1
|
Taeb S, Rostamzadeh D, Mafi S, Mofatteh M, Zarrabi A, Hushmandi K, Safari A, Khodamoradi E, Najafi M. Update on Mesenchymal Stem Cells: A Crucial Player in Cancer Immunotherapy. Curr Mol Med 2024; 24:98-113. [PMID: 36573062 DOI: 10.2174/1566524023666221226143814] [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: 05/17/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 12/28/2022]
Abstract
The idea of cancer immunotherapy has spread, and it has made tremendous progress with the advancement of new technology. Immunotherapy, which serves to assist the natural defenses of the body in eradicating cancerous cells, is a remarkable achievement that has revolutionized both cancer research and cancer treatments. Currently, the use of stem cells in immunotherapy is widespread and shares a special characteristic, including cancer cell migration, bioactive component release, and immunosuppressive activity. In the context of cancer, mesenchymal stem cells (MSCs) are rapidly being identified as vital stromal regulators of tumor progression. MSCs therapy has been implicated in treating a wide range of diseases, including bone damage, autoimmune diseases, and particularly hematopoietic abnormalities, providing stem cell-based therapy with an extra dimension. Moreover, the implication of MSCs does not have ethical concerns, and the complications known in pluripotent and totipotent stem cells are less common in MSCs. MSCs have a lot of distinctive characteristics that, when coupled, make them excellent for cellular-based immunotherapy and as vehicles for gene and drug delivery in a variety of inflammations and malignancies. MSCs can migrate to the inflammatory site and exert immunomodulatory responses via cell-to-cell contacts with lymphocytes by generating soluble substances. In the current review, we discuss the most recent research on the immunological characteristics of MSCs, their use as immunomodulatory carriers, techniques for approving MSCs to adjust their immunological contour, and their usages as vehicles for delivering therapeutic as well as drugs and genes engineered to destroy tumor cells.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Sahar Mafi
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohammad Mofatteh
- Sir William Dunn School of Pathology, Medical Sciences Division, University of Oxford, South Parks Road, Oxford OX1 3RE, United Kingdom
- Lincoln College, University of Oxford, Turl Street, Oxford OX1 3DR, United Kingdom
| | - Ali Zarrabi
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Arash Safari
- Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
2
|
Martin-Morales N, Padial-Molina M, Tovar I, De Araujo Farias V, Hernández-Cortés P, Ramirez-Moreno E, Caba-Molina M, Davis J, Carrero Castaño A, Ruiz de Almodovar JM, Galindo-Moreno P, Oliver-Pozo J, O'Valle Ravassa FJ. IMP3 Immunohistochemical Expression Is Related with Progression and Metastases in Xenografted and Cutaneous Melanomas. Pathobiology 2023; 91:132-143. [PMID: 37797584 DOI: 10.1159/000533916] [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: 04/21/2023] [Accepted: 08/19/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Insulin-like growth factor-II messenger RNA-binding protein-3 (IMP3) over-expression is a predictor of tumor recurrence and metastases in some types of human melanoma. Our objective was to evaluate the immunohistochemical expression of IMP3 and other molecules related to tumor prognosis in melanoma-xeno-tumors undergoing treatment. We test the effect of radiotherapy (RT) and mesenchymal stromal cells (MSCs) treatment, analyzing the tumorigenic and metastatsizing capacity in a mice melanoma xenograft model. MATERIALS AND METHODS We inoculated A375 and G361 human melanoma cell lines into NOD/SCID gamma mice (n = 64). We established a control group, a group treated with MSCs, a group treated with MSCs plus RT, and a group treated with RT. We assessed the immunohistochemical expression of IMP3, E-cadherin, N-cadherin, PARP1, HIF-1α, and the proliferation marker Ki-67. Additionally, we performed a retrospective study including 114 histological samples of patients diagnosed with malignant cutaneous superficial spreading melanoma (n = 104) and nodular melanoma (n = 10) with at least 5 years of follow-up. RESULTS Most morphological and immunohistochemical features show statistically significant differences between the 2 cell lines. The A375 cell line induced the formation of metastases, while the G361 cell line provoked tumor formation but not metastases. All three treatments reduced the cell proliferation evaluated by the Ki-67 nuclear antigen (p = 0.000, one-way ANOVA test) and reduced the number of metastases (p = 0.004, one-way ANOVA test). In addition, the tumor volumes reduced in comparison with the control groups, 31.74% for RT + MSCs in the A357 tumor cell line, and 89.84% RT + MSCs in the G361 tumor cell line. We also found that IMP3 expression is associated with greater tumor aggressiveness and was significantly correlated with cell proliferation (measured by the expression of Ki-67), the number of metastases, and reduced expression of adhesion molecules. CONCLUSIONS The combined treatment of RT and MSCs on xenografted melanomas reduces tumor size, metastases frequency, and the epithelial to mesenchymal transition/PARP1 metastatic phenotype. This treatment also reduces the expression of molecules related to cellular proliferation (Ki-67), molecules that facilitate the metastatic process (E-cadherin), and molecules related with prognosis (IMP3).
Collapse
Affiliation(s)
- Natividad Martin-Morales
- Department of Pathology, University of Granada, Granada, Spain,
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain,
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Biosanitary Institute (Ibs.GRANADA), Granada, Spain
| | - Isabel Tovar
- Department of Oncology and Radiotherapy, Virgen de las Nieves University Hospital, Granada, Spain
| | - Virginea De Araujo Farias
- Institute of Biopathology and Medicine Regenerative (IBIMER, CIBM), University of Granada, Granada, Spain
| | - Pedro Hernández-Cortés
- Biosanitary Institute (Ibs.GRANADA), Granada, Spain
- Department of Orthopedic Surgery, Clinic San Cecilio University Hospital, Granada, Spain
| | | | - Mercedes Caba-Molina
- Department of Pathology, University of Granada, Granada, Spain
- Biosanitary Institute (Ibs.GRANADA), Granada, Spain
- Intercentre Provincial Pathological Anatomy Unit of the San Cecilio Clinical University Hospital, Granada, Spain
| | - Justin Davis
- Department of Business Administration, Washington and Lee University, Lexington, Virginia, USA
| | - Alejandro Carrero Castaño
- Intercentre Provincial Pathological Anatomy Unit of the San Cecilio Clinical University Hospital, Granada, Spain
| | | | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, Granada, Spain
- Biosanitary Institute (Ibs.GRANADA), Granada, Spain
| | - Javier Oliver-Pozo
- Institute of Parasitology and Biomedicine López Neyra, CSIC, Granada, Spain
| | - Francisco Javier O'Valle Ravassa
- Department of Pathology, University of Granada, Granada, Spain
- Biosanitary Institute (Ibs.GRANADA), Granada, Spain
- Institute of Biopathology and Medicine Regenerative (IBIMER, CIBM), University of Granada, Granada, Spain
| |
Collapse
|
3
|
Khosravi N, Pishavar E, Baradaran B, Oroojalian F, Mokhtarzadeh A. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: A promising biomimetic nanoplatforms for cancer theranostics. J Control Release 2022; 348:706-722. [PMID: 35732250 DOI: 10.1016/j.jconrel.2022.06.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Nanomedicine research has advanced dramatically in recent decades. Nonetheless, traditional nanomedicine faces significant obstacles such as the low concentration of the drug at target sites and accelerated removal of the drug from blood circulation. Various techniques of nanotechnology, including cell membrane coating, have been developed to address these challenges and to improve targeted distribution and redcue cell membrane-mediated immunogenicity. Recently, stem cell (SC) membranes, owing to their immunosuppressive and regenerative properties, have grabbed attention as attractive therapeutic carriers for targeting specific tissues or organs. Bioengineering strategies that combine synthetic nanoparticles (NPs) with SC membranes, because of their homing potential and tumor tropism, have recently received a lot of publicity. Several laboratory experiments and clinical trials have indicated that the benefits of SC-based technologies are mostly related to the effects of SC-derived exosomes (SC-Exos). Exosomes are known as nano-sized extracellular vehicles (EVs) that deliver particular bioactive molecules for cell-to-cell communication. In this regard, SC-derived exosome membranes have recently been employed to improve the therapeutic capability of engineered drug delivery vehicles. Most recently, for further enhancing NPs' functionality, a new coating approach has been offered that combines membranes from two separate cells. These hybrid membrane delivery vehicles have paved the way for the development of biocompatible, high-efficiency, biomimetic NPs with varying hybrid capabilities that can overcome the drawbacks of present NP-based treatment techniques. This review explores stem cell membranes, SC-Exos, and hybrid SC-camouflaged NPs preparation methods and their importance in cancer therapy.
Collapse
Affiliation(s)
- Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Pishavar
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Kitzberger C, Spellerberg R, Morath V, Schwenk N, Schmohl KA, Schug C, Urnauer S, Tutter M, Eiber M, Schilling F, Weber WA, Ziegler S, Bartenstein P, Wagner E, Nelson PJ, Spitzweg C. The sodium iodide symporter (NIS) as theranostic gene: its emerging role in new imaging modalities and non-viral gene therapy. EJNMMI Res 2022; 12:25. [PMID: 35503582 PMCID: PMC9065223 DOI: 10.1186/s13550-022-00888-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/11/2022] [Indexed: 01/14/2023] Open
Abstract
Cloning of the sodium iodide symporter (NIS) in 1996 has provided an opportunity to use NIS as a powerful theranostic transgene. Novel gene therapy strategies rely on image-guided selective NIS gene transfer in non-thyroidal tumors followed by application of therapeutic radionuclides. This review highlights the remarkable progress during the last two decades in the development of the NIS gene therapy concept using selective non-viral gene delivery vehicles including synthetic polyplexes and genetically engineered mesenchymal stem cells. In addition, NIS is a sensitive reporter gene and can be monitored by high resolution PET imaging using the radiotracers sodium [124I]iodide ([124I]NaI) or [18F]tetrafluoroborate ([18F]TFB). We performed a small preclinical PET imaging study comparing sodium [124I]iodide and in-house synthesized [18F]TFB in an orthotopic NIS-expressing glioblastoma model. The results demonstrated an improved image quality using [18F]TFB. Building upon these results, we will be able to expand the NIS gene therapy approach using non-viral gene delivery vehicles to target orthotopic tumor models with low volume disease, such as glioblastoma. Trial registration not applicable.
Collapse
Affiliation(s)
- Carolin Kitzberger
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Rebekka Spellerberg
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Volker Morath
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nathalie Schwenk
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Kathrin A Schmohl
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Christina Schug
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Sarah Urnauer
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Mariella Tutter
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Centre for System-Based Drug Research and Centre for Nanoscience, LMU Munich, Munich, Germany
| | - Peter J Nelson
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany. .,Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Kwong GA, Ghosh S, Gamboa L, Patriotis C, Srivastava S, Bhatia SN. Synthetic biomarkers: a twenty-first century path to early cancer detection. Nat Rev Cancer 2021; 21:655-668. [PMID: 34489588 PMCID: PMC8791024 DOI: 10.1038/s41568-021-00389-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 02/08/2023]
Abstract
Detection of cancer at an early stage when it is still localized improves patient response to medical interventions for most cancer types. The success of screening tools such as cervical cytology to reduce mortality has spurred significant interest in new methods for early detection (for example, using non-invasive blood-based or biofluid-based biomarkers). Yet biomarkers shed from early lesions are limited by fundamental biological and mass transport barriers - such as short circulation times and blood dilution - that limit early detection. To address this issue, synthetic biomarkers are being developed. These represent an emerging class of diagnostics that deploy bioengineered sensors inside the body to query early-stage tumours and amplify disease signals to levels that could potentially exceed those of shed biomarkers. These strategies leverage design principles and advances from chemistry, synthetic biology and cell engineering. In this Review, we discuss the rationale for development of biofluid-based synthetic biomarkers. We examine how these strategies harness dysregulated features of tumours to amplify detection signals, use tumour-selective activation to increase specificity and leverage natural processing of bodily fluids (for example, blood, urine and proximal fluids) for easy detection. Finally, we highlight the challenges that exist for preclinical development and clinical translation of synthetic biomarker diagnostics.
Collapse
Affiliation(s)
- Gabriel A Kwong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA.
- Parker H. Petit Institute of Bioengineering and Bioscience, Atlanta, GA, USA.
- Institute for Electronics and Nanotechnology, Georgia Tech, Atlanta, GA, USA.
- The Georgia Immunoengineering Consortium, Emory University and Georgia Tech, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| | - Sharmistha Ghosh
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Lena Gamboa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, USA
| | - Christos Patriotis
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sudhir Srivastava
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
6
|
Effect of Stem Cell-Derived Extracellular Vesicles on Damaged Human Corneal Endothelial Cells. Stem Cells Int 2021; 2021:6644463. [PMID: 33531909 PMCID: PMC7834816 DOI: 10.1155/2021/6644463] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/19/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose Human corneal endothelial cells (HCECs) are essential to visual function; however, since they have limited proliferative capacity in vivo, they are prone to corneal endothelial dysfunction. At present, the only treatment is a corneal transplantation from donor cadavers. Also, due to a global shortage of donor corneas, it is important to find alternative strategies. Recent studies highlight that stem cell–derived extracellular vesicles (EVs) play a relevant role in stem cell-induced regeneration by reprogramming injured cells and inducing proregenerative pathways. The aim of this work is to evaluate whether EVs derived from mesenchymal stem cells (MSC-EVs) are able to promote regeneration of damaged HCECs. Methods We isolated HCECs from discarded corneas in patients undergoing corneal transplantation or enucleation (N = 23 patients). Bone marrow mesenchymal stem cells (MSCs) were obtained from Lonza, cultured, and characterized. MSC-EVs were obtained from supernatants of MSCs. In order to establish a valid in vitro damage model to test the regenerative potential of EVs on HCECs, we evaluated the proliferation rate and the apoptosis after exposing the cells to serum-deprived medium at different concentrations for 24 hours. We then evaluated the HCEC migration through a wound healing assay. Results In the selected serum deprivation damage conditions, the treatment with different doses of MSC-EVs resulted in a significantly higher proliferation rate of HCECs at all the tested concentrations of EVs (5‐20 × 103 MSC-EV/cell). MSC-EVs/cell induced a significant decrease in number of total apoptotic cells after 24 hours of serum deprivation. Finally, the wound healing assay showed a significantly faster repair of the wound after HCEC treatment with MSC-EVs. Conclusions Results highlight the already well-known proregenerative potential of MSC-EVs in a totally new biological model, the endothelium of the cornea. MSC-EVs, indeed, induced proliferation and survival of HCECs, promoting the migration of HCECs in vitro.
Collapse
|
7
|
Regional Hyperthermia Enhances Mesenchymal Stem Cell Recruitment to Tumor Stroma: Implications for Mesenchymal Stem Cell-Based Tumor Therapy. Mol Ther 2020; 29:788-803. [PMID: 33068779 DOI: 10.1016/j.ymthe.2020.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/22/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
The tropism of mesenchymal stem cells (MSCs) for tumors forms the basis for their use as delivery vehicles for the tumor-specific transport of therapeutic genes, such as the theranostic sodium iodide symporter (NIS). Hyperthermia is used as an adjuvant for various tumor therapies and has been proposed to enhance leukocyte recruitment. Here, we describe the enhanced recruitment of adoptively applied NIS-expressing MSCs to tumors in response to regional hyperthermia. Hyperthermia (41°C, 1 h) of human hepatocellular carcinoma cells (HuH7) led to transiently increased production of immunomodulatory factors. MSCs showed enhanced chemotaxis to supernatants derived from heat-treated cells in a 3D live-cell tracking assay and was validated in vivo in subcutaneous HuH7 mouse xenografts. Cytomegalovirus (CMV)-NIS-MSCs were applied 6-48 h after or 24-48 h before hyperthermia treatment. Using 123I-scintigraphy, thermo-stimulation (41°C, 1 h) 24 h after CMV-NIS-MSC injection resulted in a significantly increased uptake of 123I in heat-treated tumors compared with controls. Immunohistochemical staining and real-time PCR confirmed tumor-selective, temperature-dependent MSC migration. Therapeutic efficacy was significantly enhanced by combining CMV-NIS-MSC-mediated 131I therapy with regional hyperthermia. We demonstrate here for the first time that hyperthermia can significantly boost tumoral MSC recruitment, thereby significantly enhancing therapeutic efficacy of MSC-mediated NIS gene therapy.
Collapse
|
8
|
Feng Y, Zhan F, Zhong Y, Tan B. Effects of human umbilical cord mesenchymal stem cells derived from exosomes on migration ability of endometrial glandular epithelial cells. Mol Med Rep 2020; 22:715-722. [PMID: 32626977 PMCID: PMC7339775 DOI: 10.3892/mmr.2020.11137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 04/06/2020] [Indexed: 12/20/2022] Open
Abstract
The present study aimed to investigate the effects of human umbilical cord mesenchymal stem cells (Huc‑MSCs)‑derived exosomes on the migratory abilities of endometrial glandular epithelial cells, and to evaluate the underlying mechanism from the perspective of epithelial‑mesenchymal transition (EMT). Huc‑MSCs were prepared from human umbilical cord, and eutopic endometrial glandular epithelial cells were isolated from patients with endometriosis. The exosomes derived from Huc‑MSCs (Huc‑MSCs‑exo) were prepared using an exosome extraction kit. The endometrial glandular epithelial cells were randomly divided into two groups: Huc‑MSCs‑exo and control. Cell migratory ability was assessed and western blotting was used to detect the expression levels of EMT. The results of the present study demonstrated that Huc‑MSCs‑exo treatment significantly enhanced the migration of endometrial glandular epithelial cells from patients with endometriosis (P<0.05). The present study also demonstrated that treatment with Huc‑MSCs‑exo inhibited the expression levels of E‑cadherin and promoted the expression levels of Vimentin and N‑cadherin at both the mRNA and protein level. The results of the current study indicate that Huc‑MSCs‑exo enhance the migratory ability of endometrial glandular epithelial cells via promotion of EMT.
Collapse
Affiliation(s)
- Ying Feng
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fuliang Zhan
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanying Zhong
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Buzhen Tan
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
9
|
Tutter M, Schug C, Schmohl KA, Urnauer S, Schwenk N, Petrini M, Lokerse WJM, Zach C, Ziegler S, Bartenstein P, Weber WA, Wagner E, Lindner LH, Nelson PJ, Spitzweg C. Effective control of tumor growth through spatial and temporal control of theranostic sodium iodide symporter ( NIS) gene expression using a heat-inducible gene promoter in engineered mesenchymal stem cells. Am J Cancer Res 2020; 10:4490-4506. [PMID: 32292510 PMCID: PMC7150485 DOI: 10.7150/thno.41489] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/11/2020] [Indexed: 02/07/2023] Open
Abstract
Purpose: The tumor homing characteristics of mesenchymal stem cells (MSCs) make them attractive vehicles for the tumor-specific delivery of therapeutic agents, such as the sodium iodide symporter (NIS). NIS is a theranostic protein that allows non-invasive monitoring of the in vivo biodistribution of functional NIS expression by radioiodine imaging as well as the therapeutic application of 131I. To gain local and temporal control of transgene expression, and thereby improve tumor selectivity, we engineered MSCs to express the NIS gene under control of a heat-inducible HSP70B promoter (HSP70B-NIS-MSCs). Experimental Design: NIS induction in heat-treated HSP70B-NIS-MSCs was verified by 125I uptake assay, RT-PCR, Western blot and immunofluorescence staining. HSP70B-NIS-MSCs were then injected i.v. into mice carrying subcutaneous hepatocellular carcinoma HuH7 xenografts, and hyperthermia (1 h at 41°C) was locally applied to the tumor. 0 - 72 h later radioiodine uptake was assessed by 123I-scintigraphy. The most effective uptake regime was then selected for 131I therapy. Results: The HSP70B promoter showed low basal activity in vitro and was significantly induced in response to heat. In vivo, the highest tumoral iodine accumulation was seen 12 h after application of hyperthermia. HSP70B-NIS-MSC-mediated 131I therapy combined with hyperthermia resulted in a significantly reduced tumor growth with prolonged survival as compared to control groups. Conclusions: The heat-inducible HSP70B promoter allows hyperthermia-induced spatial and temporal control of MSC-mediated theranostic NIS gene radiotherapy with efficient tumor-selective and temperature-dependent accumulation of radioiodine in heat-treated tumors.
Collapse
|
10
|
de la Torre P, Pérez-Lorenzo MJ, Alcázar-Garrido Á, Flores AI. Cell-Based Nanoparticles Delivery Systems for Targeted Cancer Therapy: Lessons from Anti-Angiogenesis Treatments. Molecules 2020; 25:E715. [PMID: 32046010 PMCID: PMC7038177 DOI: 10.3390/molecules25030715] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 02/05/2023] Open
Abstract
The main strategy of cancer treatment has focused on attacking the tumor cells. Some cancers initially responsive to chemotherapy become treatment-resistant. Another strategy is to block the formation of tumor vessels. However, tumors also become resistant to anti-angiogenic treatments, mostly due to other cells and factors present in the tumor microenvironment, and hypoxia in the central part of the tumor. The need for new cancer therapies is significant. The use of nanoparticle-based therapy will improve therapeutic efficacy and targeting, while reducing toxicity. However, due to inefficient accumulation in tumor sites, clearance by reticuloendothelial organs and toxicity, internalization or conjugation of drug-loaded nanoparticles (NPs) into mesenchymal stem cells (MSCs) can increase efficacy by actively delivering them into the tumor microenvironment. Nanoengineering MSCs with drug-loaded NPs can increase the drug payload delivered to tumor sites due to the migratory and homing abilities of MSCs. However, MSCs have some disadvantages, and exosomes and membranes from different cell types can be used to transport drug-loaded NPs actively to tumors. This review gives an overview of different cancer approaches, with a focus on hypoxia and the emergence of NPs as drug-delivery systems and MSCs as cellular vehicles for targeted delivery due to their tumor-homing potential.
Collapse
Affiliation(s)
| | | | | | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas 12), Avda. de Cordoba s/n, 28041 Madrid, Spain; (P.d.l.T.); (M.J.P.-L.)
| |
Collapse
|
11
|
Liu L, Wong CW, Han M, Farhoodi HP, Liu G, Liu Y, Liao W, Zhao W. Meta-analysis of preclinical studies of mesenchymal stromal cells to treat rheumatoid arthritis. EBioMedicine 2019; 47:563-577. [PMID: 31501076 PMCID: PMC6796567 DOI: 10.1016/j.ebiom.2019.08.073] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND This study aims to evaluate the quality of preclinical data, determine the effect sizes, and identify experimental measures that inform efficacy using mesenchymal stromal (or stem) cells (MSC) therapy in animal models of rheumatoid arthritis (RA). METHODS Literature searches were performed on MSC preclinical studies to treat RA. MSC treatment effect sizes were determined by the most commonly used outcome measures, including paw thickness, clinical score, and histological score. FINDINGS A total of 48 studies and 94 treatment arms were included, among which 42 studies and 79 treatment arms reported that MSC improved outcomes. The effect sizes of RA treatments using MSC, when compared to the controls, were: paw thickness was ameliorated by 53.6% (95% confidence interval (CI): 26.7% -80.4%), histological score was decreased by 44.9% (95% CI: 33.3% -56.6%), and clinical score was decreased by 29.9% (95% CI: 16.7% -43.0%). Specifically, our results indicated that human umbilical cord derived MSC led to large improvements of the clinical score (-42.1%) and histological score (-51.4%). INTERPRETATION To the best of our knowledge, this meta-analysis is to quantitatively answer whether MSC represent a robust RA treatment in animal models. It suggests that in preclinical studies, MSC have consistently exhibited therapeutic benefits. The findings demonstrate a need for considering variations in different animal models and treatment protocols in future studies using MSC to treat RA in humans to maximise the therapeutic gains in the era of precision medicine. FUNDS NIH [1DP2CA195763], Baylx Inc.: BI-206512, NINDS/NIH Training Grant [Award# NS082174].
Collapse
Affiliation(s)
- Linan Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Chi W Wong
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Menglu Han
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Henry P Farhoodi
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Guangyang Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Surgery, University of California-Irvine, Irvine, CA 92697, USA
| | - Yongjun Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA
| | - Wenbin Liao
- Baylx, Inc., 1 Technology Dr, C511, Irvine, CA 92618, USA.
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California-Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California-Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California-Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California-Irvine, Irvine, CA 92697, USA; Departments of Biomedical Engineering and Biological Chemistry, University of California-Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
12
|
Adipose-derived stromal cell secretome disrupts autophagy in glioblastoma. J Mol Med (Berl) 2019; 97:1491-1506. [PMID: 31401659 DOI: 10.1007/s00109-019-01829-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 07/21/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
Mesenchymal stromal cells (MSCs) are frequently recruited to tumor sites to play a part in the tumor microenvironment (TME). However, their real impact on cancer cell behavior remains obscure. Here we investigated the effects of human adipose-derived stromal cell (hADSC) secretome in autophagy of glioblastoma (GBM), as a way to better comprehend how hADSCs influence the TME. GBM U-87 MG cells were treated with conditioned medium (CM) from hADSCs and autophagic flux was evaluated. hADSC CM treatment blocked the autophagic flux in tumor cells, as indicated by the accumulation of autophagosomes in the cytosol, the high LC3-II and p62/SQSTM1 protein levels, and the lack of increase in the amount of acidic vesicular organelles. These effects were further detected in other GBM cell lines tested and also in co-cultures of hADSCs and U-87 MG. hADSC CM did not compromise lysosomal acidification; however, it was able to activate mTORC1 signaling and, as a consequence, led to a decrease in the nuclear translocation of TFEB, a master transcriptional regulator of lysosomal biogenesis and autophagy, thereby contributing to a defective autophagic process. hADSCs secrete transforming growth factor beta 1 (TGFβ1) and this cytokine is an important mediator of CM effects on autophagy. A comprehensive knowledge of MSC roles in tumor biology is of great importance to shed light on the complex dialog between these cells and to explore such interactions therapeutically. The present results help to elucidate the paracrine effects of MSCs in tumors and bring attention to the potential to be explored in MSC secretome. KEY MESSAGES: hADSC secretome specifically affects the biology of GBM cells. hADSCs block the late steps of autophagic flux in GBM cells. hADSC secretome activates mTORC1 signaling and reduces TFEB nuclear translocation in GBM cells.
Collapse
|
13
|
Cheng S, Nethi SK, Rathi S, Layek B, Prabha S. Engineered Mesenchymal Stem Cells for Targeting Solid Tumors: Therapeutic Potential beyond Regenerative Therapy. J Pharmacol Exp Ther 2019; 370:231-241. [PMID: 31175219 PMCID: PMC6640188 DOI: 10.1124/jpet.119.259796] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have previously demonstrated considerable promise in regenerative medicine based on their ability to proliferate and differentiate into cells of different lineages. More recently, there has been a significant interest in using MSCs as cellular vehicles for targeted cancer therapy by exploiting their tumor homing properties. Initial studies focused on using genetically modified MSCs for targeted delivery of various proapoptotic, antiangiogenic, and therapeutic proteins to a wide variety of tumors. However, their use as drug delivery vehicles has been limited by poor drug load capacity. This review discusses various strategies for the nongenetic modification of MSCs that allows their use in tumor-targeted delivery of small molecule chemotherapeutic agents. SIGNIFICANCE STATEMENT: There has been considerable interest in exploiting the tumor homing potential of MSCs to develop them as a vehicle for the targeted delivery of cytotoxic agents to tumor tissue. The inherent tumor-tropic and drug-resistant properties make MSCs ideal carriers for toxic payload. While significant progress has been made in the area of the genetic modification of MSCs, studies focused on identification of molecular mechanisms that contribute to the tumor tropism along with optimization of the engineering conditions can further improve their effectiveness as drug delivery vehicles.
Collapse
Affiliation(s)
- Shen Cheng
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Susheel Kumar Nethi
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Sneha Rathi
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Buddhadev Layek
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| | - Swayam Prabha
- Departments of Experimental and Clinical Pharmacology (S.C., S.K.N., B.L., S.P.) and Pharmaceutics (S.R., S.P.), College of Pharmacy, University of Minnesota, Twin Cities, Minnesota
| |
Collapse
|
14
|
Affiliation(s)
- Miriam Filippi
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Marina Boido
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
15
|
Segaliny AI, Cheng JL, Farhoodi HP, Toledano M, Yu CC, Tierra B, Hildebrand L, Liu L, Liao MJ, Cho J, Liu D, Sun L, Gulsen G, Su MY, Sah RL, Zhao W. Combinatorial targeting of cancer bone metastasis using mRNA engineered stem cells. EBioMedicine 2019; 45:39-57. [PMID: 31281099 PMCID: PMC6642316 DOI: 10.1016/j.ebiom.2019.06.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Bone metastases are common and devastating to cancer patients. Existing treatments do not specifically target the disease sites and are therefore ineffective and systemically toxic. Here we present a new strategy to treat bone metastasis by targeting both the cancer cells ("the seed"), and their surrounding niche ("the soil"), using stem cells engineered to home to the bone metastatic niche and to maximise local delivery of multiple therapeutic factors. METHODS We used mesenchymal stem cells engineered using mRNA to simultaneously express P-selectin glycoprotein ligand-1 (PSGL-1)/Sialyl-Lewis X (SLEX) (homing factors), and modified versions of cytosine deaminase (CD) and osteoprotegerin (OPG) (therapeutic factors) to target and treat breast cancer bone metastases in two mouse models, a xenograft intratibial model and a syngeneic model of spontaneous bone metastasis. FINDINGS We first confirmed that MSC engineered using mRNA produced functional proteins (PSGL-1/SLEX, CD and OPG) using various in vitro assays. We then demonstrated that mRNA-engineered MSC exhibit enhanced homing to the bone metastatic niche likely through interactions between PSGL-1/SLEX and P-selectin expressed on tumour vasculature. In both the xenograft intratibial model and syngeneic model of spontaneous bone metastasis, engineered MSC can effectively kill tumour cells and preserve bone integrity. The engineered MSC also exhibited minimal toxicity in vivo, compared to its non-targeted chemotherapy counterpart (5-fluorouracil). INTERPRETATION Our combinatorial targeting of both the cancer cells and the niche represents a simple, safe and effective way to treat metastatic bone diseases, otherwise difficult to manage with existing strategies. It can also be applied to other cell types (e.g., T cells) and cargos (e.g., genome editing components) to treat a broad range of cancer and other complex diseases. FUND: National Institutes of Health, National Cancer Institute of the National Institutes of Health, Department of Defense, California Institute of Regenerative Medicine, National Science Foundation, Baylx Inc., and Fondation ARC pour la recherche sur le cancer.
Collapse
Affiliation(s)
- Aude I Segaliny
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jason L Cheng
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Henry P Farhoodi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael Toledano
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Chih Chun Yu
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Beatrice Tierra
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Leanne Hildebrand
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Linan Liu
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael J Liao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jaedu Cho
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Dongxu Liu
- Department of Civil and Environmental Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Lizhi Sun
- Department of Civil and Environmental Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Gultekin Gulsen
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Min-Ying Su
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert L Sah
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA; Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
16
|
Schug C, Kitzberger C, Sievert W, Spellerberg R, Tutter M, Schmohl KA, Eberlein B, Biedermann T, Steiger K, Zach C, Schwaiger M, Multhoff G, Wagner E, Nelson PJ, Spitzweg C. Radiation-Induced Amplification of TGFB1-Induced Mesenchymal Stem Cell-Mediated Sodium Iodide Symporter ( NIS) Gene 131I Therapy. Clin Cancer Res 2019; 25:5997-6008. [PMID: 31196853 DOI: 10.1158/1078-0432.ccr-18-4092] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/10/2019] [Accepted: 06/10/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE The innate tumor homing potential of mesenchymal stem cells (MSCs) has been used for a targeted delivery of the theranostic sodium iodide symporter (NIS) transgene into solid tumors. We have previously shown that external beam radiotherapy (EBRT) results in the enhanced recruitment of NIS-expressing MSCs into human hepatocellular carcinoma (HuH7). In parallel, the tumor-associated cytokine TGFB1 becomes strongly upregulated in HuH7 tumors in response to EBRT. EXPERIMENTAL DESIGN We therefore evaluated the effects of combining focused EBRT (5 Gy) with MSC-mediated systemic delivery of the theranostic NIS transgene under control of a synthetic TGFB1-inducible SMAD-responsive promoter (SMAD-NIS-MSCs) using 123I-scintigraphy followed by 131I therapy in CD1 nu/nu mice harboring subcutaneous human hepatocellular carcinoma (HuH7). RESULTS Following tumor irradiation and SMAD-NIS-MSC application, tumoral iodide uptake monitored in vivo by 123I-scintigraphy was enhanced as compared with nonirradiated tumors. Combination of EBRT and SMAD-NIS-MSC-mediated 131I therapy resulted in a significantly improved delay in tumor growth and prolonged survival in therapy mice as compared with the combined therapy using CMV-NIS-MSCs or to control groups receiving EBRT or saline only, or EBRT together with SMAD-NIS-MSCs and saline applications. CONCLUSIONS MSC-based NIS-mediated 131I therapy after EBRT treatment dramatically enhanced therapeutic efficacy when a TGFB1-inducible SMAD-responsive promoter was used to drive NIS expression in adoptively applied MSCs. The remarkable therapeutic effect seen is thought to be linked in large part to the enhanced TGFB1 produced in this context, which leads to a highly selective and focused amplification of MSC-based NIS expression within the tumor milieu.
Collapse
Affiliation(s)
- Christina Schug
- Medizinische Klinik und Poliklinik IV-Campus Grosshadern, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Carolin Kitzberger
- Medizinische Klinik und Poliklinik IV-Campus Grosshadern, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wolfgang Sievert
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar der Technischen Universität München, Radiation Immuno-Oncology group, Munich, Germany
| | - Rebekka Spellerberg
- Medizinische Klinik und Poliklinik IV-Campus Grosshadern, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mariella Tutter
- Medizinische Klinik und Poliklinik IV-Campus Grosshadern, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kathrin A Schmohl
- Medizinische Klinik und Poliklinik IV-Campus Grosshadern, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Bernadette Eberlein
- Department of Dermatology and Allergy Biederstein, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Tilo Biedermann
- Department of Dermatology and Allergy Biederstein, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Katja Steiger
- Department of Pathology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Christian Zach
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Markus Schwaiger
- Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Gabriele Multhoff
- Center for Translational Cancer Research (TranslaTUM), Klinikum rechts der Isar der Technischen Universität München, Radiation Immuno-Oncology group, Munich, Germany
| | - Ernst Wagner
- Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christine Spitzweg
- Medizinische Klinik und Poliklinik IV-Campus Grosshadern, University Hospital of Munich, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
17
|
Ahmed ESA, Ahmed NH, Medhat AM, Said UZ, Rashed LA, Abdel Ghaffar ARB. Mesenchymal stem cells targeting PI3K/AKT pathway in leukemic model. Tumour Biol 2019; 41:1010428319846803. [PMID: 31018830 DOI: 10.1177/1010428319846803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells have therapeutic properties that are related to their potentials for trans-differentiation, immunomodulation, anti-inflammatory, inhibitory effect on tumor proliferation, and induction of apoptosis. This study was performed to analyze the role of mesenchymal stem cells as an alternative for cellular signaling growth factors involved in the pathogenesis of leukemogenesis in rats. Treatment of rats with 7,12-dimethyl benz [a] anthracene induced leukemogenesis appeared as a significant decrease in hematological parameters with concomitant significant increase in bone marrow oxidative and inflammatory indices (transforming growth factor beta and interleukin-6) in comparison with normal groups. On the contrary, Western immunoblotting showed a significant increase in the signaling growth factors: PI3K, AKT, mTOR proteins and a significant decrease in PTEN in 7,12-dimethyl benz [a] anthracene-treated group. In addition, a significant increase in the transcript levels of B cell lymphoma-2 protein gene in the 7,12-dimethyl benz [a] anthracene group, while that of C-X-C motif chemokine receptor-4 and B cell lymphoma-2 protein associated x-protein were significantly downregulated compared to controls. Meanwhile, therapeutic mesenchymal stem cells treatment predict a significant improvement versus 7,12-dimethyl benz [a] anthracene group through the modulation of growth factors that confront bone marrow dysplasia. In the same direction treatment of 7,12-dimethyl benz [a] anthracene group with mesenchymal stem cells, it induced apoptosis and increased the homing efficacy to bone marrow. In conclusion, mesenchymal stem cells improve hematopoiesis and alleviate inflammation, and modulated PI3K/AKT signaling pathway contributed to experimental leukemogenesis.
Collapse
Affiliation(s)
- Esraa S A Ahmed
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Neamat H Ahmed
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Amina M Medhat
- 2 Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ussama Z Said
- 1 National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Laila A Rashed
- 3 Biochemistry & Molecular Biology Department, Medicine Faculty, Cairo University, Cairo, Egypt
| | | |
Collapse
|
18
|
Yang Y, Zhang X, Lin F, Xiong M, Fan D, Yuan X, Lu Y, Song Y, Zhang Y, Hao M, Ye Z, Zhang Y, Wang J, Xiong D. Bispecific CD3-HAC carried by E1A-engineered mesenchymal stromal cells against metastatic breast cancer by blocking PD-L1 and activating T cells. J Hematol Oncol 2019; 12:46. [PMID: 31023384 PMCID: PMC6482514 DOI: 10.1186/s13045-019-0723-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022] Open
Abstract
Background PD-1/PD-L1 blockade can confer durable benefits in the treatment of metastatic cancers, but the response rate remains modest and potential adverse effects occur sometimes. Concentrating immunotherapeutic agents at the site of disease was believed to break local immune tolerance and reduce systemic toxicity. E1A-engineered mesenchymal stromal cell (MSC.E1A) was an attractive transfer system that preferentially homing and treating cancer metastasis, through which the tumor cells were modified by locally replicated adenoviruses to release CD3-HAC, a bifunctional fusion protein that anti-CD3 scfv linked with high-affinity consensus (HAC) PD-1. Subsequently, CD3-HAC, wbich was bound on PD-L1-positive breast cancer cells, recruited T cells to exhibit a potent antitumor immunity incombination with immune checkpoint blockade. Methods We constructed the CD3-HAC gene driven by human telomerase reverse transcriptase (hTERT) promoter into an adenoviral vector and the E1A gene into the lentiviral vector. The homing property of MSCs in vivo was analyzed with firefly luciferase-labeled MSCs (MSC.Luc) by bioluminescent imaging (BLI). The cytotoxicity of T cells induced by CD3-HAC towards PD-L1-positive cells was detected in vitro and in vivo in combination with 5-FU. Results Our data suggest that CD3-HAC could specifically bind to PD-L1-positive tumor cells and induce lymphocyte-mediated lysis effectively both in vitro and in vivo. The intervention with HAC diminished the effects of PD-1/PD-L1 axis on T cells exposed to MDA-MB-231 cells and increased lymphocytes activation. MSCs infected by AdCD3-HAC followed by LentiR.E1A could specially migrate to metastasis of breast cancer and produce adenoviruses in the tumor sites. Furthermore, treatment with MSC.CD3-HAC.E1A in combination with 5-FU significantly inhibited the tumor growth in mice. Conclusions This adenovirus-loaded MSC.E1A system provides a promising strategy for the identification and elimination of metastasis with locally released immuno-modulator. Electronic supplementary material The online version of this article (10.1186/s13045-019-0723-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuanyuan Yang
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Xiaolong Zhang
- Department of Pharmacy, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People's Republic of China
| | - Fangzhen Lin
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Mengshang Xiong
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Dongmei Fan
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Xiangfei Yuan
- Tianjin Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, People's Republic of China
| | - Yang Lu
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Yuewen Song
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Yizi Zhang
- Central Hospital of Karamay, Karamay, Xinjiang, 834000, People's Republic of China
| | - Mu Hao
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China
| | - Zhou Ye
- Central Hospital of Karamay, Karamay, Xinjiang, 834000, People's Republic of China.
| | - Yanjun Zhang
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China.
| | - Dongsheng Xiong
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, 300020, People's Republic of China.
| |
Collapse
|
19
|
Role of protein phosphatases in the cancer microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:144-152. [DOI: 10.1016/j.bbamcr.2018.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/29/2018] [Accepted: 07/11/2018] [Indexed: 12/15/2022]
|
20
|
van der Velden DL, Houthuijzen JM, Roodhart JML, van Werkhoven E, Voest EE. Detection of endogenously circulating mesenchymal stem cells in human cancer patients. Int J Cancer 2018; 143:2516-2524. [PMID: 29992568 DOI: 10.1002/ijc.31727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/05/2018] [Accepted: 05/25/2018] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells (MSCs) can play a vital role in tumor progression and anticancer therapy response, as demonstrated by various in vitro and in vivo model systems. Their ability to home to developing tumors and modulate the tumor microenvironment, by suppressing T-cell responses and contributing to the tumor stroma, is suggested to have a significant impact on disease progression, metastasis formation, and therapy response. Most evidence, however, is derived from artificial models using exogenously administered MSCs. The contribution of endogenous MSCs to tumor progression is currently unclear. Furthermore, few studies have been conducted in humans. A prospective biomarker study was therefore undertaken in 40 human cancer patients and 10 healthy controls of similar age, aimed at (i) exploring and quantifying circulating MSC levels in healthy volunteers and patients with advanced malignancies, (ii) determining the variability of MSC levels between healthy volunteers and cancer patients with different histologic tumor types, and (iii) exploring biomarkers associated with MSC levels. Significantly increased levels of circulating MSC-like cells were observed in cancer patients when compared to healthy individuals (1.72 fold difference, 95% CI 1.03-2.81%, p = 0.03). In addition, prior systemic therapy was associated with a significant increase in MSC-like cells (1.73 fold difference, 95% CI 1.02-2.95, p = 0.04). These results indicate that the amount of endogenously circulating MSCs in humans is increased in response to cancer, and that systemic anticancer treatment can influence MSC levels. Further research is needed to determine whether MSCs have a predictive value.
Collapse
Affiliation(s)
- Daphne L van der Velden
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Julia M Houthuijzen
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jeanine M L Roodhart
- Division of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Erik van Werkhoven
- Biometrics Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emile E Voest
- Division of Molecular Oncology & Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Kopan C, Tucker T, Alexander M, Mohammadi MR, Pone EJ, Lakey JRT. Approaches in Immunotherapy, Regenerative Medicine, and Bioengineering for Type 1 Diabetes. Front Immunol 2018; 9:1354. [PMID: 29963051 PMCID: PMC6011033 DOI: 10.3389/fimmu.2018.01354] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Recent advances on using immune and stem cells as two-pronged approaches for type 1 diabetes mellitus (T1DM) treatment show promise for advancement into clinical practice. As T1DM is thought to arise from autoimmune attack destroying pancreatic β-cells, increasing treatments that use biologics and cells to manipulate the immune system are achieving better results in pre-clinical and clinical studies. Increasingly, focus has shifted from small molecule drugs that suppress the immune system nonspecifically to more complex biologics that show enhanced efficacy due to their selectivity for specific types of immune cells. Approaches that seek to inhibit only autoreactive effector T cells or enhance the suppressive regulatory T cell subset are showing remarkable promise. These modern immune interventions are also enabling the transplantation of pancreatic islets or β-like cells derived from stem cells. While complete immune tolerance and body acceptance of grafted islets and cells is still challenging, bioengineering approaches that shield the implanted cells are also advancing. Integrating immunotherapy, stem cell-mediated β-cell or islet production and bioengineering to interface with the patient is expected to lead to a durable cure or pave the way for a clinical solution for T1DM.
Collapse
Affiliation(s)
- Christopher Kopan
- Department of Surgery, University of California Irvine, Irvine, CA, United States
| | - Tori Tucker
- Department of Cell and Molecular Biosciences, University of California Irvine, Irvine, CA, United States
| | - Michael Alexander
- Department of Surgery, University of California Irvine, Irvine, CA, United States
| | - M. Rezaa Mohammadi
- Department of Chemical Engineering and Materials Science, University of California Irvine, Irvine, CA, United States
| | - Egest J. Pone
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, United States
| | - Jonathan Robert Todd Lakey
- Department of Surgery, University of California Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
22
|
Schug C, Sievert W, Urnauer S, Müller AM, Schmohl KA, Wechselberger A, Schwenk N, Lauber K, Schwaiger M, Multhoff G, Wagner E, Nelson PJ, Spitzweg C. External Beam Radiation Therapy Enhances Mesenchymal Stem Cell-Mediated Sodium-Iodide Symporter Gene Delivery. Hum Gene Ther 2018; 29:1287-1300. [PMID: 29724129 DOI: 10.1089/hum.2018.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The tumor-homing properties of mesenchymal stem cells (MSC) have led to their development as delivery vehicles for the targeted delivery of therapeutic genes such as the sodium-iodide symporter (NIS) to solid tumors. External beam radiation therapy may represent an ideal setting for the application of engineered MSC-based gene therapy, as tumor irradiation may enhance MSC recruitment into irradiated tumors through the increased production of select factors linked to MSC migration. In the present study, the irradiation of human liver cancer cells (HuH7; 1-10 Gy) showed a strong dose-dependent increase in steady-state mRNA levels of CXCL8, CXCL12, FGF2, PDGFB, TGFB1, THBS1, and VEGF (0-48 h), which was verified for most factors at the protein level (after 48 h). Radiation effects on directed MSC migration were tested in vitro using a live cell tracking migration assay and supernatants from control and irradiated HuH7 cells. A robust increase in mean forward migration index, mean center of mass, and mean directionality of MSCs toward supernatants was seen from irradiated as compared to non-irradiated tumor cells. Transferability of this effect to other tumor sources was demonstrated using the human breast adenocarcinoma cell line (MDA-MB-231), which showed a similar behavior to radiation as seen with HuH7 cells in quantitative polymerase chain reaction and migration assay. To evaluate this in a more physiologic in vivo setting, subcutaneously growing HuH7 xenograft tumors were irradiated with 0, 2, or 5 Gy followed by CMV-NIS-MSC application 24 h later. Tumoral iodide uptake was monitored using 123I-scintigraphy. The results showed increased tumor-specific dose-dependent accumulation of radioiodide in irradiated tumors. The results demonstrate that external beam radiation therapy enhances the migratory capacity of MSCs and may thus increase the therapeutic efficacy of MSC-mediated NIS radionuclide therapy.
Collapse
Affiliation(s)
- Christina Schug
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Wolfgang Sievert
- 2 Department of Radiation Oncology, Technische Universitaet Muenchen , Munich, Germany
| | - Sarah Urnauer
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Andrea M Müller
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kathrin A Schmohl
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Alexandra Wechselberger
- 3 Clinical Biochemistry Group, Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Nathalie Schwenk
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Kirsten Lauber
- 4 Department of Radiation Oncology, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Markus Schwaiger
- 5 Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universitaet Muenchen , Munich, Germany
| | - Gabriele Multhoff
- 2 Department of Radiation Oncology, Technische Universitaet Muenchen , Munich, Germany
| | - Ernst Wagner
- 6 Department of Pharmacy, Center of Drug Research, Pharmaceutical Biotechnology, LMU Munich, Munich, Germany
| | - Peter J Nelson
- 3 Clinical Biochemistry Group, Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| | - Christine Spitzweg
- 1 Department of Internal Medicine IV, University Hospital of Munich , LMU Munich, Munich, Germany
| |
Collapse
|
23
|
Liu L, Zhang SX, Liao W, Farhoodi HP, Wong CW, Chen CC, Ségaliny AI, Chacko JV, Nguyen LP, Lu M, Polovin G, Pone EJ, Downing TL, Lawson DA, Digman MA, Zhao W. Mechanoresponsive stem cells to target cancer metastases through biophysical cues. Sci Transl Med 2018; 9:9/400/eaan2966. [PMID: 28747514 DOI: 10.1126/scitranslmed.aan2966] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/23/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
Despite decades of effort, little progress has been made to improve the treatment of cancer metastases. To leverage the central role of the mechanoenvironment in cancer metastasis, we present a mechanoresponsive cell system (MRCS) to selectively identify and treat cancer metastases by targeting the specific biophysical cues in the tumor niche in vivo. Our MRCS uses mechanosensitive promoter-driven mesenchymal stem cell (MSC)-based vectors, which selectively home to and target cancer metastases in response to specific mechanical cues to deliver therapeutics to effectively kill cancer cells, as demonstrated in a metastatic breast cancer mouse model. Our data suggest a strong correlation between collagen cross-linking and increased tissue stiffness at the metastatic sites, where our MRCS is specifically activated by the specific cancer-associated mechano-cues. MRCS has markedly reduced deleterious effects compared to MSCs constitutively expressing therapeutics. MRCS indicates that biophysical cues, specifically matrix stiffness, are appealing targets for cancer treatment due to their long persistence in the body (measured in years), making them refractory to the development of resistance to treatment. Our MRCS can serve as a platform for future diagnostics and therapies targeting aberrant tissue stiffness in conditions such as cancer and fibrotic diseases, and it should help to elucidate mechanobiology and reveal what cells "feel" in the microenvironment in vivo.
Collapse
Affiliation(s)
- Linan Liu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Shirley X Zhang
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Wenbin Liao
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Henry P Farhoodi
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Chi W Wong
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Claire C Chen
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Aude I Ségaliny
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jenu V Chacko
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Lily P Nguyen
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Mengrou Lu
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - George Polovin
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Egest J Pone
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Timothy L Downing
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA
| | - Devon A Lawson
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Michelle A Digman
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Laboratory for Fluorescence Dynamics, University of California, Irvine, Irvine, CA 92697, USA.,Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale, New South Wales 2351, Australia
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, CA 92697, USA. .,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA.,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92697, USA.,Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
24
|
Phase I study of combined indomethacin and platinum-based chemotherapy to reduce platinum-induced fatty acids. Cancer Chemother Pharmacol 2018; 81:911-921. [PMID: 29574584 DOI: 10.1007/s00280-018-3563-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/14/2018] [Indexed: 01/29/2023]
Abstract
PURPOSE Chemotherapy-resistance remains a major obstacle to effective anti-cancer treatment. We previously showed that platinum analogs cause the release of two fatty acids. These platinum-induced fatty acids (PIFAs) induced complete chemoresistance in mice, whereas co-administration of a COX-1 inhibitor, indomethacin, prevented PIFA release and significantly enhanced chemosensitivity. To assess the safety of combining indomethacin with platinum-based chemotherapy, and to explore its efficacy and associated PIFA levels, a multi-center phase I trial was conducted. METHODS The study was comprised of two arms: oxaliplatin plus capecitabine (CAPOX, arm I) and cisplatin plus gemcitabine, capecitabine or 5FU (arm II) in patients for whom these regimens were indicated as standard care. Indomethacin was escalated from 25 to 75 mg TID, using a standard 3 × 3 design per arm, and was administered orally 8 days around chemo-infusion from cycle two onwards. PIFA levels were measured before and after treatment initiation, with and without indomethacin. RESULTS Thirteen patients were enrolled, of which ten were evaluable for safety analyses. In arm I, no dose-limiting toxicities were observed, and all indomethacin dose levels were well-tolerated. Partial responses were observed in three patients (30%). Indomethacin lowered plasma levels of 12-S-hydroxy-5,8,10-heptadecatrienoic acid (12-S-HHT), whereas 4,7,10,13-hexadecatetraenoic acid (16:4(n-3)) levels were not affected. Only one patient was included in arm II; renal toxicity led to closure of this cohort. CONCLUSIONS Combined indomethacin and CAPOX treatment is safe and reduces the concentrations of 12-S-HHT, which may be associated with improved chemosensitivity. The recommended phase II dose is 75 mg indomethacin TID given 8 days surrounding standard dosed CAPOX.
Collapse
|
25
|
Luo D, Hu S, Tang C, Liu G. Mesenchymal stem cells promote cell invasion and migration and autophagy-induced epithelial-mesenchymal transition in A549 lung adenocarcinoma cells. Cell Biochem Funct 2018; 36:88-94. [PMID: 29372557 DOI: 10.1002/cbf.3320] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/16/2017] [Accepted: 12/26/2017] [Indexed: 01/06/2023]
Abstract
Mesenchymal stem cells (MSCs) are recruited into the tumour microenvironment and promote tumour growth and metastasis. Tumour microenvironment-induced autophagy is considered to suppress primary tumour formation by impairing migration and invasion. Whether these recruited MSCs regulate tumour autophagy and whether autophagy affects tumour growth are controversial. Our data showed that MSCs promote autophagy activation, reactive oxygen species production, and epithelial-mesenchymal transition (EMT) as well as increased migration and invasion in A549 cells. Decreased expression of E-cadherin and increased expression of vimentin and Snail were observed in A549 cells cocultured with MSCs. Conversely, MSC coculture-mediated autophagy positively promoted tumour EMT. Autophagy inhibition suppressed MSC coculture-mediated EMT and reduced A549 cell migration and invasion slightly. Furthermore, the migratory and invasive abilities of A549 cells were additional increased when autophagy was further enhanced by rapamycin treatment. Taken together, this work suggests that microenvironments containing MSCs can promote autophagy activation for enhancing EMT; MSCs also increase the migratory and invasive abilities of A549 lung adenocarcinoma cells. Mesenchymal stem cell-containing microenvironments and MSC-induced autophagy signalling may be potential targets for blocking lung cancer cell migration and invasion.
Collapse
Affiliation(s)
- Dan Luo
- Quality Control Section, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Shiyuan Hu
- Department of Orthopedics, The Third Affiliated Hospital, Army Medical University, Chongqing, China
| | - Chunlan Tang
- Quality Control Section, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guoxiang Liu
- Department of Respiration, Army Medical University, Chongqing, China
| |
Collapse
|
26
|
You B, Shan Y, Bao L, Chen J, Yang L, Zhang Q, Zhang W, Zhang Z, Zhang J, Shi S, You Y. The biology and function of extracellular vesicles in nasopharyngeal carcinoma (Review). Int J Oncol 2017; 52:38-46. [PMID: 29138808 DOI: 10.3892/ijo.2017.4202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/02/2017] [Indexed: 11/06/2022] Open
Abstract
Extracellular vesicles are a heterogeneous group of membrane-enclosed vesicles, which play an important role in intercellular communication. Increasing number of studies have shown that tumor-derived extracellular vesicles might be involved in the transfer of oncogenic cargo (proteins, lipids, messenger RNA, microRNA, non-coding RNAs and DNA) through which cancer cells could shape the tumor microenvironment and influence tumor progression. Nasopharyngeal carcinoma-derived extracellular vesicles have also reported to facilitate tumor proliferation, metastasis and immune escape. Moreover, nasopharyngeal carcinoma-derived extracellular vesicles might serve as biomarkers for early diagnosis and therapeutic targets. The present review provides information on the biological and clinical significance of extracellular vesicles in tumors, especially in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Bo You
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ying Shan
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lili Bao
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jing Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Liu Yang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qicheng Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wei Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhenxin Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jie Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Si Shi
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yiwen You
- Department of Otorhinolaryngology, Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
27
|
The Immunomodulatory Effects of Mesenchymal Stem Cell Polarization within the Tumor Microenvironment Niche. Stem Cells Int 2017; 2017:4015039. [PMID: 29181035 PMCID: PMC5664329 DOI: 10.1155/2017/4015039] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/11/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent a promising tool for cell therapy, particularly for their antitumor effects. This cell population can be isolated from multiple tissue sources and also display an innate ability to home to areas of inflammation, such as tumors. Upon entry into the tumor microenvironment niche, MSCs promote or inhibit tumor progression by various mechanisms, largely through the release of soluble factors. These factors can be immunomodulatory by activating or inhibiting both the adaptive and innate immune responses. The mechanisms by which MSCs modulate the immune response are not well understood. Because of this, the relationship between MSCs and immune cells within the tumor microenvironment niche continues to be an active area of research in order to help explain the apparent contradictory findings currently available in the literature. The ongoing research aims to enhance the potential of MSCs in future therapeutic applications.
Collapse
|
28
|
Kucerova L, Durinikova E, Toro L, Cihova M, Miklikova S, Poturnajova M, Kozovska Z, Matuskova M. Targeted antitumor therapy mediated by prodrug-activating mesenchymal stromal cells. Cancer Lett 2017; 408:1-9. [PMID: 28838843 DOI: 10.1016/j.canlet.2017.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022]
Abstract
Mesenchymal stromal cells (MSCs) were introduced as tumor-targeted vehicles suitable for delivery of the gene-directed enzyme/prodrug therapy more than 10 years ago. Over these years key properties of tumor cells and MSCs, which are crucial for the treatment efficiency, were examined; and there are some critical issues to be considered for the maximum antitumor effect. Moreover, engineered MSCs expressing enzymes capable of activating non-toxic prodrugs achieved long-term curative effect even in metastatic and hard-to-treat tumor types in pre-clinical scenario(s). These gene-modified MSCs are termed prodrug-activating MSCs throughout the text and represent promising approach for further clinical application. This review summarizes major determinants to be considered for the application of the prodrug-activating MSCs in antitumor therapy in order to maximize therapeutic efficiency.
Collapse
Affiliation(s)
- Lucia Kucerova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia.
| | - Erika Durinikova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Lenka Toro
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Marina Cihova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Svetlana Miklikova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Martina Poturnajova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Kozovska
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Miroslava Matuskova
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| |
Collapse
|
29
|
Ramamonjisoa N, Ackerstaff E. Characterization of the Tumor Microenvironment and Tumor-Stroma Interaction by Non-invasive Preclinical Imaging. Front Oncol 2017; 7:3. [PMID: 28197395 PMCID: PMC5281579 DOI: 10.3389/fonc.2017.00003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Tumors are often characterized by hypoxia, vascular abnormalities, low extracellular pH, increased interstitial fluid pressure, altered choline-phospholipid metabolism, and aerobic glycolysis (Warburg effect). The impact of these tumor characteristics has been investigated extensively in the context of tumor development, progression, and treatment response, resulting in a number of non-invasive imaging biomarkers. More recent evidence suggests that cancer cells undergo metabolic reprograming, beyond aerobic glycolysis, in the course of tumor development and progression. The resulting altered metabolic content in tumors has the ability to affect cell signaling and block cellular differentiation. Additional emerging evidence reveals that the interaction between tumor and stroma cells can alter tumor metabolism (leading to metabolic reprograming) as well as tumor growth and vascular features. This review will summarize previous and current preclinical, non-invasive, multimodal imaging efforts to characterize the tumor microenvironment, including its stromal components and understand tumor-stroma interaction in cancer development, progression, and treatment response.
Collapse
Affiliation(s)
- Nirilanto Ramamonjisoa
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
30
|
Stab BR, Martinez L, Grismaldo A, Lerma A, Gutiérrez ML, Barrera LA, Sutachan JJ, Albarracín SL. Mitochondrial Functional Changes Characterization in Young and Senescent Human Adipose Derived MSCs. Front Aging Neurosci 2016; 8:299. [PMID: 28018212 PMCID: PMC5156959 DOI: 10.3389/fnagi.2016.00299] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are highly dynamic organelles that in response to the cell's bio-energetic state continuously undergo structural remodeling fission and fusion processes. This mitochondrial dynamic activity has been implicated in cell cycle, autophagy, and age-related diseases. Adult tissue-derived mesenchymal stromal/stem cells present a therapeutic potential. However, to obtain an adequate mesenchymal stromal/stem cell number for clinical use, extensive in vitro expansion is required. Unfortunately, these cells undergo replicative senescence rapidly by mechanisms that are not well understood. Senescence has been associated with metabolic changes in the oxidative state of the cell, a process that has been also linked to mitochondrial fission and fusion events, suggesting an association between mitochondrial dynamics and senescence. In the present work, we studied the mitochondrial structural remodeling process of mesenchymal stromal/stem cells isolated from adipose tissue in vitro to determine if mitochondrial phenotypic changes were associated with mesenchymal stromal/stem cell senescence. For this purpose, mitochondrial dynamics and oxidative state of stromal/stem cell were compared between young and old cells. With increased cell passage, we observed a significant change in cell morphology that was associated with an increase in β-galactosidase activity. In addition, old cells (population doubling seven) also showed increased mitochondrial mass, augmented superoxide production, and decreased mitochondrial membrane potential. These changes in morphology were related to slightly levels increases in mitochondrial fusion proteins, Mitofusion 1 (MFN1), and Dynamin-related GTPase (OPA1). Collectively, our results showed that adipose tissue-derived MSCs at population doubling seven developed a senescent phenotype that was characterized by metabolic cell changes that can lead to mitochondrial fusion.
Collapse
Affiliation(s)
- Bernd R Stab
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Laura Martinez
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Adriana Grismaldo
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Alejandra Lerma
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - María L Gutiérrez
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad JaverianaBogotá, Colombia; Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad JaverianaBogotá, Colombia
| | - Luis A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Jhon J Sutachan
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Sonia L Albarracín
- Department of Nutrition and Biochemistry, School of Sciences, Pontificia Universidad Javeriana Bogotá, Colombia
| |
Collapse
|
31
|
Identification of two distinct mesenchymal stromal cell populations in human malignant glioma. J Neurooncol 2016; 131:245-254. [PMID: 27757723 PMCID: PMC5306185 DOI: 10.1007/s11060-016-2302-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 10/09/2016] [Indexed: 12/25/2022]
Abstract
Gene profiling has revealed that malignant gliomas can be divided into four distinct molecular subtypes, where tumors with a mesenchymal gene expression are correlated with short survival. The present investigation was undertaken to clarify whether human malignant gliomas contain endogenous mesenchymal stromal cells (MSC), fulfilling consensus criteria defined by The International Society for Cellular Therapy, recruited from the host. We found that MSC-like cells can be isolated from primary human malignant gliomas. Two distinct MSC-like cell populations, differing in their expression of the CD90 surface marker, were discovered after cell sorting. RNA sequencing revealed further genetic differences between these two cell populations and MSC-like cells lacking CD90 produced higher amounts of VEGF and PGE2 compared to cells with the true MSC phenotype, implying that the CD90− MSC-like cells most probably are more active in tumor vascularization and immunosuppression than their CD90+ counterpart. The results highlight the CD90− subpopulation as an important tumor component, however, its functional effects in glioma remains to be resolved. Using the protocols presented here, it will be possible to isolate, characterize and analyze brain tumor-derived MSC-like cells in more detail and to further test their functions in vitro and in in vivo xenograft models of glioma.
Collapse
|
32
|
Niess H, Thomas MN, Schiergens TS, Kleespies A, Jauch KW, Bruns C, Werner J, Nelson PJ, Angele MK. Genetic engineering of mesenchymal stromal cells for cancer therapy: turning partners in crime into Trojan horses. Innov Surg Sci 2016; 1:19-32. [PMID: 31579715 PMCID: PMC6753982 DOI: 10.1515/iss-2016-0005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/03/2016] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are adult progenitor cells with a high migratory and differentiation potential, which influence a broad range of biological functions in almost every tissue of the body. Among other mechanisms, MSCs do so by the secretion of molecular cues, differentiation toward more specialized cell types, or influence on the immune system. Expanding tumors also depend on the contribution of MSCs to building a supporting stroma, but the effects of MSCs appear to go beyond the mere supply of connective tissues. MSCs show targeted "homing" toward growing tumors, which is then followed by exerting direct and indirect effects on cancer cells. Several research groups have developed novel strategies that make use of the tumor tropism of MSCs by engineering them to express a transgene that enables an attack on cancer growth. This review aims to familiarize the reader with the current knowledge about MSC biology, the existing evidence for MSC contribution to tumor growth with its underlying mechanisms, and the strategies that have been developed using MSCs to deploy an anticancer therapy.
Collapse
Affiliation(s)
- Hanno Niess
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Michael N Thomas
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Tobias S Schiergens
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Axel Kleespies
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Karl-Walter Jauch
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, Hospital of the University of Cologne, Cologne, Germany
| | - Jens Werner
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| | - Peter J Nelson
- Medizinische Klinik und Poliklinik IV, Campus Innenstadt, Klinikum der Universitaet Muenchen, Arbeitsgruppe Klinische Biochemie, Munich, Germany
| | - Martin K Angele
- Department of General, Visceral, Transplantation and Vascular Surgery, Hospital of the University of Munich, Munich, Germany
| |
Collapse
|
33
|
Yang J, Miao Y, Chang Y, Zhang F, Wang Y, Zheng S. Condition medium of HepG-2 cells induces the transdifferentiation of human umbilical cord mesenchymal stem cells into cancerous mesenchymal stem cells. Am J Transl Res 2016; 8:3429-3438. [PMID: 27648133 PMCID: PMC5009395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/17/2016] [Indexed: 06/06/2023]
Abstract
This study aimed to investigate the transdifferentiation of human umbilical cord mesenchymal stem cells (hUCMSCs) into cancer-associated mesenchymal stem cells (CA-MSCs) after incubation with condition medium (CM) from liver cancer HepG-2 cells, and the biobehaviors (proliferation and migration) of these CA-MSCs were further evaluated. The supernatant of HepG-2 cells was collected and mixed with equal volume of low glucose DMEM. The resultant medium was used to treat hUCMSCs for 48 h. The expression of CA-MSCs related proteins and miR-221 was detected in cells. The supernatant of induced hUCMSCs was mixed with equal volume of high glucose DMEM, and the resultant medium was used treat HepG-2 cells for 48 h and the proliferation and migration of HepG-2 cells were evaluated. Moreover, HepG-2 cells were co-cultured with hUCMSCs and then the proliferation and migration of HepG-2 cells were assessed. After incubation with the supernatant from HepG-2 cells, hUCMSCs showed significantly elevated expression of vimentin, fibroblast activation protein (FAP) and miR-221. The supernatant of induced hUCMSCs was able to significantly increase the proliferation and migration of HepG-2 cells. Following co-culture, the proliferation and migration of HepG-2 cells increased dramatically. These findings suggest that the supernatant of HepG-2 cells is able to induce the phenotype of CA-MSCs and the supernatant of CA-MSCs may promote the proliferation and migration of HepG-2 cells. These findings provide experimental evidence for the cellular remodeling in tumor microenvironment and the safety of clinical use of hUCMSCs.
Collapse
Affiliation(s)
- Juan Yang
- Department of Gastroenterology, The Third People’s Hospital of Yunnan ProvinceKunming 650011, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical UniversityKunming 650032, China
| | - Yefei Chang
- Department of Laboratory, The Third People’s Hospital of Yunnan ProvinceKunming 650000, China
| | - Fan Zhang
- Department of Gastroenterology, The Third People’s Hospital of Yunnan ProvinceKunming 650011, China
| | - Yubo Wang
- Department of Gastroenterology, The Third People’s Hospital of Yunnan ProvinceKunming 650011, China
| | - Sheng Zheng
- Department of Gastroenterology, The Third People’s Hospital of Yunnan ProvinceKunming 650011, China
| |
Collapse
|
34
|
Hagenhoff A, Bruns CJ, Zhao Y, von Lüttichau I, Niess H, Spitzweg C, Nelson PJ. Harnessing mesenchymal stem cell homing as an anticancer therapy. Expert Opin Biol Ther 2016; 16:1079-92. [PMID: 27270211 DOI: 10.1080/14712598.2016.1196179] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) are non-hematopoietic progenitor cells that have been exploited as vehicles for cell-based cancer therapy. The general approach is based on the innate potential of adoptively applied MSC to undergo facilitated recruitment to malignant tissue. MSC from different tissue sources have been engineered using a variety of therapy genes that have shown efficacy in solid tumor models. AREAS COVERED In this review we will focus on the current developments of MSC-based gene therapy, in particular the diverse approaches that have been used for MSCs-targeted tumor therapy. We also discuss some outstanding issues and general prospects for their clinical application. EXPERT OPINION The use of modified mesenchymal stem cells as therapy vehicles for the treatment of solid tumors has progressed to the first generation of clinical trials, but the general field is still in its infancy. There are many questions that need to be addressed if this very complex therapy approach is widely applied in clinical settings. More must be understood about the mechanisms underlying tumor tropism and we need to identify the optimal source of the cells used. Outstanding issues also include the therapy transgenes used, and which tumor types represent viable targets for this therapy.
Collapse
Affiliation(s)
- Anna Hagenhoff
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Christiane J Bruns
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Yue Zhao
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Irene von Lüttichau
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Hanno Niess
- c Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery , University of Munich , Munich , Germany
| | - Christine Spitzweg
- d Department of Internal Medicine II , University of Munich , Munich , Germany
| | - Peter J Nelson
- e Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV , University of Munich , Munich , Germany
| |
Collapse
|
35
|
Shi S, Zhang Q, Xia Y, You B, Shan Y, Bao L, Li L, You Y, Gu Z. Mesenchymal stem cell-derived exosomes facilitate nasopharyngeal carcinoma progression. Am J Cancer Res 2016; 6:459-472. [PMID: 27186416 PMCID: PMC4859673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/08/2016] [Indexed: 06/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs), which are capable of differentiating into multiple cell types, are reported to exert multiple effects on tumor development. However, the relationship between MSCs and nasopharyngeal carcinoma (NPC) cells remains unclear. Exosomes are small membrane vesicles that can be released by several cell types, including MSCs. Exosomes, which can carry membrane and cytoplasmic constituents, have been described as participants in a novel mechanism of cell-to-cell communication. In the present study, we investigated the mechanisms underlying the interaction between MSCs and NPC cells. The data showed that MSCs secreted 40-100 nm heterogeneous small vesicles, which were defined as exosomes. Incubation of NPC cells with MSC-derived exosomes resulted in the uptake of exosomes by the cells, which promoted their proliferation, migration and tumorigenesis. After an extended treatment duration, the tumor cells showed morphological changes and significant changes in the expression of epithelial-mesenchymal transition (EMT) markers. Moreover, we found that FGF19 was highly expressed in MSC-exosomes and that exosomes stimulated NPC progression by activating the FGF19-FGFR4-dependent ERK signaling cascade and by modulating the EMT. All of these data indicated that exosomes participate in a novel mechanism by which MSCs influence NPC progression.
Collapse
Affiliation(s)
- Si Shi
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Qicheng Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yunfei Xia
- Department of Rheumatology, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Bo You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Ying Shan
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Lili Bao
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Li Li
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Yiwen You
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong UniversityNantong, Jiangsu Province, China
| |
Collapse
|
36
|
Zhou K, Xia M, Tang B, Yang D, Liu N, Tang D, Xie H, Wang X, Zhu H, Liu C, Zuo C. Isolation and comparison of mesenchymal stem cell‑like cells derived from human gastric cancer tissues and corresponding ovarian metastases. Mol Med Rep 2015; 13:1788-94. [PMID: 26718033 DOI: 10.3892/mmr.2015.4735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 12/02/2015] [Indexed: 11/06/2022] Open
Abstract
Mesenchymal stem cell (MSC)-like cells have been isolated from various types of tumor. It has previously been reported that MSCs are involved in tumorigenesis and its prognosis. The aim of the present study was to isolate and compare MSC-like cells from human gastric cancer (GC) and its metastatic deposits in ovarian tissue. MSC-like cells were isolated from human gastric cancer (hGC-MSCs) and the corresponding ovarian metastatic tissues (hGCOM-MSCs) from 40 patients. The characteristics of hGC-MSCs and hGCOM-MSCs, including their morphology, surface antigens, specific gene expression and differentiation potential, were similar to those of MSCs derived from human bone marrow (hBM-MSCs) but different from GC cells. In conclusion, the present study demonstrated that MSC-like cells could be isolated from GC tissue and its ovarian metastatic tissues. The existence of MSC-like cells in GC tissues and its ovarian metastatic tissues suggests that they may be a potential target for cancer therapy, and provides an experimental foundation for investigating their role in the initiation and progression of ovarian metastasis of GC.
Collapse
Affiliation(s)
- Kunyan Zhou
- Department of Gastroduodenal and Pancreatic Surgery, Laboratory of Digestive Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Man Xia
- Department of Gynaecological Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Bo Tang
- Department of Gastroduodenal and Pancreatic Surgery, Laboratory of Digestive Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Darong Yang
- Department of Gastroduodenal and Pancreatic Surgery, Laboratory of Digestive Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Nianli Liu
- Department of Molecular Medicine, College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan 410082, P.R. China
| | - Dihong Tang
- Department of Gynaecological Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Hailong Xie
- Cancer Research Institute, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaohong Wang
- Department of Gastroduodenal and Pancreatic Surgery, Laboratory of Digestive Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Haizhen Zhu
- Department of Gastroduodenal and Pancreatic Surgery, Laboratory of Digestive Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chen Liu
- Department of Pathology, Immunology and Laboratory Medicine, Shands Cancer Center, University of Florida, Gainesville, FL 32610‑0275, USA
| | - Chaohui Zuo
- Department of Gastroduodenal and Pancreatic Surgery, Laboratory of Digestive Oncology, Hunan Cancer Hospital and Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
37
|
Abstract
Compared with traditional 2D adherent cell culture, 3D spheroidal cell aggregates, or spheroids, are regarded as more physiological, and this technique has been exploited in the field of oncology, stem cell biology, and tissue engineering. Mesenchymal stem cells (MSCs) cultured in spheroids have enhanced anti-inflammatory, angiogenic, and tissue reparative/regenerative effects with improved cell survival after transplantation. Cytoskeletal reorganization and drastic changes in cell morphology in MSC spheroids indicate a major difference in mechanophysical properties compared with 2D culture. Enhanced multidifferentiation potential, upregulated expression of pluripotency marker genes, and delayed replicative senescence indicate enhanced stemness in MSC spheroids. Furthermore, spheroid formation causes drastic changes in the gene expression profile of MSC in microarray analyses. In spite of these significant changes, underlying molecular mechanisms and signaling pathways triggering and sustaining these changes are largely unknown.
Collapse
|
38
|
Mesenchymal stem cells engineered to express selectin ligands and IL-10 exert enhanced therapeutic efficacy in murine experimental autoimmune encephalomyelitis. Biomaterials 2015; 77:87-97. [PMID: 26584349 DOI: 10.1016/j.biomaterials.2015.11.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022]
Abstract
Systemic administration of mesenchymal stem cells (MSCs) affords the potential to ameliorate the symptoms of Multiple Sclerosis (MS) in both preclinical and clinical studies. However, the efficacy of MSC-based therapy for MS likely depends on the number of cells that home to inflamed tissues and on the controlled production of paracrine and immunomodulatory factors. Previously, we reported that engineered MSCs expressing P-selectin glycoprotein ligand-1 (PSGL-1) and Sialyl-Lewis(x) (SLeX) via mRNA transfection facilitated the targeted delivery of anti-inflammatory cytokine interleukin-10 (IL-10) to inflamed ear. Here, we evaluated whether targeted delivery of MSCs with triple PSGL1/SLeX/IL-10 engineering improves therapeutic outcomes in mouse experimental autoimmune encephalomyelitis (EAE), a murine model for human MS. We found PSGL-1/SLeX mRNA transfection significantly enhanced MSC homing to the inflamed spinal cord. This is consistent with results from in vitro flow chamber assays in which PSGL-1/SleX mRNA transfection significantly increased the percentage of rolling and adherent cells on activated brain microvascular endothelial cells, which mimic the inflamed endothelium of blood brain/spinal cord barrier in EAE. In addition, IL-10-transfected MSCs show significant inhibitory activity on the proliferation of CD4(+) T lymphocytes from EAE mice. In vivo treatment with MSCs engineered with PSGL-1/SLeX/IL-10 in EAE mice exhibited a superior therapeutic function over native (unmodified) MSCs, evidenced by significantly improved myelination and decreased lymphocytes infiltration into the white matter of the spinal cord. Our strategy of targeted delivery of performance-enhanced MSCs could potentially be utilized to increase the effectiveness of MSC-based therapy for MS and other central nervous system (CNS) disorders.
Collapse
|
39
|
Liu L, Zhang SX, Aeran R, Liao W, Lu M, Polovin G, Pone EJ, Zhao W. Exogenous marker-engineered mesenchymal stem cells detect cancer and metastases in a simple blood assay. Stem Cell Res Ther 2015; 6:181. [PMID: 26391980 PMCID: PMC4578609 DOI: 10.1186/s13287-015-0151-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/05/2015] [Accepted: 08/11/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) are adult multipotent stem cells that possess regenerative and immunomodulatory properties. They have been widely investigated as therapeutic agents for a variety of disease conditions, including tissue repair, inflammation, autoimmunity, and organ transplantation. Importantly, systemically infused MSCs selectively home to primary and metastatic tumors, though the molecular mechanisms of tumor tropism of MSCs remain incompletely understood. We have exploited the active and selective MSCs homing to cancer microenvironments to develop a rapid and selective blood test for the presence of cancer. Methods We tested the concept of using transplanted MSCs as the basis for a simple cancer blood test. MSCs were engineered to express humanized Gaussia luciferase (hGluc). In a minimally invasive fashion, hGluc secreted by MSCs into circulation as a reporter for cancer presence, was assayed to probe whether MSCs co-localize with and persist in cancerous tissue. Results In vitro, hGluc secreted by engineered MSCs was detected stably over a period of days in the presence of serum. In vivo imaging showed that MSCs homed to breast cancer lung metastases and persisted longer in tumor-bearing mice than in tumor-free mice (P < 0.05). hGluc activity in blood of tumor-bearing mice was significantly higher than in their tumor-free counterparts (P < 0.05). Conclusions Both in vitro and in vivo data show that MSCs expressing hGluc can identify and report small tumors or metastases in a simple blood test format. Our novel and simple stem cell-based blood test can potentially be used to screen, detect, and monitor cancer and metastasis at early stages and during treatment. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0151-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Linan Liu
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| | - Shirley X Zhang
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| | - Rangoli Aeran
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| | - Wenbin Liao
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| | - Mengrou Lu
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| | - George Polovin
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Boulevard, Long Beach, CA, 90840, USA.
| | - Egest J Pone
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| | - Weian Zhao
- Department of Pharmaceutical Sciences, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Department of Biomedical Engineering, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA. .,Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA, 92697, USA.
| |
Collapse
|
40
|
Ruvolo PP. Galectin 3 as a guardian of the tumor microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:427-437. [PMID: 26264495 DOI: 10.1016/j.bbamcr.2015.08.008] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 01/12/2023]
Abstract
Galectin 3 is a member of a family of β-galactoside binding proteins and has emerged as an important regulator of diverse functions critical in cancer biology including apoptosis, metastasis, immune surveillance, molecular trafficking, mRNA splicing, gene expression, and inflammation. Galectin 3's ability to support cancer cell survival by intra-cellular and extra-cellular mechanisms suggests this molecule is an important component of the tumor microenvironment that potentially could be targeted for therapy. Data is emerging that Galectin 3 is elevated in many cancers including solid tumors and the cancers of the blood. Galectin 3 also appears to be a key molecule produced by tumor microenvironment support cells including mesenchymal stromal cells (MSC) to suppress immune surveillance by killing T cells and interfering with NK cell function and by supporting metastasis. Levels of Galectin 3 increase in the MSC of aging mice and perhaps this contributes to the development of cancer in the elderly. Galectin 3 modulates surface protein expression of a diverse set of glycoproteins including CD44 by regulating endocytosis of these proteins. In addition, Galectin 3 binding to receptor kinases such as CD45 and the T cell receptor is critical in the regulation of their function. In this review I will examine the various mechanisms how Galectin 3 supports chemoresistance and metastasis in solid tumors and in leukemia and lymphoma. I will also discuss possible therapeutic strategies to target this Galectin for cancer therapy. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
Affiliation(s)
- Peter P Ruvolo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, United States.
| |
Collapse
|
41
|
Lai RC, Yeo RWY, Lim SK. Mesenchymal stem cell exosomes. Semin Cell Dev Biol 2015; 40:82-8. [PMID: 25765629 DOI: 10.1016/j.semcdb.2015.03.001] [Citation(s) in RCA: 381] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/18/2022]
Abstract
MSCs are an extensively used cell type in clinical trials today. The initial rationale for their clinical testing was based on their differentiation potential. However, the lack of correlation between functional improvement and cell engraftment or differentiation at the site of injury has led to the proposal that MSCs exert their effects not through their differentiation potential but through their secreted product, more specifically, exosomes, a type of extracellular vesicle. We propose here that MSC exosomes function as an extension of MSC's biological role as tissue stromal support cells. Like their cell source, MSC exosomes help maintain tissue homeostasis for optimal tissue function. They target housekeeping biological processes that operate ubiquitously in all tissues and are critical in maintaining tissue homeostasis, enabling cells to recover critical cellular functions and begin repair and regeneration. This hypothesis provides a rationale for the therapeutic efficacy of MSCs and their secreted exosomes in a wide spectrum of diseases. Here, we give a brief introduction of the biogenesis of MSC exosomes, review their physiological functions and highlight some of their biochemical potential to illustrate how MSC exosomes could restore tissue homeostasis leading to tissue recovery and repair.
Collapse
Affiliation(s)
- Ruenn Chai Lai
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Ronne Wee Yeh Yeo
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
42
|
Nicolay NH, Liang Y, Perez RL, Bostel T, Trinh T, Sisombath S, Weber KJ, Ho AD, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells are resistant to carbon ion radiotherapy. Oncotarget 2015; 6:2076-87. [PMID: 25504442 PMCID: PMC4385837 DOI: 10.18632/oncotarget.2857] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/02/2015] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) participate in regeneration of tissues damaged by ionizing radiation. However, radiation can damage MSCs themselves. Here we show that cellular morphology, adhesion and migration abilities were not measurably altered by photon or carbon ion irradiation. The potential for differentiation was unaffected by either form of radiation, and established MSC surface markers were found to be stably expressed irrespective of radiation treatment. MSCs were able to efficiently repair DNA double strand breaks induced by both high-dose photon and carbon ion radiation. We have shown for the first time that MSCs are relatively resistant to therapeutic carbon ion radiotherapy. Additionally, this form of radiation did not markedly alter the defining stem cell properties or the expression of established surface markers in MSCs.
Collapse
Affiliation(s)
- Nils H. Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld, Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld, Heidelberg, Germany
| | - Yingying Liang
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld, Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld, Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld, Heidelberg, Germany
| | - Tilman Bostel
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld, Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld, Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld, Heidelberg, Germany
| | - Anthony D. Ho
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld, Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
| | - Peter E. Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld, Heidelberg, Germany
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld, Heidelberg, Germany
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld, Heidelberg, Germany
| |
Collapse
|
43
|
Eterno V, Zambelli A, Pavesi L, Villani L, Zanini V, Petrolo G, Manera S, Tuscano A, Amato A. Adipose-derived Mesenchymal Stem Cells (ASCs) may favour breast cancer recurrence via HGF/c-Met signaling. Oncotarget 2015; 5:613-33. [PMID: 24327602 PMCID: PMC3996669 DOI: 10.18632/oncotarget.1359] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Adipose tissue is a reservoir of Mesenchymal Stem Cells (Adipose-derived Mesenchymal Stem Cells, ASCs), endowed with regenerative properties. Fat graft was proposed for breast reconstruction in post-surgery cancer patients achieving good aesthetic results and tissues regeneration. However, recent findings highlight a potential tumorigenic role that ASCs may have in cancer recurrence, raising some concerns about their safety in clinical application. To address this issue, we established a model where autologous ASCs were combined with primary normal or cancer cells from breast of human donors, in order to evaluate potential effects of their interactions, in vitro and in vivo. Surprisingly, we found that ASCs are not tumorigenic per sè, as they are not able to induce a neoplastic transformation of normal mammary cells, however they could exhacerbate tumorigenic behaviour of c-Met-expressing breast cancer cells, creating an inflammatory microenvironment which sustained tumor growth and angiogenesis. Pharmacological c-Met inhibition showed that a HGF/c-Met crosstalk between ASCs and breast cancer cells enhanced tumor cells migration, acquiring a metastatic signature, and sustained tumor self-renewal. The master role of HGF/c-Met pathway in cancer recurrence was further confirmed by c-Met immunostaining in primary breast cancer from human donors, revealing a strong positivity in patients displaying a recurrent pathology after fat grafts and a weak/moderate staining in patients without signs of recurrence. Altogether our findings, for the first time, suggest c-Met expression, as predictive to evaluate risk of cancer recurrence after autologous fat graft in post-surgery breast cancer patients, increasing the safety of fat graft in clinical application.
Collapse
|
44
|
MSCs derived from iPSCs with a modified protocol are tumor-tropic but have much less potential to promote tumors than bone marrow MSCs. Proc Natl Acad Sci U S A 2014; 112:530-5. [PMID: 25548183 DOI: 10.1073/pnas.1423008112] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem or stromal cells (MSCs) have many potential therapeutic applications including therapies for cancers and tissue damages caused by cancers or radical cancer treatments. However, tissue-derived MSCs such as bone marrow MSCs (BM-MSCs) may promote cancer progression and have considerable donor variations and limited expandability. These issues hinder the potential applications of MSCs, especially those in cancer patients. To circumvent these issues, we derived MSCs from transgene-free human induced pluripotent stem cells (iPSCs) efficiently with a modified protocol that eliminated the need of flow cytometric sorting. Our iPSC-derived MSCs were readily expandable, but still underwent senescence after prolonged culture and did not form teratomas. These iPSC-derived MSCs homed to cancers with efficiencies similar to BM-MSCs but were much less prone than BM-MSCs to promote the epithelial-mesenchymal transition, invasion, stemness, and growth of cancer cells. The observations were probably explained by the much lower expression of receptors for interleukin-1 and TGFβ, downstream protumor factors, and hyaluronan and its cofactor TSG6, which all contribute to the protumor effects of BM-MSCs. The data suggest that iPSC-derived MSCs prepared with the modified protocol are a safer and better alternative to BM-MSCs for therapeutic applications in cancer patients. The protocol is scalable and can be used to prepare the large number of cells required for "off-the-shelf" therapies and bioengineering applications.
Collapse
|
45
|
Del Fattore A, Luciano R, Saracino R, Battafarano G, Rizzo C, Pascucci L, Alessandri G, Pessina A, Perrotta A, Fierabracci A, Muraca M. Differential effects of extracellular vesicles secreted by mesenchymal stem cells from different sources on glioblastoma cells. Expert Opin Biol Ther 2014; 15:495-504. [PMID: 25539575 DOI: 10.1517/14712598.2015.997706] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Malignant glial tumors, including glioblastoma multiforme, account for 15 - 20% of pediatric CNS malignancies. They are most resistant to therapy and are associated with a poor prognosis. OBJECTIVE Given the ability of mesenchymal stem cells (MSCs) to affect glioma growth, we investigated the effects of extracellular vesicles (EVs) derived from MSCs on U87MG glioblastoma cells line. METHODS EVs were isolated from culture media of MSCs from different sources, including bone marrow (BM), umbilical cord (UC) and adipose tissue (AT) and added to U87MG culture. The internalization and the effects of BM-, UC- and AT-MSC-EVs on proliferation and apoptosis of tumor cells were evaluated. RESULTS Both confocal microscopy and FACS analysis showed internalization of EVs into tumor cells. BM- and UC-MSC-EVs decreased cell proliferation, while an opposite effect was observed with AT-MSC-EVs. Moreover, both BM- and UC-MSC-EVs induced apoptosis of glioblastoma cells, while AT-MSC-EVs had no effect. Loading UC-MSC-EVs with Vincristine further increased cytotoxicity when compared both to the free drug and to untreated EVs. CONCLUSIONS Different effects of MSC-EVs on cancer cells were observed depending on their tissue of origin. Moreover, MSC-EVs can deliver antiblastic drugs to glioblastoma cells.
Collapse
Affiliation(s)
- Andrea Del Fattore
- Bambino Gesù Children's Hospital, Regenerative Medicine Unit, IRCCS , Piazza Sant'Onofrio 4, Rome 00165 , Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kim SH, Bang SH, Kang SY, Park KD, Eom JH, Oh IU, Yoo SH, Kim CW, Baek SY. Human amniotic membrane-derived stromal cells (hAMSC) interact depending on breast cancer cell type through secreted molecules. Tissue Cell 2014; 47:10-6. [PMID: 25441616 DOI: 10.1016/j.tice.2014.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/06/2014] [Accepted: 10/10/2014] [Indexed: 12/26/2022]
Abstract
Human amniotic membrane-derived stromal cells (hAMSC) are candidates for cell-based therapies. We examined the characteristics of hAMSC including the interaction between hAMSC and breast cancer cells, MCF-7, and MDA-MB-231. Human amniotic membrane-derived stromal cells showed typical MSC properties, including fibroblast-like morphology, surface antigen expression, and mesodermal differentiation. To investigate cell-cell interaction via secreted molecules, we cultured breast cancer cells in hAMSC-conditioned medium (hAMSC-CM) and analyzed their proliferation, migration, and secretome profiles. MCF-7 and MDA-MB-231 cells exposed to hAMSC-CM showed increased proliferation and migration. However, in hAMSC-CM, MCF-7 cells proliferated significantly faster than MDA-MB-231 cells. When cultured in hAMSC-CM, MCF-7 cells migrated faster than MDA-MB-231 cells. Two cell types showed different profiles of secreted factors. MCF-7 cells expressed much amounts of IL-8, GRO, and MCP-1 in hAMSC-CM. Human amniotic membrane-derived stromal cells interact with breast cancer cells through secreted molecules. Factors secreted by hAMSCs promote the proliferation and migration of MCF-7 breast cancer cells. For much safe cell-based therapies using hAMSC, it is necessary to study carefully about interaction between hAMSC and cancer cells.
Collapse
Affiliation(s)
- Sun-Hee Kim
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea; Laboratory of Biopharmaceutical Process, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Republic of Korea.
| | - So Hee Bang
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| | - So Yeong Kang
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| | - Ki Dae Park
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| | - Jun Ho Eom
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| | - Il Ung Oh
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| | - Si Hyung Yoo
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| | - Chan-Wha Kim
- Laboratory of Biopharmaceutical Process, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Republic of Korea
| | - Sun Young Baek
- National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Chuncheongbuk-do 363-700, Republic of Korea
| |
Collapse
|
47
|
CXXC5 (retinoid-inducible nuclear factor, RINF) is a potential therapeutic target in high-risk human acute myeloid leukemia. Oncotarget 2014; 4:1438-48. [PMID: 23988457 PMCID: PMC3824541 DOI: 10.18632/oncotarget.1195] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The retinoid-responsive gene CXXC5 localizes to the 5q31.2 chromosomal region and encodes a retinoid-inducible nuclear factor (RINF) that seems important during normal myelopoiesis. We investigated CXXC5/RINF expression in primary human acute myeloid leukemia (AML) cells derived from 594 patients, and a wide variation in CXXC5/RINF mRNA levels was observed both in the immature leukemic myeloblasts and in immature acute lymphoblastic leukemia cells. Furthermore, patients with low-risk cytogenetic abnormalities showed significantly lower levels compared to patients with high-risk abnormalities, and high RINF/CXXC5/ mRNA levels were associated with decreased overall survival for patients receiving intensive chemotherapy for newly diagnosed AML. This association with prognosis was seen both when investigating (i) an unselected patient population as well as for patients with (ii) normal cytogenetic and (iii) core-binding factor AML. CXXC5/RINF knockdown in AML cell lines caused increased susceptibility to chemotherapy-induced apoptosis, and regulation of apoptosis also seemed to differ between primary human AML cells with high and low RINF expression. The association with adverse prognosis together with the antiapoptotic effect of CXXC5/RINF suggests that targeting of CXXC5/RINF should be considered as a possible therapeutic strategy, especially in high-risk patients who show increased expression in AML cells compared with normal hematopoietic cells.
Collapse
|
48
|
Dittmer J, Leyh B. The impact of tumor stroma on drug response in breast cancer. Semin Cancer Biol 2014; 31:3-15. [PMID: 24912116 DOI: 10.1016/j.semcancer.2014.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/27/2014] [Accepted: 05/30/2014] [Indexed: 02/06/2023]
Abstract
In the last two decades the breast cancer mortality rate has steadily declined, in part, due to the availability of better treatment options. However, drug resistance still remains a major challenge. Resistance can be an inherent feature of breast cancer cells, but can also arise from the tumor microenvironment. This review aims to focus on the modulatory effect of the tumor microenvironment on the differing response of breast cancer subtypes to targeted drugs and chemotherapy.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, University of Halle, Halle/Saale, Germany.
| | - Benjamin Leyh
- Clinic for Gynecology, University of Halle, Halle/Saale, Germany
| |
Collapse
|
49
|
L. Berg E, Hsu YC, Lee JA. Consideration of the cellular microenvironment: physiologically relevant co-culture systems in drug discovery. Adv Drug Deliv Rev 2014; 69-70:190-204. [PMID: 24524933 DOI: 10.1016/j.addr.2014.01.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/16/2014] [Accepted: 01/28/2014] [Indexed: 01/15/2023]
Abstract
There is renewed interest in phenotypic approaches to drug discovery, using cell-based assays to select new drugs, with the goal of improving pharmaceutical success. Assays that are more predictive of human biology can help researchers achieve this goal. Primary cells are more physiologically relevant to human biology and advances are being made in methods to expand the available cell types and improve the potential clinical translation of these assays through the use of co-cultures or three-dimensional (3D) technologies. Of particular interest are assays that may be suitable for industrial scale drug discovery. Here we review the use of primary human cells and co-cultures in drug discovery and describe the characteristics of co-culture models for inflammation biology (BioMAP systems), neo-vascularization and tumor microenvironments. Finally we briefly describe technical trends that may enable and impact the development of physiologically relevant co-culture assays in the near future.
Collapse
|
50
|
Leatherman J. Stem cells supporting other stem cells. Front Genet 2013; 4:257. [PMID: 24348512 PMCID: PMC3847370 DOI: 10.3389/fgene.2013.00257] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 11/11/2013] [Indexed: 01/03/2023] Open
Abstract
Adult stem cell therapies are increasingly prevalent for the treatment of damaged or diseased tissues, but most of the improvements observed to date are attributed to the ability of stem cells to produce paracrine factors that have a trophic effect on existing tissue cells, improving their functional capacity. It is now clear that this ability to produce trophic factors is a normal and necessary function for some stem cell populations. In vivo adult stem cells are thought to self-renew due to local signals from the microenvironment where they live, the niche. Several niches have now been identified which harbor multiple stem cell populations. In three of these niches - the Drosophila testis, the bulge of the mammalian hair follicle, and the mammalian bone marrow - one type of stem cell has been found to produce factors that contribute to the maintenance of a second stem cell population in the shared niche. In this review, I will examine the architecture of these three niches and discuss the molecular signals involved. Together, these examples establish a new paradigm for stem cell behavior, that stem cells can promote the maintenance of other stem cells.
Collapse
Affiliation(s)
- Judith Leatherman
- School of Biological Sciences, University of Northern Colorado Greeley, CO, USA
| |
Collapse
|