1
|
Dutour A, Pasello M, Farrow L, Amer MH, Entz-Werlé N, Nathrath M, Scotlandi K, Mittnacht S, Gomez-Mascard A. Microenvironment matters: insights from the FOSTER consortium on microenvironment-driven approaches to osteosarcoma therapy. Cancer Metastasis Rev 2025; 44:44. [PMID: 40210800 PMCID: PMC11985652 DOI: 10.1007/s10555-025-10257-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Osteosarcoma (OS), a prevalent malignant bone tumor, has seen limited progress in treatment efficacy and patient outcomes over decades. Recent insights into the tumor microenvironment (TME) have revealed its crucial role in tumor progression and therapeutic resistance, particularly in OS. This review offers a comprehensive exploration of the OS microenvironment, meticulously dissecting its crucial components: the mesenchymal stromal TME, the immune microenvironment, hypoxia-induced adaptations, and the impact of the physical microenvironment. By demonstrating how these elements collectively drive tumor proliferation, immune evasion, and invasion, this review explores the intricate molecular and cellular dynamics at play. Furthermore, innovative approaches targeting the OS microenvironment, such as immunotherapies, are presented. This review highlights the importance of the TME in OS progression and its potential as a source of novel therapeutic strategies, offering new hope for improved patient outcomes.
Collapse
Affiliation(s)
- Aurelie Dutour
- Childhood Cancer & Cell Death Team, Centre de Recherche en Cancérologie de Lyon (CRCL), Centre Léon Bérard, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008, Lyon, France
| | - Michela Pasello
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luke Farrow
- University College London Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley St, London, WC1E 6DD, UK
| | - Mahetab H Amer
- Division of Cell Matrix & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Natacha Entz-Werlé
- Pediatric Onco-Hematology Unit, University Hospitals of Strasbourg, Strasbourg, France
- Translational, Transversal and Therapeutic Oncology Team, Laboratory of Bioimaging and Pathologies, Faculty of Pharmacy, CNRS UMR 7021, Illkirch, France
| | - Michaela Nathrath
- Department of Pediatric Hemato-Oncology, Psychosomatics and Systemic Diseases, Children's Hospital Kassel, Kassel, Germany
- Department of Pediatrics, Children'S Cancer Research Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sibylle Mittnacht
- University College London Cancer Institute, University College London, Paul O'Gorman Building, 72 Huntley St, London, WC1E 6DD, UK
| | - Anne Gomez-Mascard
- Department of Pathology, CHU, IUCT-Oncopole, University of Toulouse, Eq19. ONCOSARC CRCT, UMR 1037 Inserm/UT3, ERL 5294 CNRS, 1 Avenue Irène Joliot-Curie, 31059, Toulouse Cedex 9, France.
| |
Collapse
|
2
|
Chen Y, Chen QW, Fu FS, Gu HY, Yu A, Zhang XZ. Bone Destruction-Chemotactic Osteoprogenitor Cells Deliver Liposome Nanomedicines for the Treatment of Osteosarcoma and Osteoporosis. ACS NANO 2024; 18:29864-29879. [PMID: 39424791 DOI: 10.1021/acsnano.4c10053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Therapeutic efficacy of skeletal diseases is usually limited by unfavorable drug delivery due to incapable bone targeting and low bone affinity of conventional drug carriers, as well as relatively reduced vascularization and dense structure of bone tissues. Due to CXC chemokine receptor 4 (CXCR4)/CXC chemokine ligand 12 (CXCL12) signal axis-guided recruitment, osteoprogenitor cells (OPCs) can actively migrate to bone disease nidus. Here, drugs-loaded nanoliposomes are prepared and decorated onto OPCs by biotin-streptavidin linkage for precise bone disease targeting and effective drug delivery. In mouse models of tibia defect and orthotopic osteosarcoma, superior targeting property of OPCs-based drug delivery systems toward diseased bone niduses is verified. By encapsulating antitumor and antiosteoporosis drugs into nanoliposomes, OPCs-based drug delivery systems effectively inhibit disease development and restore bone destruction in mouse models of orthotopic osteosarcoma and ovariectomized osteoporosis. This study reveals a cell-based drug delivery system for precise bone disease targeting and highly effective drug delivery, which will find great potential as a universal drug delivery platform for targeting treatment of various bone diseases.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Fang-Sheng Fu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Hui-Yun Gu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
3
|
Akabane M, Imaoka Y, Kawashima J, Endo Y, Schenk A, Sasaki K, Pawlik TM. Innovative Strategies for Liver Transplantation: The Role of Mesenchymal Stem Cells and Their Cell-Free Derivatives. Cells 2024; 13:1604. [PMID: 39404368 PMCID: PMC11475694 DOI: 10.3390/cells13191604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Despite being the standard treatment for end-stage liver disease, liver transplantation has limitations like donor scarcity, high surgical costs, and immune rejection risks. Mesenchymal stem cells (MSCs) and their derivatives offer potential for liver regeneration and transplantation. MSCs, known for their multipotency, low immunogenicity, and ease of obtainability, can differentiate into hepatocyte-like cells and secrete bioactive factors that promote liver repair and reduce immune rejection. However, the clinical application of MSCs is limited by risks such as aberrant differentiation and low engraftment rates. As a safer alternative, MSC-derived secretomes and extracellular vesicles (EVs) offer promising therapeutic benefits, including enhanced graft survival, immunomodulation, and reduced ischemia-reperfusion injury. Current research highlights the efficacy of MSC-derived therapies in improving liver transplant outcomes, but further studies are necessary to standardize clinical applications. This review highlights the potential of MSCs and EVs to address key challenges in liver transplantation, paving the way for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.A.); (J.K.); (A.S.)
| | - Yuki Imaoka
- Division of Abdominal Transplant, Department of Surgery, Stanford University, Stanford, CA 94305, USA; (Y.I.); (K.S.)
| | - Jun Kawashima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.A.); (J.K.); (A.S.)
| | - Yutaka Endo
- Department of Transplant Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Austin Schenk
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.A.); (J.K.); (A.S.)
| | - Kazunari Sasaki
- Division of Abdominal Transplant, Department of Surgery, Stanford University, Stanford, CA 94305, USA; (Y.I.); (K.S.)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (M.A.); (J.K.); (A.S.)
| |
Collapse
|
4
|
Law YY, Lee HL, Lin CL, Chen PN, Wang PH, Hsieh YH, Chen CM. Asiatic acid inhibits osteosarcoma cell migration and invasion via the AKT/Sp1/MMP1 axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:3920-3929. [PMID: 38567545 DOI: 10.1002/tox.24246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
Osteosarcoma is a malignant bone tumor affecting adolescents and children. No effective treatment is currently available. Asiatic acid (AA), a triterpenoid compound found in Centella asiatica, possesses anti-tumor, anti-inflammatory, and anti-oxidant properties in various types of tumor cells. This study aims to determine whether AA exerts antitumor effects in human osteosarcoma cells. Our results indicate that AA does not influence the viability, proliferative rate, or cell cycle phase of human osteosarcoma cells under non-toxic conditions. AA suppressed osteosarcoma cell migration and invasion by down-regulating matrix metalloproteinase 1 (MMP1) expression. Data in the TNMplot database suggested MMP1 expression was higher in osteosarcoma than in normal tissues, with associated clinical significance observed in osteosarcoma patients. Overexpression of MMP1 in osteosarcoma cells reversed the AA-induced suppression of cell migration and invasion. AA treatment decreased the expression of specificity protein 1 (Sp1), while Sp1 overexpression abolished the effect of AA on MMP1 expression and cell migration and invasion. AA inhibited AKT phosphorylation, and treatment with a PI3K inhibitor (wortmannin) increased the anti-invasive effect of AA on osteosarcoma cells via the p-AKT/Sp1/MMP1 axis. Thus, AA exhibits the potential for use as an anticancer drug against human osteosarcoma.
Collapse
Affiliation(s)
- Yat-Yin Law
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chu-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Han Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Department of Leisure Industry Management, National Chin-Yi University of Technology, Taichung, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
5
|
Feng D, Wang Z, Cao S, Xu H, Li S. Identification of lipid metabolism-related gene signature in the bone marrow microenvironment of multiple myelomas through deep analysis of transcriptomic data. Clin Exp Med 2024; 24:136. [PMID: 38916672 PMCID: PMC11199273 DOI: 10.1007/s10238-024-01398-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
Dysregulated lipid metabolism in the bone marrow microenvironment (BMM) plays a vital role in multiple myeloma (MM) development, progression, and drug resistance. However, the exact mechanism by which lipid metabolism impacts the BMM, promotes tumorigenesis, and triggers drug resistance remains to be fully elucidated.By analyzing the bulk sequencing and single-cell sequencing data of MM patients, we identified lipid metabolism-related genes differential expression significantly associated with MM prognosis, referred to as LMRPgenes. Using a cohort of ten machine learning algorithms and 117 combinations, LMRPgenes predictive models were constructed. Further exploration of the effects of the model risk score (RS) on the survival status, immune status of patients with BMM, and response to immunotherapy was conducted. The study also facilitated the identification of personalized therapeutic strategies targeting specified risk categories within patient cohorts.Analysis of the scRNA-seq data revealed increased lipid metabolism-related gene enrichment scores (LMESs) in erythroblasts and progenitor, malignant, and Tprolif cells but decreased LMESs in lymphocytes. LMESs were also strongly correlated with most of the 50 hallmark pathways within these cell populations. An elevated malignant cell ratio and reduced lymphocytes were observed in the high LMES group. Moreover, the LMRPgenes predictive model, consisting of 14 genes, showed great predictive power. The risk score emerged as an independent indicator of poor outcomes. Inverse relationships between the RS and immune status were noted, and a high RS was associated with impaired immunotherapy responses. Drug sensitivity assays indicated the effectiveness of bortezomib, buparlisib, dinaciclib, staurosporine, rapamycin, and MST-312 in the high-RS group, suggesting their potential for treating patients with high-RS values and poor response to immunotherapy. Ultimately, upon verification via qRT-PCR, we observed a significant upregulation of ACBD6 in NDMM group compared to the control group.Our research enhances the knowledge base regarding the association between lipid metabolism-related genes (LMRGs) and the BMM in MM patients, offering substantive insights into the mechanistic effects of the BMM mediated by LMRGs.
Collapse
Affiliation(s)
- Dan Feng
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China
| | - Zhen Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China
| | - Shengji Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China
| | - Hui Xu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150000, Heilongjiang, China
| | - Shijun Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Liaoning, Dalian, 116011, China.
- College of Laboratory Medicine, Dalian Medical University, Liaoning, Dalian, 116044, China.
| |
Collapse
|
6
|
Martins-Neves SR, Sampaio-Ribeiro G, Gomes CMF. Self-Renewal and Pluripotency in Osteosarcoma Stem Cells' Chemoresistance: Notch, Hedgehog, and Wnt/β-Catenin Interplay with Embryonic Markers. Int J Mol Sci 2023; 24:8401. [PMID: 37176108 PMCID: PMC10179672 DOI: 10.3390/ijms24098401] [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: 03/31/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Osteosarcoma is a highly malignant bone tumor derived from mesenchymal cells that contains self-renewing cancer stem cells (CSCs), which are responsible for tumor progression and chemotherapy resistance. Understanding the signaling pathways that regulate CSC self-renewal and survival is crucial for developing effective therapies. The Notch, Hedgehog, and Wnt/β-Catenin developmental pathways, which are essential for self-renewal and differentiation of normal stem cells, have been identified as important regulators of osteosarcoma CSCs and also in the resistance to anticancer therapies. Targeting these pathways and their interactions with embryonic markers and the tumor microenvironment may be a promising therapeutic strategy to overcome chemoresistance and improve the prognosis for osteosarcoma patients. This review focuses on the role of Notch, Hedgehog, and Wnt/β-Catenin signaling in regulating CSC self-renewal, pluripotency, and chemoresistance, and their potential as targets for anti-cancer therapies. We also discuss the relevance of embryonic markers, including SOX-2, Oct-4, NANOG, and KLF4, in osteosarcoma CSCs and their association with the aforementioned signaling pathways in overcoming drug resistance.
Collapse
Affiliation(s)
- Sara R. Martins-Neves
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.R.M.-N.)
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Gabriela Sampaio-Ribeiro
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.R.M.-N.)
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
| | - Célia M. F. Gomes
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.R.M.-N.)
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
| |
Collapse
|
7
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Bradu P, Biswas A, Ganesan R, Renu K, Dey A, Vellingiri B, El Allali A, Alsamman AM, Zayed H, George Priya Doss C. Evolving strategies and application of proteins and peptide therapeutics in cancer treatment. Biomed Pharmacother 2023; 163:114832. [PMID: 37150032 DOI: 10.1016/j.biopha.2023.114832] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023] Open
Abstract
Several proteins and peptides have therapeutic potential and can be used for cancer therapy. By binding to cell surface receptors and other indicators uniquely linked with or overexpressed on tumors compared to healthy tissue, protein biologics enhance the active targeting of cancer cells, as opposed to the passive targeting of cells by conventional small-molecule chemotherapeutics. This study focuses on peptide medications that exist to slow or stop tumor growth and the spread of cancer, demonstrating the therapeutic potential of peptides in cancer treatment. As an alternative to standard chemotherapy, peptides that selectively kill cancer cells while sparing healthy tissue are developing. A mountain of clinical evidence supports the efficacy of peptide-based cancer vaccines. Since a single treatment technique may not be sufficient to produce favourable results in the fight against cancer, combination therapy is emerging as an effective option to generate synergistic benefits. One example of this new area is the use of anticancer peptides in combination with nonpeptidic cytotoxic drugs or the combination of immunotherapy with conventional therapies like radiation and chemotherapy. This review focuses on the different natural and synthetic peptides obtained and researched. Discoveries, manufacture, and modifications of peptide drugs, as well as their contemporary applications, are summarized in this review. We also discuss the benefits and difficulties of potential advances in therapeutic peptides.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| | - Pragya Bradu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Antara Biswas
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, South Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077 Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco.
| | - Alsamman M Alsamman
- Department of Genome Mapping, Molecular Genetics, and Genome Mapping Laboratory, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - C George Priya Doss
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
8
|
Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma. Biomolecules 2023; 13:biom13020398. [PMID: 36830767 PMCID: PMC9953423 DOI: 10.3390/biom13020398] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a cellular process that has been linked to the promotion of aggressive cellular features in many cancer types. It is characterized by the loss of the epithelial cell phenotype and a shift to a more mesenchymal phenotype and is accompanied by an associated change in cell markers. EMT is highly complex and regulated via multiple signaling pathways. While the importance of EMT is classically described for carcinomas-cancers of epithelial origin-it has also been clearly demonstrated in non-epithelial cancers, including osteosarcoma (OS), a primary bone cancer predominantly affecting children and young adults. Recent studies examining EMT in OS have highlighted regulatory roles for multiple proteins, non-coding nucleic acids, and components of the tumor micro-environment. This review serves to summarize these experimental findings, identify key families of regulatory molecules, and identify potential therapeutic targets specific to the EMT process in OS.
Collapse
|
9
|
Sarnella A, Ferrara Y, Albanese S, Omodei D, Cerchia L, De Simone G, Supuran CT, Zannetti A. Inhibition of Bone Marrow-Mesenchymal Stem Cell-Induced Carbonic Anhydrase IX Potentiates Chemotherapy Efficacy in Triple-Negative Breast Cancer Cells. Cells 2023; 12:cells12020298. [PMID: 36672233 PMCID: PMC9857137 DOI: 10.3390/cells12020298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Conventional chemotherapy represents the main systemic treatment used for triple-negative breast cancer (TNBC) patients, although many of them develop drug resistance. The hypoxic TME is the crucial driver in the onset of insensitivity to chemotherapy. In this research, we elucidated the role played by bone marrow-derived mesenchymal stem cells (BM-MSCs) in reducing cisplatin effects in TNBC. BT-549 and MDA-MB-231 cells, grown under hypoxic conditions in the presence of conditioned medium obtained from BM-MSCs (CM-MSCs), showed a strong cisplatin insensitivity and increased expression levels of carbonic anhydrase IX (CA IX). Therefore, we inhibited CM-MSC-induced CA IX by SLC-0111 to potentiate chemotherapy efficacy in TNBC cells. Our results showed that CM-MSCs under hypoxic conditions caused an increase in the ability of TNBC cells to form vascular structures, migrate and invade Matrigel. Cell treatment with cisplatin plus SLC-0111 was able to block these mechanisms, as well as the signaling pathways underlying them, such as p-AKT, p-ERK, CD44, MMP-2, vimentin, β-catenin, and N-cadherin, more effectively than treatment with single agents. In addition, a significant enhancement of apoptosis assessed by annexin V, caspase-3 expression and activity was also shown. Taken together, our results demonstrated the possibility, through CA IX inhibition, of returning TNBC cells to a more chemosensitive state.
Collapse
Affiliation(s)
| | - Ylenia Ferrara
- Institute of Biostructures and Bioimaging, CNR, 80145 Naples, Italy
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, CNR, 80145 Naples, Italy
| | - Daniela Omodei
- Institute of Biostructures and Bioimaging, CNR, 80145 Naples, Italy
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, CNR, 80131 Naples, Italy
| | | | | | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, CNR, 80145 Naples, Italy
- Correspondence: ; Tel.: +39-081-220-3431
| |
Collapse
|
10
|
The multifaceted roles of peptides in “always-on” near-infrared fluorescent probes for tumor imaging. Bioorg Chem 2022; 129:106182. [DOI: 10.1016/j.bioorg.2022.106182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 11/20/2022]
|
11
|
Targeting CXCR4 and CD47 Receptors: An Overview of New and Old Molecules for a Biological Personalized Anticancer Therapy. Int J Mol Sci 2022; 23:ijms232012499. [PMID: 36293358 PMCID: PMC9604048 DOI: 10.3390/ijms232012499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Biological therapy, with its multifaceted applications, has revolutionized the treatment of tumors, mainly due to its ability to exclusively target cancer cells and reduce the adverse effects on normal tissues. This review focuses on the therapies targeting the CXCR4 and CD47 receptors. We surveyed the results of early clinical trials testing compounds classified as nonpeptides, small peptides, CXCR4 antagonists or specific antibodies whose activity reduces or completely blocks the intracellular signaling pathways and cell proliferation. We then examined antibodies and fusion proteins against CD47, the receptor that acts as a “do not eat me” signal to phagocytes escaping immune surveillance. Despite these molecules being tested in early clinical trials, some drawbacks are emerging that impair their use in practice. Finally, we examined the ImmunoGenic Surrender mechanism that involves crosstalk and co-internalization of CXCR4 and CD47 upon engagement of CXCR4 by ligands or other molecules. The favorable effect of such compounds is dual as CD47 surface reduction impact on the immune response adds to the block of CXCR4 proliferative potential. These results suggest that a combination of different therapeutic approaches has more beneficial effects on patients’ survival and may pave the way for new accomplishments in personalized anticancer therapy.
Collapse
|
12
|
Guo Y, Wang M, Zou Y, Jin L, Zhao Z, Liu Q, Wang S, Li J. Mechanisms of chemotherapeutic resistance and the application of targeted nanoparticles for enhanced chemotherapy in colorectal cancer. J Nanobiotechnology 2022; 20:371. [PMID: 35953863 PMCID: PMC9367166 DOI: 10.1186/s12951-022-01586-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Colorectal cancer is considered one of the major malignancies that threaten the lives and health of people around the world. Patients with CRC are prone to post-operative local recurrence or metastasis, and some patients are advanced at the time of diagnosis and have no chance for complete surgical resection. These factors make chemotherapy an indispensable and important tool in treating CRC. However, the complex composition of the tumor microenvironment and the interaction of cellular and interstitial components constitute a tumor tissue with high cell density, dense extracellular matrix, and high osmotic pressure, inevitably preventing chemotherapeutic drugs from entering and acting on tumor cells. As a result, a novel drug carrier system with targeted nanoparticles has been applied to tumor therapy. It can change the physicochemical properties of drugs, facilitate the crossing of drug molecules through physiological and pathological tissue barriers, and increase the local concentration of nanomedicines at lesion sites. In addition to improving drug efficacy, targeted nanoparticles also reduce side effects, enabling safer and more effective disease diagnosis and treatment and improving bioavailability. In this review, we discuss the mechanisms by which infiltrating cells and other stromal components of the tumor microenvironment comprise barriers to chemotherapy in colorectal cancer. The research and application of targeted nanoparticles in CRC treatment are also classified.
Collapse
Affiliation(s)
- Yu Guo
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Min Wang
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Yongbo Zou
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Longhai Jin
- Department of Radiology, Jilin University Second Hospital, Changchun, 130000, China
| | - Zeyun Zhao
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Qi Liu
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Shuang Wang
- Department of the Dermatology, Jilin University Second Hospital, Changchun, 130000, China.
| | - Jiannan Li
- Department of the General Surgery, Jilin University Second Hospital, Changchun, 130000, China.
| |
Collapse
|
13
|
Bone mesenchymal stem cell-derived extracellular vesicles containing NORAD promote osteosarcoma by miR-30c-5p. J Transl Med 2022; 102:826-837. [PMID: 35332261 DOI: 10.1038/s41374-021-00691-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 11/08/2022] Open
Abstract
Osteosarcoma is a bone tumor that often affects children, adolescents and young people. Non-coding RNA activated by DNA damage (NORAD) can promote the proliferation of cancer cells in multiple tumors. Thus, the current study set out to explore the role of NORAD derived from extracellular vesicles (EVs) of bone mesenchymal stem cells (BMSCs) in osteosarcoma. First, NORAD was highly expressed in osteosarcoma cells and tissues, which might be associated with the progression and metastasis of osteosarcoma. We isolated EVs from the characterized BMSCs, and found that NORAD was transferred from BMSCs to osteosarcoma cells via EVs in the co-culture system. Consequently, NORAD delivered by BMSC-derived EVs promoted the proliferation and invasion of osteosarcoma cells. Subsequently, bioinformatics analyses suggested potential binding relationship between NORAD and microRNA-30c-5p (miR-30c-5p) as well as between miR-30c-5p and Krueppel-like factor 10 (KLF10), and the results of which were further verified by dual luciferase reporter gene assay, RNA immunoprecipitation, and RNA pull-down assay. Mechanistically, NORAD acted as a sponge of miR-30c-5p and up-regulated the expression of KLF10 where miR-30-c-5p mimic declined the effect induced by NORAD on cancer cells. The osteosarcoma cells were injected into mice to develop tumor growth and metastasis models. In these two models, injection of BMSC-EVs elevated NORAD expression and KLF10 but reduced miR-30c-5p expression, whereby suppressing tumor growth and lung metastasis. To conclude, BMSC-EVs deliver NORAD to osteosarcoma cells to regulate the miR-30c-5p/KLF10 axis, thereby accelerating the progression and metastasis of osteosarcoma.
Collapse
|
14
|
Sarnella A, Ferrara Y, Auletta L, Albanese S, Cerchia L, Alterio V, De Simone G, Supuran CT, Zannetti A. Inhibition of carbonic anhydrases IX/XII by SLC-0111 boosts cisplatin effects in hampering head and neck squamous carcinoma cell growth and invasion. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:122. [PMID: 35365193 PMCID: PMC8976345 DOI: 10.1186/s13046-022-02345-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023]
Abstract
Background Hypoxic tumor microenvironment (TME) contributes to the onset of many aspects of the cancer biology associated to the resistance to conventional therapies. Hypoxia is a common characteristic and negative prognostic factor in the head and neck squamous carcinomas (HNSCC) and is correlated with aggressive and invasive phenotype as well as with failure to chemo- and radio-therapies. The carbonic anhydrase isoenzymes IX and XII (CA IX/XII), regulators of extra and intracellular pH, are overexpressed in TME and are involved in adaptative changes occurring in cancer cells to survive at low O2. In this study, we aim to investigate in HNSCC cells and murine models the possibility to target CA IX/XII by the specific inhibitor SLC-0111 to potentiate the effects of cisplatin in hampering cell growth, migration and invasion. Furthermore, we analyzed the signal pathways cooperating in acquisition of a more aggressive phenotype including stemness, epithelial-mesenchymal transition and apoptotic markers. Methods The effects of cisplatin, CA IX/XII specific inhibitor SLC-0111, and the combinatorial treatment were tested on proliferation, migration, invasion of HNSCC cells grown in 2D and 3D models. Main signal pathways and the expression of stemness, mesenchymal and apoptotic markers were analyzed by western blotting. Molecular imaging using NIR-Annexin V and NIR-Prosense was performed in HNSCC xenografts to detect tumor growth and metastatic spread. Results HNSCC cells grown in 2D and 3D models under hypoxic conditions showed increased levels of CA IX/XII and greater resistance to cisplatin than cells grown under normoxic conditions. The addition of CA IX/XII inhibitor SLC-0111 to cisplatin sensitized HNSCC cells to the chemotherapeutic agent and caused a reduction of proliferation, migration and invasiveness. Furthermore, the combination therapy hampered activation of STAT3, AKT, ERK, and EMT program, whereas it induced apoptosis. In HNSCC xenografts the treatment with cisplatin plus SLC-0111 caused an inhibition of tumor growth and an induction of apoptosis as well as a reduction of metastatic spread at a higher extent than single agents. Conclusion Our results highlight the ability of SLC-0111 to sensitize HNSCC to cisplatin by hindering hypoxia-induced signaling network that are shared among mechanisms involved in therapy resistance and metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02345-x.
Collapse
Affiliation(s)
- Annachiara Sarnella
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy
| | - Ylenia Ferrara
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy
| | - Luigi Auletta
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", CNR, Via S. Pansini 5, 80131, Naples, Italy
| | - Vincenzo Alterio
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy
| | - Giuseppina De Simone
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff, 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging-CNR, Via T. De Amicis, 95, 80145, Naples, Italy.
| |
Collapse
|
15
|
Shao H, Ge M, Zhang J, Zhao T, Zhang S. Osteoclasts differential-related prognostic biomarker for osteosarcoma based on single cell, bulk cell and gene expression datasets. BMC Cancer 2022; 22:288. [PMID: 35300639 PMCID: PMC8932072 DOI: 10.1186/s12885-022-09380-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma (OS) is one of the most common primary bone malignant tumors. Osteoclasts have been shown to have a valuable role in OS. In the present study, we analyzed the differentiation states of osteoclasts in OS and their prognostic significance based on integrated scRNA-seq and bulk RNA-seq data. Osteoclasts in distinct differentiation states were characterized, and 661 osteoclasts differentiation-related genes (ODRGs) were obtained. ORDGs in distinct differentiation states were enriched in distinct functions and pathways. TPM1, S100A13, LOXL1, PSMD10, ST3GAL4, PEF1, SERPINE2, TUBB, FAM207A, TUBA1A, and DCN were identified as the significant survival-predicting ODRGs. We successfully developed a risk score model based on these survival-predicting ODRGs. In addition, we generated a nomogram applicable for clinical with both ODRGs signatures and clinicopathological parameters, and validated in OS cohorts to predict OS patient outcome. This study proposed and verified the important roles of osteoclasts differentiation in the prognosis of patients with OS, suggesting promising therapeutic targets for OS.
Collapse
Affiliation(s)
- Haiyu Shao
- Department of Orthopaedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158#, Hangzhou, 310014, Zhejiang, China
| | - Meng Ge
- Department of Orthopaedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158#, Hangzhou, 310014, Zhejiang, China.,Department of Orthopaedics, Bengbu Medical College, Bengbu, Anhui, China
| | - Jun Zhang
- Department of Orthopaedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158#, Hangzhou, 310014, Zhejiang, China
| | - Tingxiao Zhao
- Department of Orthopaedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158#, Hangzhou, 310014, Zhejiang, China
| | - Shuijun Zhang
- Department of Orthopaedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Shangtang Road 158#, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
16
|
Wei B, Feng H, Wu H. Reduced CCR2 Can Improve the Prognosis of Sarcoma by Remodeling the Tumor Microenvironment. Int J Gen Med 2022; 15:3043-3053. [PMID: 35308572 PMCID: PMC8932926 DOI: 10.2147/ijgm.s349295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Background Aim Methods and Results Conclusion
Collapse
Affiliation(s)
- Baixing Wei
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Hao Feng
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Han Wu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
- Correspondence: Han Wu, Tel +86 043189876902, Email
| |
Collapse
|
17
|
Vicinanza C, Lombardi E, Da Ros F, Marangon M, Durante C, Mazzucato M, Agostini F. Modified mesenchymal stem cells in cancer therapy: A smart weapon requiring upgrades for wider clinical applications. World J Stem Cells 2022; 14:54-75. [PMID: 35126828 PMCID: PMC8788179 DOI: 10.4252/wjsc.v14.i1.54] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/06/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem stromal cells (MSC) are characterized by the intriguing capacity to home toward cancer cells after systemic administration. Thus, MSC can be harnessed as targeted delivery vehicles of cytotoxic agents against tumors. In cancer patients, MSC based advanced cellular therapies were shown to be safe but their clinical efficacy was limited. Indeed, the amount of systemically infused MSC actually homing to human cancer masses is insufficient to reduce tumor growth. Moreover, induction of an unequivocal anticancer cytotoxic phenotype in expanded MSC is necessary to achieve significant therapeutic efficacy. Ex vivo cell modifications are, thus, required to improve anti-cancer properties of MSC. MSC based cellular therapy products must be handled in compliance with good manufacturing practice (GMP) guidelines. In the present review we include MSC-improving manipulation approaches that, even though actually tested at preclinical level, could be compatible with GMP guidelines. In particular, we describe possible approaches to improve MSC homing on cancer, including genetic engineering, membrane modification and cytokine priming. Similarly, we discuss appropriate modalities aimed at inducing a marked cytotoxic phenotype in expanded MSC by direct chemotherapeutic drug loading or by genetic methods. In conclusion, we suggest that, to configure MSC as a powerful weapon against cancer, combinations of clinical grade compatible modification protocols that are currently selected, should be introduced in the final product. Highly standardized cancer clinical trials are required to test the efficacy of ameliorated MSC based cell therapies.
Collapse
Affiliation(s)
- Carla Vicinanza
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| | - Elisabetta Lombardi
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| | - Francesco Da Ros
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| | - Miriam Marangon
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| | - Cristina Durante
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| | - Mario Mazzucato
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| | - Francesco Agostini
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano 33081, Italy
| |
Collapse
|
18
|
Zhang X, Li N, Zhu Y, Wen W. The role of mesenchymal stem cells in the occurrence, development, and therapy of hepatocellular carcinoma. Cancer Med 2022; 11:931-943. [PMID: 34981659 PMCID: PMC8855904 DOI: 10.1002/cam4.4521] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver malignant tumor, with high recurrence and mortality rates. Mesenchymal stem cells (MSCs) are multipotent cells that can be recruited into the tumor microenvironment (TME). What is known, TME plays a vital part in tumor progression. In recent years, accumulating studies have found that MSCs have a dual role of promotion and inhibition in the occurrence and development of HCC. In this review, we analyzed the role of MSCs in TME and summarized the relationship between MSCs and liver cancer stem cells, the molecular signaling pathway mechanisms of MSCs promoting and inhibiting HCC, and the latest research progress of MSCs in the treatment of HCC.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Na Li
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ying Zhu
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Wei Wen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
19
|
Rosner M, Hengstschläger M. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:26-34. [PMID: 35641164 PMCID: PMC8895487 DOI: 10.1093/stcltm/szab003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/12/2021] [Indexed: 12/03/2022] Open
Abstract
It is the hope of clinicians and patients alike that stem cell-based therapeutic products will increasingly become applicable remedies for many diseases and injuries. Whereas some multipotent stem cells are already routinely used in regenerative medicine, the efficacious and safe clinical translation of pluripotent stem cells is still hampered by their inherent immunogenicity and tumorigenicity. In addition, stem cells harbor the paracrine potential to affect the behavior of cells in their microenvironment. On the one hand, this property can mediate advantageous supportive effects on the overall therapeutic concept. However, in the last years, it became evident that both, multipotent and pluripotent stem cells, are capable of inducing adjacent cells to become motile. Not only in the context of tumor development but generally, deregulated mobilization and uncontrolled navigation of patient’s cells can have deleterious consequences for the therapeutic outcome. A more comprehensive understanding of this ubiquitous stem cell feature could allow its proper clinical handling and could thereby constitute an important building block for the further development of safe therapies.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
- Corresponding author: Markus Hengstschläger, PhD, Professor, Institute of Medical Genetics, Medical University of Vienna, Währinger Strasse 10, 1090 Vienna, Austria. Tel: +43 1 40160 56500; Fax: +43 1 40160 956501;
| |
Collapse
|
20
|
Xu C, Wang M, Zandieh-Doulabi B, Sun W, Wei L, Liu Y. To B (Bone Morphogenic Protein-2) or Not to B (Bone Morphogenic Protein-2): Mesenchymal Stem Cells May Explain the Protein's Role in Osteosarcomagenesis. Front Cell Dev Biol 2021; 9:740783. [PMID: 34869325 PMCID: PMC8635864 DOI: 10.3389/fcell.2021.740783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS), a primary malignant bone tumor, stems from bone marrow-derived mesenchymal stem cells (BMSCs) and/or committed osteoblast precursors. Distant metastases, in particular pulmonary and skeletal metastases, are common in patients with OS. Moreover, extensive resection of the primary tumor and bone metastases usually leads to bone defects in these patients. Bone morphogenic protein-2 (BMP-2) has been widely applied in bone regeneration with the rationale that BMP-2 promotes osteoblastic differentiation of BMSCs. Thus, BMP-2 might be useful after OS resection to repair bone defects. However, the potential tumorigenicity of BMP-2 remains a concern that has impeded the administration of BMP-2 in patients with OS and in populations susceptible to OS with severe bone deficiency (e.g., in patients with genetic mutation diseases and aberrant activities of bone metabolism). In fact, some studies have drawn the opposite conclusion about the effect of BMP-2 on OS progression. Given the roles of BMSCs in the origination of OS and osteogenesis, we hypothesized that the responses of BMSCs to BMP-2 in the tumor milieu may be responsible for OS development. This review focuses on the relationship among BMSCs, BMP-2, and OS cells; a better understanding of this relationship may elucidate the accurate mechanisms of actions of BMP-2 in osteosarcomagenesis and thereby pave the way for clinically safer and broader administration of BMP-2 in the future. For example, a low dosage of and a slow-release delivery strategy for BMP-2 are potential topics for exploration to treat OS.
Collapse
Affiliation(s)
- Chunfeng Xu
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mingjie Wang
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Behrouz Zandieh-Doulabi
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wei Sun
- Department of Mechanical Engineering, Drexel University, Philadelphia, PA, United States.,Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Lingfei Wei
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Oral Implantology, Yantai Stomatological Hospital, Yantai, China
| | - Yuelian Liu
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
21
|
Monteiro CF, Custódio CA, Mano JF. Bioengineering a humanized 3D tri-culture osteosarcoma model to assess tumor invasiveness and therapy response. Acta Biomater 2021; 134:204-214. [PMID: 34303015 DOI: 10.1016/j.actbio.2021.07.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/03/2023]
Abstract
To date, anticancer therapies with evidenced efficacy in preclinical models fail during clinical trials. The shortage of robust drug screening platforms that accurately predict patient's response underlie these misleading results. To provide a reliable platform for tumor drug discovery, we herein propose a relevant humanized 3D osteosarcoma (OS) model exploring the potential of methacryloyl platelet lysates (PLMA)-based hydrogels to sustain spheroid growth and invasion. The architecture and synergistic cell-microenvironment interaction of an invading tumor was recapitulated encapsulating spheroids in PLMA hydrogels, alone or co-cultured with osteoblasts and mesenchymal stem cells. The stem cells alignment toward OS spheroid suggested that tumor cells chemotactically attracted the surrounding stromal cells, which supported tumor growth and invasion into the hydrogels. The exposure of established models to doxorubicin revealed an improved drug resistance of PLMA-based models, comparing with scaffold-free spheroids. The proposed OS models highlighted the feasibility of PLMA hydrogels to support tumor invasion and recapitulate tumor-stromal cell crosstalk, demonstrating the potential of this 3D platform for complex tumor modelling. STATEMENT OF SIGNIFICANCE: Cell invasion mechanisms involved in tumor progression have been recapitulated in the field of 3D in vitro modeling, leveraging the great advance in biomimetic materials. In line with the growing interest in human-derived biomaterials, the aim of this study is to explore for the first time the potential of methacryloyl platelet lysates (PLMA)-based hydrogels to develop a humanized 3D osteosarcoma model to assess tumor invasiveness and drug sensitivity. By co-culturing tumor spheroids with human osteoblasts and human mesenchymal stem cells, this study demonstrated the importance of the synergistic tumor cell-microenvironment interaction in tumor growth, invasion and drug resistance. The established 3D osteosarcoma model highlighted the feasibility of PLMA hydrogels as a relevant 3D platform for complex tumor modelling.
Collapse
Affiliation(s)
- Cátia F Monteiro
- CICECO - Aveiro Institute of Materials, University of Aveiro, Department of Chemistry, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Catarina A Custódio
- CICECO - Aveiro Institute of Materials, University of Aveiro, Department of Chemistry, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - João F Mano
- CICECO - Aveiro Institute of Materials, University of Aveiro, Department of Chemistry, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
22
|
Xing L, Lv HT, Liu SG, Wang WB, Zhang TF, Liu JH, Bian W. The effect of gemcitabine combined with AMD3100 applying to cholangiocarcinoma RBE cell lines to CXCR4/CXCL12 axis. Scand J Gastroenterol 2021; 56:914-919. [PMID: 34165373 DOI: 10.1080/00365521.2021.1906944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE To evaluate the effect of AMD3100 treatment to cholangiocarcinoma by analyzing the relationship between them, and provide experimental evidence for whether AMD3100 can become a clinical treatment drug for cholangiocarcinoma. MATERIALS AND METHODS Cholangiocarcinoma RBE cell lines were used in this study. MTT cell proliferation test was used for evaluating the effect of gemcitabine and AMD3100 to cell. CXCR4, N-cadherin, VEGF-C and MMP-9 were detect by RT-PCR and western. Transwell was used for evaluating the invasion effect. RESULTS We demonstrated that as the concentration of gemcitabine increasing from 0.33, 3.33 to 33.33 uM, the cell survival rate was 76.65%, 71.40%, 52.25%, respectively. RT-PCR and Western blot that gemcitabine could affect the expression of CXCR4 protein and the level of mRNA transcription in a dose-dependent manner. N-cadherin VEGF-C, MMP-9 mRNA transcription level showed a significant upward trend in gemcitabine group. In Transwell test, the number of cells in the gemcitabine group was significantly higher than that in the no-medication group (p < .05), the AMD3100 group and the combination group of gemcitabine and AMD3100, the difference between the no-medication group and the AMD3100 monotherapy group was not significant, and the combination group was between them. CONCLUSIONS This study showed that gemcitabine significantly inhibited the growth of cholangiocarcinoma RBE cells in a dose-dependent manner, and gemcitabine can affect the expression of CXCR4, N-cadherin, VEGF-C, MMP-9 protein and mRNA. Cell invasion and metastasis-related factors decreased after AMD3100 combined with gemcitabine.
Collapse
Affiliation(s)
- Li Xing
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Hai-Tao Lv
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - San-Guang Liu
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Wen-Bin Wang
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Teng-Fei Zhang
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Jian-Hua Liu
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Wei Bian
- Department of Hepatobiliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
23
|
Hassanzadeh A, Shamlou S, Yousefi N, Nikoo M, Verdi J. Genetically-Modified Stem Cell in Regenerative Medicine and Cancer Therapy; A New Era. Curr Gene Ther 2021; 22:23-39. [PMID: 34238158 DOI: 10.2174/1566523221666210707125342] [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] [Received: 02/04/2021] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 11/22/2022]
Abstract
Recently, genetic engineering by various strategies to stimulate gene expression in a specific and controllable mode is a speedily growing therapeutic approach. Genetic modification of human stem or progenitor cells, such as embryonic stem cells (ESCs), neural progenitor cells (NPCs), mesenchymal stem/stromal cells (MSCs), and hematopoietic stem cells (HSCs) for direct delivery of specific therapeutic molecules or genes has been evidenced as an opportune plan in the context of regenerative medicine due to their supported viability, proliferative features, and metabolic qualities. On the other hand, a large number of studies have investigated the efficacy of modified stem cells in cancer therapy using cells from various sources, disparate transfection means for gene delivery, different transfected yields, and wide variability of tumor models. Accordingly, cell-based gene therapy holds substantial aptitude for the treatment of human malignancy as it could relieve signs or even cure cancer succeeding expression of therapeutic or suicide transgene products; however, there exist inconsistent results in this regard. Herein, we deliver a brief overview of stem cell potential to use in cancer therapy and regenerative medicine and importantly discuss stem cells based gene delivery competencies to stimulate tissue repair and replacement in concomitant with their potential to use as an anti-cancer therapeutic strategy, focusing on the last two decades in vivo studies.
Collapse
Affiliation(s)
- Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Shamlou
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Marzieh Nikoo
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Yang Y, Zhao Y, Zhang L, Zhang F, Li L. The Application of Mesenchymal Stem Cells in the Treatment of Liver Diseases: Mechanism, Efficacy, and Safety Issues. Front Med (Lausanne) 2021; 8:655268. [PMID: 34136500 PMCID: PMC8200416 DOI: 10.3389/fmed.2021.655268] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation is a novel treatment for liver diseases due to the roles of MSCs in regeneration, fibrosis inhibition and immune regulation. However, the mechanisms are still not completely understood. Despite the significant efficacy of MSC therapy in animal models and preliminary clinical trials, issues remain. The efficacy and safety of MSC-based therapy in the treatment of liver diseases remains a challenging issue that requires more investigation. This article reviews recent studies on the mechanisms of MSCs in liver diseases and the associated challenges and suggests potential future applications.
Collapse
Affiliation(s)
- Ya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yalei Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Chen H, Li G, Liu Y, Ji S, Li Y, Xiang J, Zhou L, Gao H, Zhang W, Sun X, Fu X, Li B. Pleiotropic Roles of CXCR4 in Wound Repair and Regeneration. Front Immunol 2021; 12:668758. [PMID: 34122427 PMCID: PMC8194072 DOI: 10.3389/fimmu.2021.668758] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022] Open
Abstract
Wound healing is a multi-step process that includes multiple cellular events such as cell proliferation, cell adhesion, and chemotactic response as well as cell apoptosis. Accumulating studies have documented the significance of stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor 4 (CXCR4) signaling in wound repair and regeneration. However, the molecular mechanism of regeneration is not clear. This review describes various types of tissue regeneration that CXCR4 participates in and how the efficiency of regeneration is increased by CXCR4 overexpression. It emphasizes the pleiotropic effects of CXCR4 in regeneration. By delving into the specific molecular mechanisms of CXCR4, we hope to provide a theoretical basis for tissue engineering and future regenerative medicine.
Collapse
Affiliation(s)
- Huating Chen
- Department of Wound Repair Surgery, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | | | - Yiqiong Liu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuaifei Ji
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.,Department of Southern Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Jiangbing Xiang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.,Department of School of Biological Engineering, Chongqing University, Chongqing, China
| | - Laixian Zhou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Huanhuan Gao
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenwen Zhang
- Department of Wound Repair Surgery, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College; PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Binghui Li
- Department of Wound Repair Surgery, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, Huang YH, Ling TY. Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci 2021; 28:28. [PMID: 33849537 PMCID: PMC8043779 DOI: 10.1186/s12929-021-00725-7] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a promising resource for cell-based therapy because of their high immunomodulation ability, tropism towards inflamed and injured tissues, and their easy access and isolation. Currently, there are more than 1200 registered MSC clinical trials globally. However, a lack of standardized methods to characterize cell safety, efficacy, and biodistribution dramatically hinders the progress of MSC utility in clinical practice. In this review, we summarize the current state of MSC-based cell therapy, focusing on the systemic safety and biodistribution of MSCs. MSC-associated risks of tumor initiation and promotion and the underlying mechanisms of these risks are discussed. In addition, MSC biodistribution methodology and the pharmacokinetics and pharmacodynamics of cell therapies are addressed. Better understanding of the systemic safety and biodistribution of MSCs will facilitate future clinical applications of precision medicine using stem cells.
Collapse
Affiliation(s)
- Wei-Zhan Zhuang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yi-Heng Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, 10041, Taiwan.,Department of Obstetrics and Gynecology, National Taiwan University Hospital Yunlin Branch, Yunlin, 64041, Taiwan
| | - Long-Jyun Su
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan
| | - Meng-Shiue Wu
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Han-Yin Jeng
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.,TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Huan-Cheng Chang
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106, Taiwan.,Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan. .,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan. .,TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan. .,International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan. .,Comprehensive Cancer Center of Taipei Medical University, Taipei, 11031, Taiwan. .,The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
27
|
Li J, Pu K, Li C, Wang Y, Zhou Y. A Novel Six-Gene-Based Prognostic Model Predicts Survival and Clinical Risk Score for Gastric Cancer. Front Genet 2021; 12:615834. [PMID: 33692828 PMCID: PMC7938863 DOI: 10.3389/fgene.2021.615834] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/15/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Autophagy plays a vital role in cancer initiation, malignant progression, and resistance to treatment. However, autophagy-related genes (ARGs) have rarely been analyzed in gastric cancer (GC). The purpose of this study was to analyze ARGs in GC using bioinformatic analysis and to identify new biomarkers for predicting the overall survival (OS) of patients with GC. Methods: The gene expression profiles and clinical data of patients with GC were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets, and ARGs were obtained from two other datasets (the Human Autophagy Database and Molecular Signatures Database). Lasso, univariate, and multivariate Cox regression analyses were performed to identify the OS-related ARGs. Finally, a six-ARG model was identified as a prognostic indicator using the risk-score model, and survival and prognostic performance were analyzed based on the Kaplan-Meier test and ROC curve. Estimate calculations were used to assess the immune status of this model, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed for investigating the functions and terms associated with the model-related genes in GC. Results: The six ARGs, DYNLL1, PGK2, HPR, PLOD2, PHYHIP, and CXCR4, were identified using Lasso and Cox regression analyses. Survival analysis revealed that the OS of GC patients in the high-risk group was significantly lower than that of the low-risk group (p < 0.05). The ROC curves revealed that the risk score model exhibited better prognostic performance with respect to OS. Multivariate Cox regression analysis indicated that the model was an independent predictor of OS and was not affected by most of the clinical traits (p < 0.05). The model-related genes were associated with immune suppression and several biological process terms, such as extracellular structure organization and matrix organization. Moreover, the genes were associated with the P13K-Akt signaling pathway, focal adhesion, and MAPK signaling pathway. Conclusions: This study presents potential prognostic biomarkers for GC patients that would aid in determining the best patient-specific course of treatment.
Collapse
Affiliation(s)
- Juan Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, China
| | - Ke Pu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Chunmei Li
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
28
|
Prediction of disease-associated circRNAs via circRNA–disease pair graph and weighted nuclear norm minimization. Knowl Based Syst 2021. [DOI: 10.1016/j.knosys.2020.106694] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Portella L, Bello AM, Scala S. CXCL12 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:51-70. [PMID: 34286441 DOI: 10.1007/978-3-030-62658-7_5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor microenvironment (TME) is the local environment of tumor, composed of tumor cells and blood vessels, extracellular matrix (ECM), immune cells, and metabolic and signaling molecules. Chemokines and their receptors play a fundamental role in the crosstalk between tumor cells and TME, regulating tumor-related angiogenesis, specific leukocyte infiltration, and activation of the immune response and directly influencing tumor cell growth, invasion, and cancer progression. The chemokine CXCL12 is a homeostatic chemokine that regulates physiological and pathological process such as inflammation, cell proliferation, and specific migration. CXCL12 activates CXCR4 and CXCR7 chemokine receptors, and the entire axis has been shown to be dysregulated in more than 20 different tumors. CXCL12 binding to CXCR4 triggers multiple signal transduction pathways that regulate intracellular calcium flux, chemotaxis, transcription, and cell survival. CXCR7 binds with high-affinity CXCL12 and with lower-affinity CXCL11, which binds also CXCR3. Although CXCR7 acts as a CXCL12 scavenger through ligand internalization and degradation, it transduces the signal mainly through β-arrestin with a pivotal role in endothelial and neural cells. Recent studies demonstrate that TME rich in CXCL12 leads to resistance to immune checkpoint inhibitors (ICI) therapy and that CXCL12 axis inhibitors sensitize resistant tumors to ICI effect. Thus targeting the CXCL12-mediated axis may control tumor and tumor microenvironment exerting an antitumor dual action. Herein CXCL12 physiology, role in cancer biology and in composite TME, prognostic role, and the relative inhibitors are addressed.
Collapse
Affiliation(s)
- Luigi Portella
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Anna Maria Bello
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
30
|
Zannetti A, Benga G, Brunetti A, Napolitano F, Avallone L, Pelagalli A. Role of Aquaporins in the Physiological Functions of Mesenchymal Stem Cells. Cells 2020; 9:2678. [PMID: 33322145 PMCID: PMC7763964 DOI: 10.3390/cells9122678] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Aquaporins (AQPs) are a family of membrane water channel proteins that control osmotically-driven water transport across cell membranes. Recent studies have focused on the assessment of fluid flux regulation in relation to the biological processes that maintain mesenchymal stem cell (MSC) physiology. In particular, AQPs seem to regulate MSC proliferation through rapid regulation of the cell volume. Furthermore, several reports have shown that AQPs play a crucial role in modulating MSC attachment to the extracellular matrix, their spread, and migration. Shedding light on how AQPs are able to regulate MSC physiological functions can increase our knowledge of their biological behaviours and improve their application in regenerative and reparative medicine.
Collapse
Affiliation(s)
- Antonella Zannetti
- Institute of Biostructure and Bioimaging, CNR, Via T. De Amicis 95, 80145 Naples, Italy;
| | - Gheorghe Benga
- Romanian Academy, Cluj-Napoca Branch, Strada Republicii 9, 400015 Cluj-Napoca, Romania;
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples Federico II, via Pansini 5, 80131 Naples, Italy;
| | - Francesco Napolitano
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, via Veterinaria 1, 80137 Naples, Italy; (F.N.); (L.A.)
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, via Veterinaria 1, 80137 Naples, Italy; (F.N.); (L.A.)
| | - Alessandra Pelagalli
- Institute of Biostructure and Bioimaging, CNR, Via T. De Amicis 95, 80145 Naples, Italy;
- Department of Advanced Biomedical Sciences, University of Naples Federico II, via Pansini 5, 80131 Naples, Italy;
| |
Collapse
|
31
|
Lei Y, Junxin C, Yongcan H, Xiaoguang L, Binsheng Y. Role of microRNAs in the crosstalk between osteosarcoma cells and the tumour microenvironment. J Bone Oncol 2020; 25:100322. [PMID: 33083216 PMCID: PMC7554654 DOI: 10.1016/j.jbo.2020.100322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumour, with a peak incidence in adolescents, and the five-year survival rate of patients with metastasis or recurrence is much lower than that of patients without metastasis and recurrence. OS is initiated and develops in a complex tumour microenvironment (TME) that contains many different components, such as osteoblasts, osteoclasts, mesenchymal stem cells, fibroblasts, immune cells, extracellular matrix (ECM), extracellular vesicles, and cytokines. The extensive interaction between OS and the TME underlies OS progression. Therefore, rather than targeting OS cells, targeting the key factors in the TME may yield novel therapeutic approaches. MicroRNAs (miRNAs) play multiple roles in the biological behaviours of OS, and recent studies have implied that miRNAs are involved in mediating the communication between OS cells and the surrounding TME. Here, we review the TME landscape and the miRNA dysregulation of OS, describe the role of the altered TME in OS development and highlight the role of miRNA in the crosstalk between OS cells and the TME.
Collapse
Affiliation(s)
- Yong Lei
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chen Junxin
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Huang Yongcan
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Liu Xiaoguang
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - Yu Binsheng
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
32
|
Liu Q, Gu J, Zhang E, He L, Yuan ZX. Targeted Delivery of Therapeutics to Urological Cancer Stem Cells. Curr Pharm Des 2020; 26:2038-2056. [PMID: 32250210 DOI: 10.2174/1381612826666200403131514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Urological cancer refers to cancer in organs of the urinary system and the male reproductive system. It mainly includes prostate cancer, bladder cancer, renal cancer, etc., seriously threatening patients' survival. Although there are many advances in the treatment of urological cancer, approved targeted therapies often result in tumor recurrence and therapy failure. An increasing amount of evidence indicated that cancer stem cells (CSCs) with tumor-initiating ability were the source of treatment failure in urological cancer. The development of CSCstargeted strategy can provide a possibility for the complete elimination of urological cancer. This review is based on a search of PubMed, Google scholar and NIH database (http://ClinicalTrials.gov/) for English language articles containing the terms: "biomarkers", "cancer stem cells", "targeting/targeted therapy", "prostate cancer", bladder cancer" and "kidney cancer". We summarized the biomarkers and stem cell features of the prostate, bladder and renal CSCs, outlined the targeted strategies for urological CSCs from signaling pathways, cytokines, angiogenesis, surface markers, elimination therapy, differentiation therapy, immunotherapy, microRNA, nanomedicine, etc., and highlighted the prospects and future challenges in this research field.
Collapse
Affiliation(s)
- Qiang Liu
- Yaopharma Co., Ltd. Chongqing, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, China
| | - E Zhang
- Officers college of PAP, Chengdu, Sichuan, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, China
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Sarnella A, D’Avino G, Hill BS, Alterio V, Winum JY, Supuran CT, De Simone G, Zannetti A. A Novel Inhibitor of Carbonic Anhydrases Prevents Hypoxia-Induced TNBC Cell Plasticity. Int J Mol Sci 2020; 21:ijms21218405. [PMID: 33182416 PMCID: PMC7664880 DOI: 10.3390/ijms21218405] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Cell plasticity is the ability that cells have to modify their phenotype, adapting to the environment. Cancer progression is under the strict control of the the tumor microenvironment that strongly determines its success by regulating the behavioral changes of tumor cells. The cross-talk between cancer and stromal cells and the interactions with the extracellular matrix, hypoxia and acidosis contribute to trigger a new tumor cell identity and to enhance tumor heterogeneity and metastatic spread. In highly aggressive triple-negative breast cancer, tumor cells show a significant capability to change their phenotype under the pressure of the hypoxic microenvironment. In this study, we investigated whether targeting the hypoxia-induced protein carbonic anhydrase IX (CA IX) could reduce triple-negative breast cancer (TNBC) cell phenotypic switching involved in processes associated with poor prognosis such as vascular mimicry (VM) and cancer stem cells (CSCs). The treatment of two TNBC cell lines (BT-549 and MDA-MB-231) with a specific CA IX siRNA or with a novel inhibitor of carbonic anhydrases (RC44) severely impaired their ability to form a vascular-like network and mammospheres and reduced their metastatic potential. In addition, the RC44 inhibitor was able to hamper the signal pathways involved in triggering VM and CSC formation. These results demonstrate that targeting hypoxia-induced cell plasticity through CA IX inhibition could be a new opportunity to selectively reduce VM and CSCs, thus improving the efficiency of existing therapies in TNBC.
Collapse
Affiliation(s)
- Annachiara Sarnella
- CNR Istituto di Biostrutture e Bioimmagini, 80122 Napoli, Italy; (A.S.); (G.D.); (B.S.H.); (V.A.); (G.D.S.)
| | - Giuliana D’Avino
- CNR Istituto di Biostrutture e Bioimmagini, 80122 Napoli, Italy; (A.S.); (G.D.); (B.S.H.); (V.A.); (G.D.S.)
| | - Billy Samuel Hill
- CNR Istituto di Biostrutture e Bioimmagini, 80122 Napoli, Italy; (A.S.); (G.D.); (B.S.H.); (V.A.); (G.D.S.)
| | - Vincenzo Alterio
- CNR Istituto di Biostrutture e Bioimmagini, 80122 Napoli, Italy; (A.S.); (G.D.); (B.S.H.); (V.A.); (G.D.S.)
| | - Jean-Yves Winum
- IBMM, Universite Montpellier, CNRS, ENSCM, 34296 Montpellier, France;
| | - Claudiu T. Supuran
- Dipartimento NEUROFARBA, Sezione di Scienze Farmaceutiche, Università di Firenze, Sesto Fiorentino, 50139 Firenze, Italy;
| | - Giuseppina De Simone
- CNR Istituto di Biostrutture e Bioimmagini, 80122 Napoli, Italy; (A.S.); (G.D.); (B.S.H.); (V.A.); (G.D.S.)
| | - Antonella Zannetti
- CNR Istituto di Biostrutture e Bioimmagini, 80122 Napoli, Italy; (A.S.); (G.D.); (B.S.H.); (V.A.); (G.D.S.)
- Correspondence: ; Tel.: +39-3666115319
| |
Collapse
|
34
|
Lu M, Xie K, Lu X, Lu L, Shi Y, Tang Y. Notoginsenoside R1 counteracts mesenchymal stem cell-evoked oncogenesis and doxorubicin resistance in osteosarcoma cells by blocking IL-6 secretion-induced JAK2/STAT3 signaling. Invest New Drugs 2020; 39:416-425. [PMID: 33128383 DOI: 10.1007/s10637-020-01027-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022]
Abstract
Tumor microenvironment is a critical participant in the initiation, progression and drug resistance of carcinomas, including osteosarcoma. Notoginsenoside R1 (NGR1) is a proverbial active ingredient of the traditional Chinese medicine Panax notoginseng (PN) and possess undeniable roles in several cancers. Nevertheless, its function in osteosarcoma and tumor microenvironment remains elusive. In the current study, exposure to NGR1 dose-dependently inhibited osteosarcoma cell viability and migration, and induced apoptosis. Furthermore, osteosarcoma cells that were incubated with conditioned medium (CM) from bone marrow mesenchymal stem cells (BMSCs) exhibited greater proliferation, migration capacity and MMP-2 and MMP-9 expression relative to control cells, which was reversed when BMSCs were treated with NGR1. Notably, administration with NGR1 antagonized CM-evoked doxorubicin resistance in osteosarcoma cells by decreasing cell viability and increasing cell apoptosis and caspase-3/9 activity. Mechanically, NGR1 suppressed IL-6 secretion from BMSCs, as well as the subsequent activation of the JAK2/STAT3 signaling in osteosarcoma cells. In addition, blocking the JAK2 pathway by its antagonist AG490 reversed CM-induced osteosarcoma cell proliferation, migration and doxorubicin resistance. Moreover, exogenous supplementation with IL-6 engendered not only the reactivation of the JAK2/STAT3 signaling but also muted NGR1-mediated efficacy against osteosarcoma cell malignancy and doxorubicin resistance. Collectively, NGR1 may directly restrain osteosarcoma cell growth and migration, or indirectly antagonize MSC-evoked malignancy and drug resistance by interdicting IL-6 secretion-evoked activation of the JAK2/STAT3 pathway. Consequently, the current study may highlight a promising therapeutic strategy against osteosarcoma by regulating tumor cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Minan Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Kegong Xie
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Xianzhe Lu
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Lu Lu
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Yu Shi
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Yujin Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China.
- Department of Orthopedic Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
35
|
Flavonoids Restore Platinum Drug Sensitivity to Ovarian Carcinoma Cells in a Phospho-ERK1/2-Dependent Fashion. Int J Mol Sci 2020; 21:ijms21186533. [PMID: 32906729 PMCID: PMC7555577 DOI: 10.3390/ijms21186533] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/23/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer (OC) is the second most common type of gynecological malignancy; it has poor survival rates and is frequently (>75%) diagnosed at an advanced stage. Platinum-based chemotherapy, with, e.g., carboplatin, is the standard of care for OC, but toxicity and acquired resistance to therapy have proven challenging. Despite advances in OC diagnosis and treatment, approximately 85% of patients will experience relapse, mainly due to chemoresistance. The latter is attributed to alterations in the cancer cells and is also mediated by tumor microenvironment (TME). Recently, we reported the synthesis of a platinum (IV) prodrug that exhibits equal potency toward platinum-sensitive and resistant OC cell lines. Here, we investigated the effect of TME on platinum sensitivity. Co-culture of OC cells with murine or human mesenchymal stem cells (MS-5 and HS-5, respectively) rendered them resistant to chemotherapeutic agents, including platinum, paclitaxel and colchicine. Platinum resistance was also conferred by co-culture with differentiated murine adipocyte progenitor cells. Exposure of OC cells to chemotherapeutic agents resulted in activation of phospho-ERK1/2. Co-culture with MS-5, which conferred drug resistance, was accompanied by blockage of phospho-ERK1/2 activation. The flavonoids fisetin and quercetin were active in restoring ERK phosphorylation, as well as sensitivity to platinum compounds. Exposure of OC cells to cobimetinib-a MEK1 inhibitor that also inhibits extracellular signal-regulated kinase (ERK) phosphorylation-which resulted in reduced sensitivity to the platinum compound. This suggests that ERK activity is involved in mediating the function of flavonoids in restoring platinum sensitivity to OC co-cultured with cellular components of the TME. Our data show the potential of combining flavonoids with standard therapy to restore drug sensitivity to OC cells and overcome TME-mediated platinum drug resistance.
Collapse
|
36
|
Niu J, Yan T, Guo W, Wang W, Zhao Z, Ren T, Huang Y, Zhang H, Yu Y, Liang X. Identification of Potential Therapeutic Targets and Immune Cell Infiltration Characteristics in Osteosarcoma Using Bioinformatics Strategy. Front Oncol 2020; 10:1628. [PMID: 32974202 PMCID: PMC7471873 DOI: 10.3389/fonc.2020.01628] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma is one of the most aggressive malignant bone tumors worldwide. Although great advancements have been made in its treatment owing to the advent of neoadjuvant chemotherapy, the problem of lung metastasis is a major obstacle in the improvement of survival outcomes. Thus, the aim of the present study is to screen novel and key biomarkers, which may act as potential prognostic markers and therapeutic targets in osteosarcoma. We utilized the robust rank aggregation (RRA) method to integrate three osteosarcoma microarray datasets downloaded from the Gene Expression Omnibus (GEO) database, and we identified the robust differentially expressed genes (DEGs) between primary and metastatic osteosarcoma tissues. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to explore the functions of robust DEGs. The results of enrichment analysis showed that the robust DEGs were closely associated with osteosarcoma development and progression. Immune cell infiltration analysis was also conducted by CIBERSORT algorithm, and we found that macrophages are the most principal infiltrating immune cells in osteosarcoma, especially macrophages M0 and M2. Then, the protein–protein interaction network and key modules were constructed by Cytoscape, and 10 hub genes were selected by plugin cytoHubba from the whole network. The survival analysis of hub genes was also carried out based on the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. The integrated bioinformatics analysis was utilized to provide new insight into osteosarcoma development and metastasis and identified EGR1, CXCL10, MYC, and CXCR4 as potential biomarkers for prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Jianfang Niu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Taiqiang Yan
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Wei Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Zhiqing Zhao
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Hongliang Zhang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yiyang Yu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Xin Liang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| |
Collapse
|
37
|
New CXCR4 Antagonist Peptide R (Pep R) Improves Standard Therapy in Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12071952. [PMID: 32708431 PMCID: PMC7409147 DOI: 10.3390/cancers12071952] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/18/2022] Open
Abstract
The chemokine receptor CXCR4 is overexpressed and functional in colorectal cancer. To investigate the role of CXCR4 antagonism in potentiating colon cancer standard therapy, the new peptide CXCR4 antagonist Peptide R (Pep R) was employed. Human colon cancer HCT116 xenograft-bearing mice were treated with chemotherapeutic agents (CT) 5-Fluorouracil (5FU) and oxaliplatin (OX) or 5FU and radio chemotherapy (RT-CT) in the presence of Pep R. After two weeks, CT plus Pep R reduced by 4-fold the relative tumor volume (RTV) as compared to 2- and 1.6-fold reductions induced, respectively, by CT and Pep R. In vitro Pep R addition to CT/RT-CT impaired HCT116 cell growth and further reduced HCT116 and HT29 clonal capability. Thus, the hypothesis that Pep R could target the epithelial mesenchyme transition (EMT) process was evaluated. While CT decreased ECAD and increased ZEB-1 and CD90 expression, the addition of Pep R restored the pretreatment expression. In HCT116 and HT29 cells, CT/RT-CT induced a population of CD133+CXCR4+ cells, supposedly a stem-resistant cancer cell population, while Pep R reduced it. Taken together, the results showed that targeting CXCR4 ameliorates the effect of treatment in colon cancer through inhibition of cell growth and reversal of EMT treatment-induced markers, supporting further clinical studies.
Collapse
|
38
|
Li Y, Wang H, Wang J, Sun B, Li L. Chemokine receptor 4 expression is correlated with the occurrence and prognosis of gastric cancer. FEBS Open Bio 2020; 10:1149-1161. [PMID: 32306562 PMCID: PMC7262922 DOI: 10.1002/2211-5463.12864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) is a common tumor with a low 5-year survival rate. The chemokine receptor 4 (CXCR4) protein contributes to the progression and prognosis of GC, but the relationship between CXCR4 and immune infiltration, somatic copy number alteration (SCNA), tumor purity, tumor mutation burden (TMB), cytolytic activity (CYT), and drug sensitivity in GC is poorly understood. This study aimed to systematically explore the role of CXCR4 in GC. Microarray and RNA-seq data were collected from the Gene Expression Omnibus and The Cancer Genome Atlas. Our analysis shows that CXCR4 is correlated with various types of immune cells. Patients with high CXCR4 expression had a higher fraction of B cells and CD8+ T cells, and a lower fraction of CD4+ T cells. In addition, high CXCR4 expression was associated with more advanced tumor stage, worse prognosis and higher stromal score, immune score, and cytolytic activity (P < 0.05). High CXCR4 expression also correlated with lower tumor purity and TMB. In summary, our analyses suggest that CXCR4 may affect the progression and prognosis of GC by influencing immune infiltration, TMB, CYT, tumor purity, and drug sensitivity.
Collapse
Affiliation(s)
- Yang Li
- Department of GastrointestinalShandong provincial hospital affiliated to Shandong universityJinanChina
| | - Hong‐Chang Wang
- Department of GastrointestinalShandong provincial hospital affiliated to Shandong universityJinanChina
| | - Jin‐Shen Wang
- Department of GastrointestinalShandong provincial hospital affiliated to Shandong universityJinanChina
| | - Bo Sun
- Department of GastrointestinalShandong provincial hospital affiliated to Shandong universityJinanChina
| | - Le‐Ping Li
- Department of GastrointestinalShandong provincial hospital affiliated to Shandong universityJinanChina
| |
Collapse
|
39
|
LI A, ZHANG T, GAO J. [Progress on utilizing mesenchymal stem cells as cellular delivery system for targeting delivery of as drug/gene for anti-tumor therapy]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020; 49:20-34. [PMID: 32621413 PMCID: PMC8800717 DOI: 10.3785/j.issn.1008-9292.2020.02.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/15/2020] [Indexed: 06/11/2023]
Abstract
Mesenchymal stem cells (MSCs) have the inherent tumor-homing ability with the attraction of multiple chemokines released by tumor tissues or tumor microenvironments, which can be utilized as promising cellular carriers for targeted delivery of anti-tumor drugs and genes. In most circumstances, large amount of systemicly administrated MSCs will be firstly trapped by lungs, following with re-distribution and homing to tumor tissues after lung clearance. Several approaches like enhanced interactions between chemokines and receptors on MSCs or reducing the retention of MSCs by changes of administration methods are firstly reviewed for improving the homing of MSCs towards tumor tissues. Additionally, the potentials and gains of utilizing MSCs to carry several chemotherapeutics, such as doxorubicin, paclitaxel and gemcitabine are summarized, showing the advantages of overcoming the short half-life and poor tumor targeting of these chemotherapeutics. Moreover, the applications of MSCs to protect and deliver therapeutic genes to tumor sites for selectively tumor cells eliminating or promoting immune system are highlighted. In addition, the potentials of using MSCs for tumor-targeting delivery of diagnostic and therapeutic agents are addressed. We believed that the continuous improvement and optimization of this stem cells-based cellular delivery system will provide a novel delivery strategy and option for tumor treatment.
Collapse
|
40
|
Chen H, Wahafu P, Wang L, Chen X. LncRNA LINC00313 Knockdown Inhibits Tumorigenesis and Metastasis in Human Osteosarcoma by Upregulating FOSL2 through Sponging miR-342-3p. Yonsei Med J 2020; 61:359-370. [PMID: 32390359 PMCID: PMC7214116 DOI: 10.3349/ymj.2020.61.5.359] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/31/2019] [Accepted: 12/09/2019] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Osteosarcoma (OS) is the most common primary bone tumor, with high morbidity in infants and adolescents. Long noncoding RNA LINC00313 has been found to modulate papillary thyroid cancer tumorigenesis and to be dysregulate in lung cancer. However, the role of LINC00313 in OS has not yet been addressed. MATERIALS AND METHODS We evaluated mRNA and protein expression using real-time quantitative PCR and Western blotting. Cell proliferation was evaluated using MTT; apoptosis and autophagy were assessed with flow cytometry, Western blotting, and/or GFP-LC3 assay. Transwell assay was conducted to measure cell migration and invasion. Potential target sites for LINC00313 and miR-342-3p were predicted with starBase v.2.0 and TargetScan Human, and verified using luciferase reporter assay, RNA immunoprecipitation, and RNA pull-down assay. In vivo, xenogeneic tumors were induced with U2OS and MG-63 cells, separately. RESULTS LINC00313 was upregulated and miR-342-3p was downregulated in OS tissues and cells. High expression of LINC00313 was associated with shorter overall survival. FOSL2 downregulation and miR-342-3p overexpression suppressed cell proliferation and migratory and invasive abilities while promoting apoptosis and autophagy, all of which were consistent with the effects of LINC00313 knockdown. miR-342-3p, sponged by LINC00313, inversely modulated FOSL2 by targeting MG-63 cells, and FOSL2 expression was positively controlled by LINC00313. LINC00313 knockdown suppressed tumor growth in vivo. CONCLUSION LINC00313 is upregulated in OS, and LINC00313 knockdown plays a vital anti-tumor role in OS cell progression through a miR-342-3p/FOSL2 axis. Our study suggests that LINC00313 may be a novel, promising biomarker for diagnosis and prognosis of OS.
Collapse
Affiliation(s)
- Hongtao Chen
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China.
| | - Paerhati Wahafu
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Leilei Wang
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Xuan Chen
- Department of Orthopedics, The Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| |
Collapse
|
41
|
Zhang S, Yang Y, Fan L, Zhang F, Li L. The clinical application of mesenchymal stem cells in liver disease: the current situation and potential future. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:565. [PMID: 32775366 PMCID: PMC7347776 DOI: 10.21037/atm.2020.03.218] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver disease is a major health issue which present poor clinical treatment performance. Cirrhosis and liver failure are common clinical manifestations of liver diseases. Liver transplantation is recognized as the ultimate and most efficient therapy to the end stage of liver disease. But it was limited by the shortage of honor organs and high cost. Nowadays, stem cell therapy gained more and more attention due to its attractive efficacy in treating liver disease especially in cirrhosis during the clinical trials. Mesenchymal stem cell (MSC) can be differentiated into hepatocytes, promote liver regeneration, inhibit liver fibrosis and induce liver apoptosis, particularly via paracrine mechanisms. This review will highlight recent clinical applications of MSC, providing the available evidence and discussing some unsolved questions in treating liver disease.
Collapse
Affiliation(s)
- Sainan Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Ya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Linxiao Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
42
|
Zhang L, Yang L, Xia ZW, Yang SC, Li WH, Liu B, Yu ZQ, Gong PF, Yang YL, Sun WZ, Mo J, Li GS, Wang TY, Wang K. The role of fibroblast activation protein in progression and development of osteosarcoma cells. Clin Exp Med 2020; 20:121-130. [PMID: 31745677 DOI: 10.1007/s10238-019-00591-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
Abstract
To investigate the expression levels of fibroblast activation protein (FAP) in human osteosarcoma tissues and its possible correlations with clinical pathological characteristics of patients with osteosarcoma, and to explore the potential effects of FAP on progression and development of osteosarcoma. Immunohistochemistry (IHC) assay was initially performed to detect the expression levels of FAP in 66 tumor tissues and adjacent non-tumor tissues. Patients were sequentially divided into two groups based on different expression levels of FAP. The correlations between the expression levels of FAP and the clinical pathological characteristics were investigated, and the role of FAP in proliferation, migration, and invasion of osteosarcoma cells was assessed via colony formation, MTT, wound healing, and transwell assays, respectively. The possible effects of FAP on tumor growth and metastasis were evaluated in vivo. We further attempted to reveal the underlying mechanism of FAP involved in tumor growth through bioinformatics and IHC assays. High expression levels of FAP were noted in human osteosarcoma tissues. It also was unveiled that FAP was significantly associated with the tumor size (P = 0.005*) and clinical stage (P = 0.017*). Our data further confirmed that knockdown of FAP remarkably blocked proliferation, migration, and invasion of osteosarcoma cells in vitro, and suppressed tumor growth and metastasis in mice via AKT signaling pathway. The possible role of FAP in progression and development of osteosarcoma could be figured out. Our data may be helpful to develop a novel therapeutic target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Li Yang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zi-Wei Xia
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Shi-Chang Yang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wen-Hui Li
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Bin Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zi-Qi Yu
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Peng-Fei Gong
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Ya-Lin Yang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wei-Zong Sun
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jing Mo
- Department of Pathology, Tianjin Medical University, Tianjin, 300070, China
| | - Gui-Shi Li
- Department of Joint Orthopedics, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong Province, China
| | - Tian-Yi Wang
- Department of Orthopedics, The 981st Hospital of the Chinese People's Liberation Army, Chengde, 067000, Hebei Province, China.
| | - Kai Wang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| |
Collapse
|
43
|
Long non-coding RNA PVT1 encapsulated in bone marrow mesenchymal stem cell-derived exosomes promotes osteosarcoma growth and metastasis by stabilizing ERG and sponging miR-183-5p. Aging (Albany NY) 2019; 11:9581-9596. [PMID: 31699956 PMCID: PMC6874467 DOI: 10.18632/aging.102406] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/26/2019] [Indexed: 12/21/2022]
Abstract
Exosomes secreted by bone marrow mesenchymal stem cells (BMSCs) promote osteosarcoma cell proliferation and migration, while the underlying mechanism remains unknown. Since the long non-coding RNA PVT1 has been reported to be upregulated in osteosarcoma cells and contributes to its growth and metastasis, we aim to investigate whether BMSC-derived exosomes promote osteosarcoma growth and metastasis via transporting PVT1 into osteosarcoma cells. The PVT1 expression in BMSC-derived exosomes was markedly higher than that in osteosarcoma cell-derived exosomes. The co-culturing of BMSC-derived exosomes and osteosarcoma cells (Saos-2, MG-63, and MNNG/HOS cell lines) significantly raised PVT1 expression of osteosarcoma cells. The direct binding between PVT1 and the oncogenic protein ERG was confirmed using RNA immunoprecipitation and RNA pull-down assays, and the transported PVT1 promotes osteosarcoma cell proliferation and migration via inhibiting degradation and ubiquitination of ERG. PVT1 also increased ERG expression through sponging miR-183-5p. In summary, our findings indicated that BMSC-derived exosomes encapsulate PVTl and transport it into osteosarcoma cells, and the transported PVT1 promotes tumor growth and metastasis by inhibiting ubiquitination and promoting expression of ERG in osteosarcoma cells. These data provide a novel insight into the mechanism of BMSC-derived exosomes in affecting osteosarcoma progression.
Collapse
|
44
|
Guo Z, Chen W, Dai G, Huang Y. Cordycepin suppresses the migration and invasion of human liver cancer cells by downregulating the expression of CXCR4. Int J Mol Med 2019; 45:141-150. [PMID: 31746344 PMCID: PMC6889938 DOI: 10.3892/ijmm.2019.4391] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is a worldwide threat to human health. High expression levels of C-X-C chemokine receptor type 4 (CXCR4) have been reported to promote the migration and invasion capacities of liver cancer cells. Cordycepin, extracted from Cordyceps militaris, has anti-inflammatory, antioxidant and anticancerous properties. Therefore, in the present study, migration assays, western blotting, reverse transcription-quantitative PCR and immunofluorescence analyses were conducted to determine whether cordycepin was able to suppress the migration and invasion abilities of liver cancer cells by inhibiting CXCR4 expression. The results suggested that cordycepin notably inhibited migration and invasion, and decreased the expression of CXCR4 in a dose-dependent manner. Activation of phosphorylated (p-) NF-κB inhibitor α (IκBα) and p-P65, the principal components of the NF-κB signaling pathway, was also downregulated. In addition, cordycepin markedly suppressed the nuclear translocation of P65, but had no effect on the expression of total IκBα (t-IκBα) and total P65 (t-P65). JSH-23, an inhibitor of the NF-κB pathway, impaired the migration of liver cancer cells, and was found to act synergistically with cordycepin. Furthermore, cordycepin treatment reduced the chemotactic migration ability of liver cancer cells to stromal cell-derived factor 1 (SDF1), which was significantly enhanced following treatment with JSH-23. Collectively, the present results indicated that cordycepin inhibited the nuclear translocation of P65 by preventing p-IκBα activation; this resulted in the downregulation of CXCR4 expression, and subsequently, in the impaired migration and invasion abilities of liver cancer cells and attenuated reactivity to SDF1. The current study revealed a novel mechanism for the antimetastatic activity of cordycepin and its potential to exert positive synergistic effects with other compounds for the treatment of liver cancer.
Collapse
Affiliation(s)
- Zhongrong Guo
- First Department of General Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian 363000, P.R. China
| | - Wen Chen
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Guisen Dai
- Department of Hepatobiliary Surgery, Longyan First Hospital, Longyan, Fujian 364000, P.R. China
| | - Yuanliang Huang
- Interventional Diagnosis and Treatment Section, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361001, P.R. China
| |
Collapse
|
45
|
D'Alterio C, Buoncervello M, Ieranò C, Napolitano M, Portella L, Rea G, Barbieri A, Luciano A, Scognamiglio G, Tatangelo F, Anniciello AM, Monaco M, Cavalcanti E, Maiolino P, Romagnoli G, Arra C, Botti G, Gabriele L, Scala S. Targeting CXCR4 potentiates anti-PD-1 efficacy modifying the tumor microenvironment and inhibiting neoplastic PD-1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:432. [PMID: 31661001 PMCID: PMC6819555 DOI: 10.1186/s13046-019-1420-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/10/2019] [Indexed: 12/21/2022]
Abstract
Background Inefficient T-cell access to the tumor microenvironment (TME) is among the causes of tumor immune-resistance. Previous evidence demonstrated that targeting CXCR4 improves anti-PD-1/PD-L1 efficacy reshaping TME. To evaluate the role of newly developed CXCR4 antagonists (PCT/IB2011/000120/ EP2528936B1/US2013/0079292A1) in potentiating anti-PD-1 efficacy two syngeneic murine models, the MC38 colon cancer and the B16 melanoma-human CXCR4-transduced, were employed. Methods Mice were subcutaneously injected with MC38 (1 × 106) or B16-hCXCR4 (5 × 105). After two weeks, tumors bearing mice were intraperitoneally (ip) treated with murine anti-PD-1 [RMP1–14] (5 mg/kg, twice week for 2 weeks), Pep R (2 mg/kg, 5 days per week for 2 weeks), or both agents. The TME was evaluated through immunohistochemistry and flow-cytometry. In addition, the effects of the human-anti-PD-1 nivolumab and/or Peptide-R54 (Pep R54), were evaluated on human melanoma PES43 cells and xenografts treated. Results The combined treatment, Pep R plus anti-PD-1, reduced the MC38 Relative Tumor Volume (RTV) by 2.67 fold (p = 0.038) while nor anti-PD-1, neither Pep R significantly impacted on tumor growth. Significant higher number of Granzyme B (GZMB) positive cells was detected in MC38 tumors from mice treated with the combined treatment (p = 0.016) while anti-PD-1 determined a modest but significant increase of tumor-infiltrating GZMB positive cells (p = 0.035). Also, a lower number of FoxP3 positive cells was detected (p = 0.022). In the B16-hCXCR4 tumors, two weeks of combined treatment reduced tumor volume by 2.27 fold while nor anti-PD-1 neither Pep R significantly impacted on tumor growth. A significant higher number of GRZB positive cells was observed in B16-hCXCR4 tumors treated with combined treatment (p = 0,0015) as compared to anti-PD-1 (p = 0.028). The combined treatment reduced CXCR4, CXCL12 and PD-L1 expression in MC38 tumors. In addition, flow cytometry on fresh B16-hCXCR4 tumors showed significantly higher Tregs number following anti-PD-1 partially reversed by the combined treatment Pep R and anti-PD-1. Combined treatment determined an increase of CD8/Tregs and CD8/MDSC ratio. To dissect the effect of anti-PD-1 and CXCR4 targeting on PD-1 expressed by human cancer cells, PES43 human melanoma xenograft model was employed. In vitro human anti-PD-1 nivolumab or pembrolizumab (10 μM) reduced PES43 cells growth while nivolumab (10 μM) inhibited pERK1/2, P38 MAPK, pAKT and p4EBP. PES43 xenograft mice were treated with Pep R54, a newly developed Pep R derivative (AcHN-Arg-Ala-[DCys-Arg- Nal(2′)-His-Pen]- COOH), plus nivolumab. After 3 weeks of combined treatment a significant reduction in tumor growth was shown (p = 0.038). PES43 lung disseminated tumor cells (DTC) were detected in fresh lung tissues as melanoma positive MCSP-APC+ cells. Although not statistically significant, DTC-PES43 cells were reduced in mice lungs treated with combined treatment while nivolumab or Pep R54 did not affect DTC number. Conclusion Combined treatment with the new developed CXCR4 antagonist, Pep R, plus anti-PD-1, reduced tumor-growth in two syngeneic murine models, anti-PD-1 sensitive and resistant, potentiating Granzyme and reducing Foxp3 cells infiltration. In addition, the human specific CXCR4 antagonist, Pep R54, cooperated with nivolumab in inhibiting the growth of the PD-1 expressing human PES43 melanoma xenograft. This evidence sheds light on PD-1 targeting mechanisms and paves the way for CXCR4/PD-1 targeting combination therapy.
Collapse
Affiliation(s)
- Crescenzo D'Alterio
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Maria Buoncervello
- Research Coordination and Support Service, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Caterina Ieranò
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Maria Napolitano
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Luigi Portella
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Giuseppina Rea
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Giosuè Scognamiglio
- Pathology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Fabiana Tatangelo
- Pathology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Anna Maria Anniciello
- Pathology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Mario Monaco
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Ernesta Cavalcanti
- Division of Laboratory Medicine, Department of Pathology and Laboratory Diagnostics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Piera Maiolino
- Pharmacy, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Giulia Romagnoli
- Department of Haematology, Oncology and Molecular Biology Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Claudio Arra
- Animal Facility, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Gerardo Botti
- Pathology, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy
| | - Lucia Gabriele
- Department of Haematology, Oncology and Molecular Biology Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy
| | - Stefania Scala
- Functional Genomics, Istituto Nazionale Tumori "Fondazione G. Pascale", IRCCS, 80,131, Naples, Italy.
| |
Collapse
|
46
|
Shang S, Wang J, Chen S, Tian R, Zeng H, Wang L, Xia M, Zhu H, Zuo C. Exosomal miRNA-1231 derived from bone marrow mesenchymal stem cells inhibits the activity of pancreatic cancer. Cancer Med 2019; 8:7728-7740. [PMID: 31642612 PMCID: PMC6912060 DOI: 10.1002/cam4.2633] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/28/2019] [Accepted: 10/06/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a highly malignant tumor with increased morbidity and mortality, which is difficult to diagnose and cure in the clinic. Through secreting exosomes containing biological molecules, including diverse RNAs and proteins, bone marrow mesenchymal stem cells (BM-MSCs) influence the immunity, inflammation, tumor environment, and cancer metastasis. In this study, low expression of miRNA-1231 (miR-1231) in exosomes derived from the peripheral blood was significantly correlated with the TNM stage of PC, suggesting the potential inhibitory effect of exosomal miR-1231 on PC occurrence and development. The proliferation, migration, invasion, and adhesion to the matrix of PC cells BxPC-3 and PANC-1 were negatively regulated by exosomes derived from the supernatants of BM-MSCs that transfected with miR-1231 oligonucleotides. Simultaneously, tumor growth in vivo was seriously restrained in BALB/C nude mice by tail vein injection with exosomes originated from BM-MSCs that transfected with miR-1231 mimics. The exosomes extracted from BM-MSCs with high level of miR-1231 inhibit the activity of PC, providing the potential application for developing new and efficient medicine for cancer therapy, especially for PC treatment. The exosomal miR-1231 of peripheral blood may also be a potential indicator for PC diagnosis in the future.
Collapse
Affiliation(s)
- Song Shang
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Jinfeng Wang
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Shilin Chen
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Renyun Tian
- Department of Molecular Medicine, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Hui Zeng
- Graduates School, University of South China, Hengyang, China
| | - Liang Wang
- Graduates School, University of South China, Hengyang, China
| | - Man Xia
- Department of Gynecological Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| | - Haizhen Zhu
- Department of Molecular Medicine, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, China
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Laboratory of Digestive Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, China
| |
Collapse
|
47
|
Hill BS, Sarnella A, D'Avino G, Zannetti A. Recruitment of stromal cells into tumour microenvironment promote the metastatic spread of breast cancer. Semin Cancer Biol 2019; 60:202-213. [PMID: 31377307 DOI: 10.1016/j.semcancer.2019.07.028] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
Currently, metastasis remains the primary cause of death of patients with breast cancer despite the important advances in the treatment of this disease. In the complex tumour microenvironment network, several malignant and non-malignant cell types as well as components of extracellular matrix cooperate in promoting the metastatic spread of breast carcinoma. Many components of the stromal compartment are recruited from distant sites to the tumour including mesenchymal stem cells, endothelial cells, macrophages and other immune cells whereas other cells such as fibroblasts are already present in both primary and secondary lesions. When these cells come into contact with cancer cells they are "educated" and acquire a pro-tumoural phenotype, which support all the steps of the metastatic cascade. In this Review, we highlight the role played by each stromal component in guiding cancer cells in their venture towards colonizing metastatic sites.
Collapse
|
48
|
Ou X, Zhang GT, Tian PK, Chen JS, Lin ZW, Xie Y, Wang AH, Liu XP, Liu JK. Forkhead box P3 gene silencing inhibits the expression of chemokines and chemokine receptors associated with cell growth, migration, and apoptosis in hepatocellular carcinoma cells. Exp Ther Med 2019; 18:1091-1098. [PMID: 31316604 PMCID: PMC6601415 DOI: 10.3892/etm.2019.7658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 05/02/2019] [Indexed: 01/23/2023] Open
Abstract
The aberrant expression of forkhead box P3 (FOXP3) leads to the formation of malignant tumors. FOXP3 expression levels are also elevated in hepatocellular carcinoma (HCC). The aim of the present study was to investigate the effects of FOXP3 silencing on cell proliferation, migration, apoptosis and chemokine/chemokine receptor expression in the MHCC-97H HCC cell line. Three FOXP3 short hairpin (sh)RNA constructs were designed: Sh-FOXP3-1-pGreenPuro, sh-FOXP3-2-pGreenPuro, and sh-FOXP3-3-pGreenPuro. MHCC-97H cells were transfected with shRNA-FOXP3, and the mRNA and protein expression levels of C-X-C motif chemokine (CXC) ligand 12 (CXCL12), CXCL11, CXC receptor 4 (CXCR4) and CXCR7 were measured. Cell Counting Kit-8, terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling and Transwell assays were used to evaluate cell proliferation, apoptosis and migration, respectively. Of the three FOXP3 lentivirus carriers constructed, sh-FOXP3-1 significantly reduced FOXP3 expression levels and was chosen for further experiments. sh-FOXP3-1 inhibited cell proliferation, promoted apoptosis and inhibited cell migration compared with the negative control. The mRNA and protein expression levels of CXCL12, CXCL11, CXCR4 and CXCR7 were decreased significantly in response to FOXP3 silencing. FOXP3 silencing may therefore inhibit cell growth, induce apoptosis and inhibit migration in HCC cells, possibly by impairing the chemokine/chemokine receptor axes.
Collapse
Affiliation(s)
- Xi Ou
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Guang-Tao Zhang
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Pei-Kai Tian
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Jing-Sen Chen
- Department of Breast Surgery, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong 518028, P.R. China
| | - Ze-Wei Lin
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yong Xie
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Ai-Hong Wang
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xiao-Ping Liu
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Ji-Kui Liu
- Department of Hepatopancreatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
49
|
Stamatopoulos A, Stamatopoulos T, Gamie Z, Kenanidis E, Ribeiro RDC, Rankin KS, Gerrand C, Dalgarno K, Tsiridis E. Mesenchymal stromal cells for bone sarcoma treatment: Roadmap to clinical practice. J Bone Oncol 2019; 16:100231. [PMID: 30956944 PMCID: PMC6434099 DOI: 10.1016/j.jbo.2019.100231] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, there has been growing interest in understanding the molecular mechanisms of cancer pathogenesis and progression, as it is still associated with high morbidity and mortality. Current management of large bone sarcomas typically includes the complex therapeutic approach of limb salvage or sacrifice combined with pre- and postoperative multidrug chemotherapy and/or radiotherapy, and is still associated with high recurrence rates. The development of cellular strategies against specific characteristics of tumour cells appears to be promising, as they can target cancer cells selectively. Recently, Mesenchymal Stromal Cells (MSCs) have been the subject of significant research in orthopaedic clinical practice through their use in regenerative medicine. Further research has been directed at the use of MSCs for more personalized bone sarcoma treatments, taking advantage of their wide range of potential biological functions, which can be augmented by using tissue engineering approaches to promote healing of large defects. In this review, we explore the use of MSCs in bone sarcoma treatment, by analyzing MSCs and tumour cell interactions, transduction of MSCs to target sarcoma, and their clinical applications on humans concerning bone regeneration after bone sarcoma extraction.
Collapse
Key Words
- 5-FC, 5-fluorocytosine
- AAT, a1-antitrypsin
- APCs, antigen presenting cells
- ASC, adipose-derived stromal/stem cells
- Abs, antibodies
- Ang1, angiopoietin-1
- BD, bone defect
- BMMSCs, bone marrow-derived mesenchymal stromal cells
- Biology
- Bone
- CAM, cell adhesion molecules
- CCL5, chemokine ligand 5
- CCR2, chemokine receptor 2
- CD, classification determinants
- CD, cytosine deaminase
- CLUAP1, clusterin associated protein 1
- CSPG4, Chondroitin sulfate proteoglycan 4
- CX3CL1, chemokine (C-X3-C motif) ligand 1
- CXCL12/CXCR4, C-X-C chemokine ligand 12/ C-X-C chemokine receptor 4
- CXCL12/CXCR7, C-X-C chemokine ligand 12/ C-X-C chemokine receptor 7
- CXCR4, chemokine receptor type 4
- Cell
- DBM, Demineralized Bone Marrow
- DKK1, dickkopf-related protein 1
- ECM, extracellular matrix
- EMT, epithelial-mesenchymal transition
- FGF-2, fibroblast growth factors-2
- FGF-7, fibroblast growth factors-7
- GD2, disialoganglioside 2
- HER2, human epidermal growth factor receptor 2
- HGF, hepatocyte growth factor
- HMGB1/RACE, high mobility group box-1 protein/ receptor for advanced glycation end-products
- IDO, indoleamine 2,3-dioxygenase
- IFN-α, interferon alpha
- IFN-β, interferon beta
- IFN-γ, interferon gamma
- IGF-1R, insulin-like growth factor 1 receptor
- IL-10, interleukin-10
- IL-12, interleukin-12
- IL-18, interleukin-18
- IL-1b, interleukin-1b
- IL-21, interleukin-21
- IL-2a, interleukin-2a
- IL-6, interleukin-6
- IL-8, interleukin-8
- IL11RA, Interleukin 11 Receptor Subunit Alpha
- MAGE, melanoma antigen gene
- MCP-1, monocyte chemoattractant protein-1
- MMP-2, matrix metalloproteinase-2
- MMP2/9, matrix metalloproteinase-2/9
- MRP, multidrug resistance protein
- MSCs, mesenchymal stem/stromal cells
- Mesenchymal
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- OPG, osteoprotegerin
- Orthopaedic
- PBS, phosphate-buffered saline
- PDGF, platelet-derived growth factor
- PDX, patient derived xenograft
- PEDF, pigment epithelium-derived factor
- PGE2, prostaglandin E2
- PI3K/Akt, phosphoinositide 3-kinase/protein kinase B
- PTX, paclitaxel
- RANK, receptor activator of nuclear factor kappa-B
- RANKL, receptor activator of nuclear factor kappa-B ligand
- RBCs, red blood cells
- RES, reticuloendothelial system
- RNA, ribonucleic acid
- Regeneration
- SC, stem cells
- SCF, stem cells factor
- SDF-1, stromal cell-derived factor 1
- STAT-3, signal transducer and activator of transcription 3
- Sarcoma
- Stromal
- TAAs, tumour-associated antigens
- TCR, T cell receptor
- TGF-b, transforming growth factor beta
- TGF-b1, transforming growth factor beta 1
- TNF, tumour necrosis factor
- TNF-a, tumour necrosis factor alpha
- TRAIL, tumour necrosis factor related apoptosis-inducing ligand
- Tissue
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
- WBCs, white blood cell
- hMSCs, human mesenchymal stromal cells
- rh-TRAIL, recombinant human tumour necrosis factor related apoptosis-inducing ligand
Collapse
Affiliation(s)
- Alexandros Stamatopoulos
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Theodosios Stamatopoulos
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Zakareya Gamie
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Eustathios Kenanidis
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Ricardo Da Conceicao Ribeiro
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Kenneth Samora Rankin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Craig Gerrand
- Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, HA7 4LP, UK
| | - Kenneth Dalgarno
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Eleftherios Tsiridis
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| |
Collapse
|
50
|
Jiang Y, Wells A, Sylakowski K, Clark AM, Ma B. Adult Stem Cell Functioning in the Tumor Micro-Environment. Int J Mol Sci 2019; 20:ijms20102566. [PMID: 31130595 PMCID: PMC6566759 DOI: 10.3390/ijms20102566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor progression from an expanded cell population in a primary location to disseminated lethal growths subverts attempts at cures. It has become evident that these steps are driven in a large part by cancer cell-extrinsic signaling from the tumor microenvironment (TME), one cellular component of which is becoming more appreciated for potential modulation of the cancer cells directly and the TME globally. That cell is a heterogenous population referred to as adult mesenchymal stem cells/multipotent stromal cells (MSCs). Herein, we review emerging evidence as to how these cells, both from distant sources, mainly the bone marrow, or local resident cells, can impact the progression of solid tumors. These nascent investigations raise more questions than they answer but paint a picture of an orchestrated web of signals and interactions that can be modulated to impact tumor progression.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| | - Kyle Sylakowski
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| | - Bo Ma
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| |
Collapse
|