cell killing. Thrombospondin-1 differentially regulates the sensitivity of malignant and nonmalignant cells to genotoxic stress caused by radiotherapy and chemotherapy. The diverse activities of thrombospondin-1 to regulate autophagy, senescence, stem cell maintenance, extracellular vesicle function, and metabolic responses to ischemic and genotoxic stress are mediated by several cell surface receptors and by regulating the functions of several secreted proteins. This review highlights progress in understanding thrombospondin-1 functions in cancer and the challenges that remain in harnessing its therapeutic potential.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| | - Steven M. Bronson
- Department of Internal Medicine, Section of Molecular Medicine, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Dipasmita Pal-Nath
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| | - Thomas W. Miller
- Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, 13273 Marseille, France
| | - David R. Soto-Pantoja
- Department of Surgery and Department of Cancer Biology, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; (S.K.); (D.P.-N.)
| |
Collapse
|
19
|
Directly targeting c-Myc contributes to the anti-multiple myeloma effect of anlotinib. Cell Death Dis 2021; 12:396. [PMID: 33854043 PMCID: PMC8046985 DOI: 10.1038/s41419-021-03685-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/21/2022]
Abstract
Despite the significant advances in the treatment of multiple myeloma (MM), this disease is still considered incurable because of relapse and chemotherapy resistance, underscoring the need to seek novel therapies with different mechanisms. Anlotinib, a novel multi-targeted tyrosine kinase inhibitor (TKI), has exhibited encouraging antitumor activity in several preclinical and clinical trials, but its effect on MM has not been studied yet. In this study, we found that anlotinib exhibits encouraging cytotoxicity in MM cells, overcomes the protective effect of the bone marrow microenvironment and suppresses tumor growth in the MM mouse xenograft model. We further examined the underlying molecular mechanism and found that anlotinib provokes cell cycle arrest, induces apoptosis and inhibits multiple signaling pathways. Importantly, we identify c-Myc as a novel direct target of anlotinib. The enhanced ubiquitin proteasomal degradation of c-Myc contributes to the cell apoptosis induced by anlotinib. In addition, anlotinib also displays strong cytotoxicity against bortezomib-resistant MM cells. Our study demonstrates the extraordinary anti-MM effect of anlotinib both in vitro and in vivo, which provides solid evidence and a promising rationale for future clinical application of anlotinib in the treatment of human MM.
Collapse
|
20
|
Fang L, Xia C, Li S, Xiao L, Jin H. Osteosclerosis and osteoporosis - different bone impairments from high bone turnover of skeletal fluorosis: a case study on the femoral head. Pathology 2021:S0031-3025(21)00082-9. [PMID: 33823986 DOI: 10.1016/j.pathol.2020.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/14/2020] [Accepted: 11/20/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Liang Fang
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, China
| | - Chenjie Xia
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, China
| | - Shaoguang Li
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Luwei Xiao
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, China; Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou, China; Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
21
|
Immunogenomic identification and characterization of granulocytic myeloid-derived suppressor cells in multiple myeloma. Blood 2021; 136:199-209. [PMID: 32325491 DOI: 10.1182/blood.2019004537] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/03/2020] [Indexed: 12/17/2022] Open
Abstract
Granulocytic myeloid-derived suppressor cells (G-MDSCs) promote tumor growth and immunosuppression in multiple myeloma (MM). However, their phenotype is not well established for accurate monitoring or clinical translation. We aimed to provide the phenotypic profile of G-MDSCs based on their prognostic significance in MM, immunosuppressive potential, and molecular program. The preestablished phenotype of G-MDSCs was evaluated in bone marrow samples from controls and MM patients using multidimensional flow cytometry; surprisingly, we found that CD11b+CD14-CD15+CD33+HLADR- cells overlapped with common eosinophils and neutrophils, which were not expanded in MM patients. Therefore, we relied on automated clustering to unbiasedly identify all granulocytic subsets in the tumor microenvironment: basophils, eosinophils, and immature, intermediate, and mature neutrophils. In a series of 267 newly diagnosed MM patients (GEM2012MENOS65 trial), only the frequency of mature neutrophils at diagnosis was significantly associated with patient outcome, and a high mature neutrophil/T-cell ratio resulted in inferior progression-free survival (P < .001). Upon fluorescence-activated cell sorting of each neutrophil subset, T-cell proliferation decreased in the presence of mature neutrophils (0.5-fold; P = .016), and the cytotoxic potential of T cells engaged by a BCMA×CD3-bispecific antibody increased notably with the depletion of mature neutrophils (fourfold; P = .0007). Most interestingly, RNA sequencing of the 3 subsets revealed that G-MDSC-related genes were specifically upregulated in mature neutrophils from MM patients vs controls because of differential chromatin accessibility. Taken together, our results establish a correlation between the clinical significance, immunosuppressive potential, and transcriptional network of well-defined neutrophil subsets, providing for the first time a set of optimal markers (CD11b/CD13/CD16) for accurate monitoring of G-MDSCs in MM.
Collapse
|
22
|
Song Y, Du T, Ray A, Chauhan K, Samur M, Munshi N, Chauhan D, Anderson KC. Identification of novel anti-tumor therapeutic target via proteomic characterization of ubiquitin receptor ADRM1/Rpn13. Blood Cancer J 2021; 11:13. [PMID: 33441535 PMCID: PMC7806750 DOI: 10.1038/s41408-020-00398-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/28/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023] Open
Affiliation(s)
- Yan Song
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ting Du
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Arghya Ray
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Krishan Chauhan
- Department of Biomedical Engineering (BME), Wentworth Institute of Technology, Boston, MA, USA
| | - Mehmet Samur
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nikhil Munshi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dharminder Chauhan
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
The extracellular matrix: A key player in the pathogenesis of hematologic malignancies. Blood Rev 2020; 48:100787. [PMID: 33317863 DOI: 10.1016/j.blre.2020.100787] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/10/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Hematopoietic stem and progenitor cells located in the bone marrow lay the foundation for multiple lineages of mature hematologic cells. Bone marrow niches are architecturally complex with specific cellular, physiochemical, and biomechanical factors. Increasing evidence suggests that the bone marrow microenvironment contributes to the pathogenesis of hematological neoplasms. Numerous studies have deciphered the role of genetic mutations and chromosomal translocations in the development hematologic malignancies. Significant progress has also been made in understanding how the cellular components and cytokine interactions within the bone marrow microenvironment promote the evolution of hematologic cancers. Although the extracellular matrix is known to be a key player in the pathogenesis of various diseases, it's role in the progression of hematologic malignancies is less understood. In this review, we discuss the interactions between the extracellular matrix and malignant cells, and provide an overview of the role of extracellular matrix remodeling in sustaining hematologic malignancies.
Collapse
|
24
|
Diaz-delCastillo M, Chantry AD, Lawson MA, Heegaard AM. Multiple myeloma-A painful disease of the bone marrow. Semin Cell Dev Biol 2020; 112:49-58. [PMID: 33158730 DOI: 10.1016/j.semcdb.2020.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Multiple myeloma is a bone marrow neoplasia with an incidence of 6/100,000/year in Europe. While the disease remains incurable, the development of novel treatments such as autologous stem cell transplantation, proteasome inhibitors and monoclonal antibodies has led to an increasing subset of patients living with long-term myeloma. However, more than two thirds of patients suffer from bone pain, often described as severe, and knowledge on the pain mechanisms and its effect on their health-related quality of life (HRQoL) is limited. In this review, we discuss the mechanisms of myeloma bone disease, the currently available anti-myeloma treatments and the lessons learnt from clinical studies regarding HRQoL in myeloma patients. Moreover, we discuss the mechanisms of cancer-induced bone pain and the knowledge that animal models of myeloma-induced bone pain can provide to identify novel analgesic targets. To date, information regarding bone pain and HRQoL in myeloma patients is still scarce and an effort should be made to use standardised questionnaires to assess patient-reported outcomes that allow inter-study comparisons of the available clinical data.
Collapse
Affiliation(s)
- Marta Diaz-delCastillo
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, Copenhagen Ø DK-2100, Denmark; Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK.
| | - Andrew D Chantry
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK
| | - Michelle A Lawson
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, Copenhagen Ø DK-2100, Denmark
| |
Collapse
|
25
|
Influence of the TGF-β Superfamily on Osteoclasts/Osteoblasts Balance in Physiological and Pathological Bone Conditions. Int J Mol Sci 2020; 21:ijms21207597. [PMID: 33066607 PMCID: PMC7589189 DOI: 10.3390/ijms21207597] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/19/2022] Open
Abstract
The balance between bone forming cells (osteoblasts/osteocytes) and bone resorbing cells (osteoclasts) plays a crucial role in tissue homeostasis and bone repair. Several hormones, cytokines, and growth factors-in particular the members of the TGF-β superfamily such as the bone morphogenetic proteins-not only regulate the proliferation, differentiation, and functioning of these cells, but also coordinate the communication between them to ensure an appropriate response. Therefore, this review focuses on TGF-β superfamily and its influence on bone formation and repair, through the regulation of osteoclastogenesis, osteogenic differentiation of stem cells, and osteoblasts/osteoclasts balance. After introducing the main types of bone cells, their differentiation and cooperation during bone remodeling and fracture healing processes are discussed. Then, the TGF-β superfamily, its signaling via canonical and non-canonical pathways, as well as its regulation by Wnt/Notch or microRNAs are described and discussed. Its important role in bone homeostasis, repair, or disease is also highlighted. Finally, the clinical therapeutic uses of members of the TGF-β superfamily and their associated complications are debated.
Collapse
|
26
|
Carminati L, Taraboletti G. Thrombospondins in bone remodeling and metastatic bone disease. Am J Physiol Cell Physiol 2020; 319:C980-C990. [PMID: 32936697 DOI: 10.1152/ajpcell.00383.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thrombospondins (TSPs) are a family of five multimeric matricellular proteins. Through a wide range of interactions, TSPs play pleiotropic roles in embryogenesis and in tissue remodeling in adult physiology as well as in pathological conditions, including cancer development and metastasis. TSPs are active in bone remodeling, the process of bone resorption (osteolysis) and deposition (osteogenesis) that maintains bone homeostasis. TSPs are particularly involved in aberrant bone remodeling, including osteolytic and osteoblastic skeletal cancer metastasis, frequent in advanced cancers such as breast and prostate carcinoma. TSPs are major players in the bone metastasis microenvironment, where they finely tune the cross talk between tumor cells and bone resident cells in the metastatic niche. Each TSP family member has different effects on the differentiation and activity of bone cells-including the bone-degrading osteoclasts and the bone-forming osteoblasts-with different outcomes on the development and growth of osteolytic and osteoblastic metastases. Here, we overview the involvement of TSP family members in the bone tissue microenvironment, focusing on their activity on osteoclasts and osteoblasts in bone remodeling, and present the evidence to date of their roles in bone metastasis establishment and growth.
Collapse
Affiliation(s)
- Laura Carminati
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giulia Taraboletti
- Laboratory of Tumor Microenvironment, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
27
|
Liu Z, Gao H, Peng Q, Yang Y. Long Noncoding RNA LUCAT1 Promotes Multiple Myeloma Cell Growth by Regulating the TGF-β Signaling Pathway. Technol Cancer Res Treat 2020; 19:1533033820945770. [PMID: 32812490 PMCID: PMC7440725 DOI: 10.1177/1533033820945770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: Long noncoding RNAs (lncRNAs) are potential biomarkers for cancers. Nevertheless, the ability of long noncoding RNA lung cancer-associated transcript 1 in patients with multiple myeloma remains unknown. The purpose of this current study was to figure out its function in multiple myeloma. Methods: Firstly, the expression of long noncoding RNA lung cancer-associated transcript 1 in cancer or normal tissues and serum from patients with multiple myeloma and normal donors was detected. Secondly, the expression of long noncoding RNA lung cancer-associated transcript 1 was overexpressed or silenced in U266 and H929 cells, respectively to detect changes of proliferation and apoptosis in multiple myeloma in vitro. Subsequently, the expression of transforming growth factor-β signaling pathway-related proteins was detected by western blot analysis. Finally, the effect of long noncoding RNA lung cancer-associated transcript 1 on the growth of multiple myeloma cells in vivo was evaluated by tumor xenograft in nude mice. Results: Long noncoding RNA lung cancer-associated transcript 1 was increased in cancer tissues and serum of patients with multiple myeloma as well as multiple myeloma cells, which was correlated with dismal prognosis of patients with multiple myeloma. Overexpression of long noncoding RNA lung cancer-associated transcript 1 promoted the activity of U266 and H929 cells, while inhibition of long noncoding RNA lung cancer-associated transcript 1 suppressed the activity of U266 and H929 cells. In addition, long noncoding RNA lung cancer-associated transcript 1 was found to promote activation of the transforming growth factor-β signaling pathway. Furthermore, long noncoding RNA lung cancer-associated transcript 1 knockdown restricted the growth of multiple myeloma cells in vivo. Conclusion: This study suggests that suppression of long noncoding RNA lung cancer-associated transcript 1 inhibits the activation of transforming growth factor-β signaling pathway, thereby inhibiting the growth of multiple myeloma cells.
Collapse
Affiliation(s)
- Zhaoyu Liu
- 74648Department of Medical Oncology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Hong Gao
- 74648Department of Gastroenterology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Qing Peng
- 74648Department of Medical Oncology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Yongheng Yang
- 74648Department of Medical Oncology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| |
Collapse
|
28
|
Deng H, Sun Y, Zeng W, Li H, Guo M, Yang L, Lu B, Yu B, Fan G, Gao Q, Jiang X. New Classification of Macrophages in Plaques: a Revolution. Curr Atheroscler Rep 2020; 22:31. [PMID: 32556603 DOI: 10.1007/s11883-020-00850-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Macrophages play vital roles in the development of atherosclerosis in responding to lipid accumulation and inflammation. Macrophages were classified as inflammatory (M1) and alternatively activated (M2) macrophage types based on results of in vitro experiments. On the other hand, the composition of macrophages in vivo is more complex and remains unresolved. This review summarizes the transcriptional variations of macrophages in atherosclerosis plaques that were discovered by single-cell RNA sequencing (scRNA-seq) to better understand their contribution to atherosclerosis. RECENT FINDINGS ScRNA-seq provides a more detailed transcriptional landscape of macrophages in atherosclerosis, which challenges the traditional view. By mining the data of GSE97310, we discovered the transcriptional variations of macrophages in LDLR-/- mice that were fed with high-fat diet (HFD) for 11 and 20 weeks. Cells were represented in a two-dimensional tSNE plane and clusters were identified and annotated via Seurat and SingleR respectively, which were R toolkits for single-cell genomics. The results showed that in healthy conditions, Trem2hi (high expression of triggering receptors expressed on myeloid cells 2)-positive, inflammatory, and resident-like macrophages make up 68%, 18%, and 6% of total macrophages respectively. When mice were fed with HFD for 11 weeks, Trem2hi, monocytes, and monocyte-derived dendritic cells take possession of 40%, 18%, and 17% of total macrophages respectively. After 20 weeks of HFD feeding, Trem2hi, inflammatory, and resident-like macrophages occupied 12%, 37%, and 35% of total macrophages respectively. The phenotypes of macrophages are very different from the previous studies. In general, Trem2hi macrophages are the most abundant population in healthy mice, while the proportion of monocytes increases after 11 weeks of HFD. Most importantly, inflammatory and resident-like macrophages make up 70% of the macrophage populations after 20 weeks of HFD. These strongly indicate that inflammatory and resident-like macrophages promote the progression of atherosclerosis plaques.
Collapse
Affiliation(s)
- Hao Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingxin Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenyun Zeng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huhu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lin Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Lu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Gao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
29
|
Suto MJ, Gupta V, Mathew B, Zhang W, Pallero MA, Murphy-Ullrich JE. Identification of Inhibitors of Thrombospondin 1 Activation of TGF-β. ACS Med Chem Lett 2020; 11:1130-1136. [PMID: 32550992 DOI: 10.1021/acsmedchemlett.9b00540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
TGF-β has been a target of interest for the treatment of fibrotic diseases and certain cancers. Approaches to target TGF-β include antagonists of the active ligand or TGF-β receptor kinase activity. These approaches have failed in clinical trials due to a lack of effectiveness and a limited therapeutic window. In this context, newer and more selective approaches to target TGF-β are needed. We previously reported that the matricellular protein, thrombospondin 1, activates the latent TGF-β complex and that antagonism of this pathway using tri/tetrapeptides in various animal models reduces fibrosis. The tripeptide, SRI-31277 (1), is effective in vivo but has a short plasma half life (0.2 h). Herein we describe the design and synthesis SRI-31277 analogs, specifically smaller peptides that retain potency and have improved bioavailability. We identified SRI-35241 (36) with a single chiral center, which blocks TGF-β activation (pIC50 = 8.12 nM) and has a plasma half life of 1.8 h (iv).
Collapse
Affiliation(s)
- Mark J. Suto
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Vandana Gupta
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Bini Mathew
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Wei Zhang
- Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, Alabama 35205, United States
| | - Manuel A. Pallero
- Department of Pathology, University of Alabama at Birmingham, VH G001A, 1720 Second Avenue South, Birmingham, Alabama 35294, United States
| | - Joanne E. Murphy-Ullrich
- Department of Pathology, University of Alabama at Birmingham, VH G001A, 1720 Second Avenue South, Birmingham, Alabama 35294, United States
| |
Collapse
|
30
|
Marino S, Petrusca DN, Roodman GD. Therapeutic targets in myeloma bone disease. Br J Pharmacol 2020; 178:1907-1922. [PMID: 31647573 DOI: 10.1111/bph.14889] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy and is characterized by a clonal proliferation of neoplastic plasma cells within the bone marrow. MM is the most frequent cancer involving the skeleton, causing osteolytic lesions, bone pain and pathological fractures that dramatically decrease MM patients' quality of life and survival. MM bone disease (MBD) results from uncoupling of bone remodelling in which excessive bone resorption is not compensated by new bone formation, due to a persistent suppression of osteoblast activity. Current management of MBD includes antiresorptive agents, bisphosphonates and denosumab, that are only partially effective due to their inability to repair the existing lesions. Thus, research into agents that prevent bone destruction and more importantly repair existing lesions by inducing new bone formation is essential. This review discusses the mechanisms regulating the uncoupled bone remodelling in MM and summarizes current advances in the treatment of MBD. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-related bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Medicine, Division Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Daniela N Petrusca
- Department of Medicine, Division Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - G David Roodman
- Department of Medicine, Division Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Roudebush VA Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
31
|
Green AC, Lath D, Hudson K, Walkley B, Down JM, Owen R, Evans HR, Paton-Hough J, Reilly GC, Lawson MA, Chantry AD. TGFβ Inhibition Stimulates Collagen Maturation to Enhance Bone Repair and Fracture Resistance in a Murine Myeloma Model. J Bone Miner Res 2019; 34:2311-2326. [PMID: 31442332 DOI: 10.1002/jbmr.3859] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/09/2019] [Accepted: 08/17/2019] [Indexed: 12/12/2022]
Abstract
Multiple myeloma is a plasma cell malignancy that causes debilitating bone disease and fractures, in which TGFβ plays a central role. Current treatments do not repair existing damage and fractures remain a common occurrence. We developed a novel low tumor phase murine model mimicking the plateau phase in patients as we hypothesized this would be an ideal time to treat with a bone anabolic. Using in vivo μCT we show substantial and rapid bone lesion repair (and prevention) driven by SD-208 (TGFβ receptor I kinase inhibitor) and chemotherapy (bortezomib and lenalidomide) in mice with human U266-GFP-luc myeloma. We discovered that lesion repair occurred via an intramembranous fracture repair-like mechanism and that SD-208 enhanced collagen matrix maturation to significantly improve fracture resistance. Lesion healing was associated with VEGFA expression in woven bone, reduced osteocyte-derived PTHrP, increased osteoblasts, decreased osteoclasts, and lower serum tartrate-resistant acid phosphatase 5b (TRACP-5b). SD-208 also completely prevented bone lesion development in mice with aggressive JJN3 tumors, and was more effective than an anti-TGFβ neutralizing antibody (1D11). We also discovered that SD-208 promoted osteoblastic differentiation (and overcame the TGFβ-induced block in osteoblastogenesis) in myeloma patient bone marrow stromal cells in vitro, comparable to normal donors. The improved bone quality and fracture-resistance with SD-208 provides incentive for clinical translation to improve myeloma patient quality of life by reducing fracture risk and fatality. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Alanna C Green
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Darren Lath
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Katie Hudson
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Brant Walkley
- Department of Materials Science and Engineering, The University of Sheffield, Sheffield, UK
| | - Jennifer M Down
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Robert Owen
- INSIGNEO Institute of In Silico Medicine, Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
| | - Holly R Evans
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Julia Paton-Hough
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Gwendolen C Reilly
- INSIGNEO Institute of In Silico Medicine, Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
| | - Michelle A Lawson
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Andrew D Chantry
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK.,Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| |
Collapse
|
32
|
Rebolledo DL, González D, Faundez-Contreras J, Contreras O, Vio CP, Murphy-Ullrich JE, Lipson KE, Brandan E. Denervation-induced skeletal muscle fibrosis is mediated by CTGF/CCN2 independently of TGF-β. Matrix Biol 2019; 82:20-37. [DOI: 10.1016/j.matbio.2019.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
|
33
|
Calvi LM, Hofbauer LC. A Novel Strategy for Repairing Multiple Myeloma Bone Lesions: Lessons From Murine Models. J Bone Miner Res 2019; 34:781-782. [PMID: 31071234 DOI: 10.1002/jbmr.3723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Laura M Calvi
- Division of Endocrinology and Metabolism, Department of Medicine and Wilmot Cancer Institute, University of Rochester School of Medicine, Rochester, NY, USA
| | - Lorenz C Hofbauer
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
34
|
Paton-Hough J, Tazzyman S, Evans H, Lath D, Down JM, Green AC, Snowden JA, Chantry AD, Lawson MA. Preventing and Repairing Myeloma Bone Disease by Combining Conventional Antiresorptive Treatment With a Bone Anabolic Agent in Murine Models. J Bone Miner Res 2019; 34:783-796. [PMID: 30320927 PMCID: PMC6607020 DOI: 10.1002/jbmr.3606] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/27/2018] [Accepted: 10/06/2018] [Indexed: 12/14/2022]
Abstract
Multiple myeloma is a plasma cell malignancy, which develops in the bone marrow and frequently leads to severe bone destruction. Current antiresorptive therapies to treat the bone disease do little to repair damaged bone; therefore, new treatment strategies incorporating bone anabolic therapies are urgently required. We hypothesized that combination therapy using the standard of care antiresorptive zoledronic acid (Zol) with a bone anabolic (anti-TGFβ/1D11) would be more effective at treating myeloma-induced bone disease than Zol therapy alone. JJN3 myeloma-bearing mice (n = 8/group) treated with combined Zol and 1D11 resulted in a 48% increase (p ≤ 0.001) in trabecular bone volume (BV/TV) compared with Zol alone and a 65% increase (p ≤ 0.0001) compared with 1D11 alone. Our most significant finding was the substantial repair of U266-induced osteolytic bone lesions with combination therapy (n = 8/group), which resulted in a significant reduction in lesion area compared with vehicle (p ≤ 0.01) or Zol alone (p ≤ 0.01). These results demonstrate that combined antiresorptive and bone anabolic therapy is significantly more effective at preventing myeloma-induced bone disease than Zol alone. Furthermore, we demonstrate that combined therapy is able to repair established myelomatous bone lesions. This is a highly translational strategy that could significantly improve bone outcomes and quality of life for patients with myeloma. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Julia Paton-Hough
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Simon Tazzyman
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Holly Evans
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Darren Lath
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Jenny M Down
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - Alanna C Green
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Andrew D Chantry
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK.,Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Michelle A Lawson
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK.,Mellanby Centre for Bone Research, University of Sheffield Medical School, University of Sheffield, Sheffield, UK
| |
Collapse
|
35
|
Green AC, Rudolph-Stringer V, Chantry AD, Wu JY, Purton LE. Mesenchymal lineage cells and their importance in B lymphocyte niches. Bone 2019; 119:42-56. [PMID: 29183783 PMCID: PMC11488667 DOI: 10.1016/j.bone.2017.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 02/06/2023]
Abstract
Early B lymphopoiesis occurs in the bone marrow and is reliant on interactions with numerous cell types in the bone marrow microenvironment, particularly those of the mesenchymal lineage. Each cellular niche that supports the distinct stages of B lymphopoiesis is unique. Different cell types and signaling molecules are important for the progressive stages of B lymphocyte differentiation. Cells expressing CXCL12 and IL-7 have long been recognized as having essential roles in facilitating progression through stages of B lymphopoiesis. Recently, a number of other factors that extrinsically mediate B lymphopoiesis (positively or negatively) have been identified. In addition, the use of transgenic mouse models to delete specific genes in mesenchymal lineage cells has further contributed to our understanding of how B lymphopoiesis is regulated in the bone marrow. This review will cover the current understanding of B lymphocyte niches in the bone marrow and key extrinsic molecules and signaling pathways involved in these niches, with a focus on how mesenchymal lineage cells regulate B lymphopoiesis.
Collapse
Affiliation(s)
- Alanna C Green
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia; Sheffield Myeloma Research Team, Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK; The Mellanby Centre for Bone Research, Sheffield, UK.
| | - Victoria Rudolph-Stringer
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Andrew D Chantry
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK; The Mellanby Centre for Bone Research, Sheffield, UK
| | - Joy Y Wu
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Louise E Purton
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia.
| |
Collapse
|
36
|
Wang Q, Ju X, Wang J, Fan Y, Ren M, Zhang H. Immunogenic cell death in anticancer chemotherapy and its impact on clinical studies. Cancer Lett 2018; 438:17-23. [PMID: 30217563 DOI: 10.1016/j.canlet.2018.08.028] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/15/2018] [Accepted: 08/30/2018] [Indexed: 01/10/2023]
Abstract
The traditional view holds that apoptosis is non-immunogenic and does not induce an inflammatory response. However, recent studies have suggested that certain chemotherapeutic drugs that induce tumor cell apoptosis can induce immunogenic cell death (ICD) in cancer cells. This process is characterized by not only up-regulation of a series of signaling molecules in cancer cells, including expose of calreticulin (CRT), secretion of adenosine triphosphate (ATP) and release of high mobility group box 1 (HMGB1). In this review, we summarize recent progress in identifying and classifying ICD inducers; concepts and molecular mechanisms of ICD; and the impact and potential applications of ICD in clinical studies. We also discuss the contributions of ICD inducers in combination with other anticancer drugs in clinical applications.
Collapse
Affiliation(s)
- Qiang Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Xiaoli Ju
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiayou Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Yu Fan
- Department of Molecular Biology and Translational Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Meijia Ren
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Heng Zhang
- Department of General Surgery, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China.
| |
Collapse
|
37
|
Prete A, Lo AS, Sadow PM, Bhasin SS, Antonello ZA, Vodopivec DM, Ullas S, Sims JN, Clohessy J, Dvorak AM, Sciuto T, Bhasin M, Murphy-Ullrich JE, Lawler J, Karumanchi SA, Nucera C. Pericytes Elicit Resistance to Vemurafenib and Sorafenib Therapy in Thyroid Carcinoma via the TSP-1/TGFβ1 Axis. Clin Cancer Res 2018; 24:6078-6097. [PMID: 30076136 DOI: 10.1158/1078-0432.ccr-18-0693] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/27/2018] [Accepted: 07/30/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE The BRAFV600E oncogene modulates the papillary thyroid carcinoma (PTC) microenvironment, in which pericytes are critical regulators of tyrosine-kinase (TK)-dependent signaling pathways. Although BRAFV600E and TK inhibitors are available, their efficacy as bimodal therapeutic agents in BRAFV600E-PTC is still unknown. EXPERIMENTAL DESIGN We assessed the effects of vemurafenib (BRAFV600E inhibitor) and sorafenib (TKI) as single agents or in combination in BRAFWT/V600E-PTC and BRAFWT/WT cells using cell-autonomous, pericyte coculture, and an orthotopic mouse model. We also used BRAFWT/V600E-PTC and BRAFWT/WT-PTC clinical samples to identify differentially expressed genes fundamental to tumor microenvironment. RESULTS Combined therapy blocks tumor cell proliferation, increases cell death, and decreases motility via BRAFV600E inhibition in thyroid tumor cells in vitro. Vemurafenib produces cytostatic effects in orthotopic tumors, whereas combined therapy (likely reflecting sorafenib activity) generates biological fluctuations with tumor inhibition alternating with tumor growth. We demonstrate that pericytes secrete TSP-1 and TGFβ1, and induce the rebound of pERK1/2, pAKT and pSMAD3 levels to overcome the inhibitory effects of the targeted therapy in PTC cells. This leads to increased BRAFV600E-PTC cell survival and cell death refractoriness. We find that BRAFWT/V600E-PTC clinical samples are enriched in pericytes, and TSP1 and TGFβ1 expression evoke gene-regulatory networks and pathways (TGFβ signaling, metastasis, tumor growth, tumor microenvironment/ECM remodeling functions, inflammation, VEGF ligand-VEGF receptor interactions, immune modulation, etc.) in the microenvironment essential for BRAFWT/V600E-PTC cell survival. Critically, antagonism of the TSP-1/TGFβ1 axis reduces tumor cell growth and overcomes drug resistance. CONCLUSIONS Pericytes shield BRAFV600E-PTC cells from targeted therapy via TSP-1 and TGFβ1, suggesting this axis as a new therapeutic target for overcoming resistance to BRAFV600E and TK inhibitors.
Collapse
Affiliation(s)
- Alessandro Prete
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Agnes S Lo
- Department of Medicine, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Swati S Bhasin
- Bioinformatic and Systems Biology Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Zeus A Antonello
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Danica M Vodopivec
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Soumya Ullas
- Longwood Small Animal Imaging Facility (LSAIF), Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jennifer N Sims
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - John Clohessy
- Division of Cancer Genetics, Department of Medicine, Beth Israel Deaconess Medical School, Harvard Medical School, Boston, Massachusetts
| | - Ann M Dvorak
- Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Tracey Sciuto
- Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Manoj Bhasin
- Bioinformatic and Systems Biology Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Joanne E Murphy-Ullrich
- Departments of Pathology, Cell Developmental and Integrative Biology, and Ophthalmology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jack Lawler
- Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - S Ananth Karumanchi
- Department of Medicine, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Carmelo Nucera
- Laboratory of Human Thyroid Cancers Preclinical and Translational Research, Division of Experimental Pathology, Cancer Research Institute (CRI), Cancer Center, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts. .,Department of Pathology, Center for Vascular Biology Research (CVBR), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|
38
|
Murphy-Ullrich JE, Suto MJ. Thrombospondin-1 regulation of latent TGF-β activation: A therapeutic target for fibrotic disease. Matrix Biol 2018; 68-69:28-43. [PMID: 29288716 PMCID: PMC6015530 DOI: 10.1016/j.matbio.2017.12.009] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/14/2017] [Accepted: 12/16/2017] [Indexed: 12/12/2022]
Abstract
Transforming growth factor-β (TGF-β) is a central player in fibrotic disease. Clinical trials with global inhibitors of TGF-β have been disappointing, suggesting that a more targeted approach is warranted. Conversion of the latent precursor to the biologically active form of TGF-β represents a novel approach to selectively modulating TGF-β in disease, as mechanisms employed to activate latent TGF-β are typically cell, tissue, and/or disease specific. In this review, we will discuss the role of the matricellular protein, thrombospondin 1 (TSP-1), in regulation of latent TGF-β activation and the use of an antagonist of TSP-1 mediated TGF-β activation in a number of diverse fibrotic diseases. In particular, we will discuss the TSP-1/TGF-β pathway in fibrotic complications of diabetes, liver fibrosis, and in multiple myeloma. We will also discuss emerging evidence for a role for TSP-1 in arterial remodeling, biomechanical modulation of TGF-β activity, and in immune dysfunction. As TSP-1 expression is upregulated by factors induced in fibrotic disease, targeting the TSP-1/TGF-β pathway potentially represents a more selective approach to controlling TGF-β activity in disease.
Collapse
Affiliation(s)
- Joanne E Murphy-Ullrich
- Departments of Pathology, Cell Developmental and Integrative Biology, and Ophthalmology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, United States.
| | - Mark J Suto
- Southern Research, 2000 Ninth Avenue South, Birmingham, AL 35205, United States
| |
Collapse
|
39
|
Saba F, Soleimani M, Abroun S. New role of hypoxia in pathophysiology of multiple myeloma through miR-210. EXCLI JOURNAL 2018; 17:647-662. [PMID: 30108468 PMCID: PMC6088223 DOI: 10.17179/excli2018-1109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/29/2018] [Indexed: 12/31/2022]
Abstract
Bone is one of the most common sites of complication in multiple myeloma (MM) progression and bone remodeling gets definitively perturbed during disease progression. Hypoxia and miR-210 play an important role in hematological malignancies. In an attempt to elucidate the specificity of the pathways of hypoxia and miR-210 in suppression of osteoblastic differentiation in MM patients, we examined the effect of miR-210 and hypoxia on expression of important cytokines and genes of myeloma cells. Differentiation of BM-MSCs towards osteoblastic cells in response to microvesicles (MVs) was also investigated. Finally, we proposed a molecular model on how HIF-1α may promote bone lesions in MM patients. To validate the effect of miR-210 and HIF-1α on targeted genes, the shRNA of HIF-1α and off-hsa-miR-210 were transfected into RPMI-8226 cells. BM-MSCs were cultured in osteoblastic inducer and 50 µg/mL of MVs derived from both hypoxic and normoxic myeloma cells. We designed an in vitro study to establish the effects of HIF-1α and miR-210 on the crosstalk between MM and osteoblasts. We here showed that hypoxia-induced miR-210 increased the mRNA expression of VLA-4, CXCR4, IL-6 and TGF-β in myeloma cells. MiR-210 is mandatory for the hypoxia-increased resistance of MM cells to melphalan. Moreover, MVs derived from hypoxic myeloma cells substantially decreased osteoblast differentiation. Considered comprehensively, our findings explain one of the reasons of bone loss that occurs at the sites of MM and a nascent crosstalk model in MM pathogenesis.
Collapse
Affiliation(s)
- Fakhredin Saba
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University,Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University,Tehran, Iran
| | - Saeid Abroun
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University,Tehran, Iran
| |
Collapse
|
40
|
Ring ES, Lawson MA, Snowden JA, Jolley I, Chantry AD. New agents in the Treatment of Myeloma Bone Disease. Calcif Tissue Int 2018; 102:196-209. [PMID: 29098361 PMCID: PMC5805798 DOI: 10.1007/s00223-017-0351-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/19/2017] [Indexed: 12/17/2022]
Abstract
Patients with multiple myeloma develop a devastating bone disease driven by the uncoupling of bone remodelling, excess osteoclastic bone resorption and diminished osteoblastic bone formation. The bone phenotype is typified by focal osteolytic lesions leading to pathological fractures, hypercalcaemia and other catastrophic bone events such as spinal cord compression. This causes bone pain, impaired functional status, decreased quality of life and increased mortality. Early in the disease, malignant plasma cells occupy a niche environment that encompasses their interaction with other key cellular components of the bone marrow microenvironment. Through these interactions, osteoclast-activating factors and osteoblast inhibitory factors are produced, which together uncouple the dynamic process of bone remodelling, leading to net bone loss and focal osteolytic lesions. Current management includes antiresorptive therapies, i.e. bisphosphonates, palliative support and orthopaedic interventions. Bisphosphonates are the mainstay of treatment for myeloma bone disease (MBD), but are only partially effective and do have some significant disadvantages; for example, they do not lead to the repair of existing bone destruction. Thus, newer agents to prevent bone destruction and also promote bone formation and repair existing lesions are warranted. This review summarises novel ways that MBD is being therapeutically targeted.
Collapse
Affiliation(s)
- Elizabeth S Ring
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, The University of Sheffield Medical School, Beech Hill Road, Sheffield, South Yorkshire, S10 2RX, UK.
| | - Michelle A Lawson
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Mellanby Bone Centre, School of Medicine and Biomedical Sciences, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Ingrid Jolley
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, The University of Sheffield Medical School, Beech Hill Road, Sheffield, South Yorkshire, S10 2RX, UK
- Department of Radiology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Andrew D Chantry
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, The University of Sheffield Medical School, Beech Hill Road, Sheffield, South Yorkshire, S10 2RX, UK
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Mellanby Bone Centre, School of Medicine and Biomedical Sciences, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
41
|
Yu H, Jiang N, Yu X, Zhao Z, Zhang X, Xu H. The role of TGFβ receptor 1-smad3 signaling in regulating the osteoclastic mode affected by fluoride. Toxicology 2017; 393:73-82. [PMID: 29127033 DOI: 10.1016/j.tox.2017.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/26/2022]
Abstract
Studies that have focused on the role TGFβ signaling plays in osteoclast activity are gradually increasing; however, literature is rare in terms of fluorosis. The aim of this study is to observe the role the TβR1/Smad3 pathway plays in fluoride regulating cellsosteoclast-like cells that are under the treatment of TGFβ receptor 1 kinase. The RANKL-mediated osteoclast-like cells from RAW264.7 cells were used as osteoclast precursor model. The profile of miRNA expression in fluoride-treated osteoclast-like cells exhibited 303 upregulated miRNAs, 61 downregulated miRNAs, and further drew 37 signaling pathway maps by KEGG and Biocarta pathway enrichment analysis. TGFβ and its downstream effectors were included among them. Osteoclast viability, formation and function were detected via MTT method, bone resorption pit and tartrate-resistant acid phosphatase (TRACP) staining, respectively. Results demonstrated that different doses of fluoride exhibited a biphasic effect on osteoclast cell viability, differentiation, formation and function. It indicated that a low dose of fluoride treatment stimulated them, but high dose inhibited them. SB431542 acted as TβR1 kinase inhibitor and blocked viability, formation and function of osteoclast-like cells regulated by fluoride. The expression of the osteoclast marker, RANK, and TβR1/Smad3 at gene and protein level was analyzed under fluoride with and without SB431542 treatment. Fluoride treatment indicated little effect on the RANK protein expression; however it significantly influenced TRACP expression in osteoclast-like cells. The stimulation of fluoride on the expression of Smad3 gene and phosphorylated Smad3 protein exhibited dose-dependent manner. SB431542 significantly impeded phosphorylation of Smad3 protein and TRACP expression in osteoclast-like cells that were exposed to fluoride. Our work demonstrated that TGFβ signaling played a key role in fluoride regulating osteoclast differentiation, formation and function. It elucidated that TβR1/Smad3 pathway participated in the mechanism of biphasic modulation of osteoclast mode regulated by fluoride.
Collapse
Affiliation(s)
- Haolan Yu
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Ningning Jiang
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - XiuHua Yu
- First Clinical Hospital, Jilin University, Changchun, 130021, People's Republic of China
| | - Zhitao Zhao
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Xiuyun Zhang
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Hui Xu
- Department of Regenerative Medical Science, School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
42
|
Sipes JM, Murphy-Ullrich JE, Roberts DD. Thrombospondins: Purification of human platelet thrombospondin-1. Methods Cell Biol 2017; 143:347-369. [PMID: 29310787 DOI: 10.1016/bs.mcb.2017.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Thrombospondins are a family of five secreted proteins that have diverse roles in modulating cellular function. Thrombospondins-1 and 2 were identified as matricellular proteins based on their functional roles combined with their transient appearance or accumulation in extracellular matrix at specific times during development and in response to injury or stress in mature tissues. Thrombospondin-1 is a major component of platelet α-granules, which provides a convenient source for purification of the protein. Methods are described to prepare thrombospondin-1 from human platelets in a biologically active form with minimal degradation or contamination with other platelet proteins. A nondenaturing method is described for removing bound transforming growth factor-β1.
Collapse
Affiliation(s)
- John M Sipes
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
43
|
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: 4.6] [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
|
44
|
Garg AD, More S, Rufo N, Mece O, Sassano ML, Agostinis P, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immunogenic cell death induction by anticancer chemotherapeutics. Oncoimmunology 2017; 6:e1386829. [PMID: 29209573 DOI: 10.1080/2162402x.2017.1386829] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022] Open
Abstract
The expression "immunogenic cell death" (ICD) refers to a functionally unique form of cell death that facilitates (instead of suppressing) a T cell-dependent immune response specific for dead cell-derived antigens. ICD critically relies on the activation of adaptive responses in dying cells, culminating with the exposure or secretion of immunostimulatory molecules commonly referred to as "damage-associated molecular patterns". Only a few agents can elicit bona fide ICD, including some clinically established chemotherapeutics such as doxorubicin, epirubicin, idarubicin, mitoxantrone, bleomycin, bortezomib, cyclophosphamide and oxaliplatin. In this Trial Watch, we discuss recent progress on the development of ICD-inducing chemotherapeutic regimens, focusing on studies that evaluate clinical efficacy in conjunction with immunological biomarkers.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Sanket More
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Nicole Rufo
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Odeta Mece
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Maria Livia Sassano
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
45
|
Terpos E, Christoulas D, Gavriatopoulou M, Dimopoulos MA. Mechanisms of bone destruction in multiple myeloma. Eur J Cancer Care (Engl) 2017; 26. [PMID: 28940410 DOI: 10.1111/ecc.12761] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2017] [Indexed: 02/02/2023]
Abstract
Osteolytic bone disease is a frequent complication of multiple myeloma, resulting in skeletal complications that are a significant cause of morbidity and mortality. It is the result of an increased activity of osteoclasts, which is not followed by reactive bone formation by osteoblasts. Recent studies have revealed novel molecules and pathways that are implicated in osteoclast activation and osteoblast inhibition. Among them, the most important include the receptor activator of nuclear factor-kappa B ligand/osteoprotegerin pathway, the macrophage inflammatory proteins and the activin-A that play a crucial role in osteoclast stimulation in myeloma, while the wingless-type (Wnt) signalling inhibitors (sclerostin and dickkopf-1) along with the growth factor independence-1 are considered the most important factors for the osteoblast dysfunction of myeloma patients. Finally, the role of osteocytes, which is the key cell for normal bone remodelling, has also revealed during the last years through their interaction with myeloma cells that leads to their apoptosis and the release of RANKL and sclerostin maintaining bone loss in these patients. This review focuses on the latest available data for the mechanisms of bone destruction in multiple myeloma.
Collapse
Affiliation(s)
- E Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra General Hospital, Athens, Greece
| | - D Christoulas
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra General Hospital, Athens, Greece
| | - M Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra General Hospital, Athens, Greece
| | - M A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra General Hospital, Athens, Greece
| |
Collapse
|
46
|
Heusschen R, Muller J, Duray E, Withofs N, Bolomsky A, Baron F, Beguin Y, Menu E, Ludwig H, Caers J. Molecular mechanisms, current management and next generation therapy in myeloma bone disease. Leuk Lymphoma 2017; 59:14-28. [PMID: 28573897 DOI: 10.1080/10428194.2017.1323272] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple myeloma (MM) bone disease is a major cause of morbidity and mortality in MM patients and persists even in patients in remission. This bone disease is caused by an uncoupling of bone remodeling, with increased osteoclast and decreased osteoblast activity and formation, culminating in lytic bone destruction. Bisphosphonates are the current standard of care but new therapies are needed. As the molecular mechanisms controlling MM bone disease are increasingly well understood, new therapeutic targets are extensively explored in the preclinical setting and initial clinical trials with novel compounds now show promising results. In this review, we will provide a comprehensive overview of the biology of MM bone disease, summarize its current clinical management and discuss preclinical and clinical data on next generation therapies.
Collapse
Affiliation(s)
- Roy Heusschen
- a Laboratory of Hematology , University of Liège, GIGA-I3 , Liège , Belgium
| | - Joséphine Muller
- a Laboratory of Hematology , University of Liège, GIGA-I3 , Liège , Belgium
| | - Elodie Duray
- a Laboratory of Hematology , University of Liège, GIGA-I3 , Liège , Belgium
| | - Nadia Withofs
- b Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics , University and CHU of Liège , Liège , Belgium
| | - Arnold Bolomsky
- c Wilhelminen Cancer Research Institute, Department of Medicine I , Center for Oncology and Hematology, Wilhelminenspital , Vienna , Austria
| | - Frédéric Baron
- a Laboratory of Hematology , University of Liège, GIGA-I3 , Liège , Belgium.,d Division of Hematology, Department of Medicine , University and CHU of Liège , Liège , Belgium
| | - Yves Beguin
- a Laboratory of Hematology , University of Liège, GIGA-I3 , Liège , Belgium.,d Division of Hematology, Department of Medicine , University and CHU of Liège , Liège , Belgium
| | - Eline Menu
- e Department of Hematology and Immunology , Myeloma Center Brussels, Vrije Universiteit Brussel , Brussels , Belgium
| | - Heinz Ludwig
- c Wilhelminen Cancer Research Institute, Department of Medicine I , Center for Oncology and Hematology, Wilhelminenspital , Vienna , Austria
| | - Jo Caers
- a Laboratory of Hematology , University of Liège, GIGA-I3 , Liège , Belgium.,d Division of Hematology, Department of Medicine , University and CHU of Liège , Liège , Belgium
| |
Collapse
|
47
|
Xi H, An R, Li L, Wang G, Tao Y, Gao L. Myeloma bone disease: Progress in pathogenesis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:149-155. [PMID: 27496181 DOI: 10.1016/j.pbiomolbio.2016.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/31/2016] [Accepted: 08/02/2016] [Indexed: 12/12/2022]
Abstract
Myeloma bone disease (MBD) is one of the most serious complications of multiple myeloma (MM) and the most severe cause of MM morbidity. Dysregulation of osteoblast and osteoclast cells plays key roles in MBD. In the bone marrow microenvironment, myeloma cells, osteoblasts, osteoclasts and bone marrow stromal cells can secrete multiple cytokines, categorized as osteoclast cell activating factors (OAFs) and osteoblast cell inactivating factors, which have been discovered to participate in bone metabolism and contribute to the pathogenesis of MBD. Several signaling pathways related to these cytokines were also revealed in the MBD pathogenesis. To better understand the pathogenesis of MBD and therefore the potential therapeutic targets of this disease, we will summarize recent study progress in the factors and underlying signaling pathways involved in the occurrence and development of MBD.
Collapse
Affiliation(s)
- Hao Xi
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Ran An
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Lu Li
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Gang Wang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Yi Tao
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Lu Gao
- Department of Physiology, Second Military Medical University, Shanghai, China.
| |
Collapse
|