1
|
Wang Z, Kregel M, Meijers JL, Franch J, Cuijpers VMJI, Ahlers D, Karst U, Slootweg P, van der Geest IC, Leeuwenburgh SC, van den Beucken JJ. Cisplatin-functionalized dual-functional bone substitute granules for bone defect treatment after bone tumor resection. Acta Biomater 2025; 191:158-176. [PMID: 39551330 DOI: 10.1016/j.actbio.2024.11.020] [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: 08/12/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Invasive bone tumors pose a significant healthcare challenge, often requiring systemic chemotherapy and limb salvage surgery. However, these strategies are hampered by severe side effects, complex post-resection bone defects, and high local recurrence rates. To address this, we developed dual-functional bone substitute biomaterials by functionalizing commercially available bone substitute granules (Bio-Oss® and MBCP®+) with the established anticancer agent cisplatin. Physicochemical characterization revealed that Bio-Oss® granules possess a higher surface area and lower crystallinity compared to MBCP®+ granules, which enhances their capacity for cisplatin adsorption and release. In co-cultures with metastatic breast and prostate cancer cells (MDA-MB-231 and PC3) and bone marrow stromal cells (hBMSCs), cisplatin-functionalized granules and their releasates exhibited dose-dependent cytotoxic effects on cancer cells while having less impact on hBMSCs. Furthermore, investigations on the mechanism of action indicated that cisplatin induced significant cell cycle arrest and apoptosis in MDA-MB-231 and PC3 cells, contrasting with minimal effects on hBMSCs. In a rat femoral condyle defect model, cisplatin-functionalized granules did not evoke adverse effects on bone tissue ingrowth or new bone formation. Importantly, local application of cisplatin-functionalized granules resulted in negligible cisplatin accumulation without signs of apoptotic damage in kidneys and livers. Taken together, we here provide hard evidence that cisplatin-functionalized granules maintain a favorable balance between biosafety, anticancer efficacy, and bone regenerative capacity. Consequently, loading granular bone substitutes with cisplatin holds promise for local treatment of bone defects following bone tumor resections, presenting a safe and potentially more effective alternative to systemic cisplatin administration. STATEMENT OF SIGNIFICANCE: Current treatments in combating malignant bone tumors are hampered by severe side effects, high local tumor recurrence, and complex bone defects after surgery. This study explores a facile manufacturing method to render two types of commercially available bone substitute granules (Bio-Oss® and MBCP®+) suitable for local delivery of cisplatin. The use of cisplatin-functionalized granules has shown promising results both in killing cancer cells in a dose-dependent manner and in aiding bone regeneration. Importantly, this local treatment strategy avoids the systemic toxicity associated with traditional chemotherapy to excretory organs. This dual-functional strategy represents a significant advancement in bone cancer treatment, offering a safe and more efficient alternative that could improve outcomes for patients following bone tumor resection.
Collapse
Affiliation(s)
- Zhule Wang
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Mark Kregel
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jean-Luc Meijers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - Jordi Franch
- Department of Small Animal Medicine and Surgery, Veterinary School, Universitat Autonoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Vincent M J I Cuijpers
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands
| | - David Ahlers
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Piet Slootweg
- Department of Pathology, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Ingrid Cm van der Geest
- Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands; Department of Orthopedics, Radboudumc, Geert Grooteplein Zuid 10, Nijmegen, the Netherlands
| | - Sander Cg Leeuwenburgh
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands
| | - Jeroen Jjp van den Beucken
- Dentistry - Regenerative Biomaterials, Radboudumc, Philips van Leydenlaan 25, 6525 EX Nijmegen, the Netherlands; Radboud Institute for Medical Innovation, Radboudumc, Geert Grooteplein 21, 6525 EZ Nijmegen, the Netherlands.
| |
Collapse
|
2
|
Strack M, Kückelhaus J, Diebold M, Wuchter P, Huber PE, Schnell O, Sankowski R, Prinz M, Grosu AL, Heiland DH, Nicolay NH, Rühle A. Effects of tumor treating fields (TTFields) on human mesenchymal stromal cells. J Neurooncol 2024; 169:329-340. [PMID: 38900237 PMCID: PMC11341748 DOI: 10.1007/s11060-024-04740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE Mesenchymal stromal cells (MSCs) within the glioblastoma microenvironment have been shown to promote tumor progression. Tumor Treating Fields (TTFields) are alternating electric fields with low intensity and intermediate frequency that exhibit anti-tumorigenic effects. While the effects of TTFields on glioblastoma cells have been studied previously, nothing is known about the influence of TTFields on MSCs. METHODS Single-cell RNA sequencing and immunofluorescence staining were employed to identify glioblastoma-associated MSCs in patient samples. Proliferation and clonogenic survival of human bone marrow-derived MSCs were assessed after TTFields in vitro. MSC' characteristic surface marker expression was determined using flow cytometry, while multi-lineage differentiation potential was examined with immunohistochemistry. Apoptosis was quantified based on caspase-3 and annexin-V/7-AAD levels in flow cytometry, and senescence was assessed with ß-galactosidase staining. MSCs' migratory potential was evaluated with Boyden chamber assays. RESULTS Single-cell RNA sequencing and immunofluorescence showed the presence of glioblastoma-associated MSCs in patient samples. TTFields significantly reduced proliferation and clonogenic survival of human bone marrow-derived MSCs by up to 60% and 90%, respectively. While the characteristic surface marker expression and differentiation capacity were intact after TTFields, treatment resulted in increased apoptosis and senescence. Furthermore, TTFields significantly reduced MSCs' migratory capacity. CONCLUSION We could demonstrate the presence of tumor-associated MSCs in glioblastoma patients, providing a rationale to study the impact of TTFields on MSCs. TTFields considerably increase apoptosis and senescence in MSCs, resulting in impaired survival and migration. The results provide a basis for further analyses on the role of MSCs in glioblastoma patients receiving TTFields.
Collapse
Affiliation(s)
- Maren Strack
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany
| | - Jan Kückelhaus
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Diebold
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Neurology and Medical Oncology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, German Red Cross Blood Service Baden- Württemberg- Hessen, Heidelberg University, Mannheim, Germany
| | - Peter E Huber
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital Center Heidelberg, Heidelberg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
- Department of Radiation Oncology, University of Leipzig Medical Center, Leipzig, Germany
- Comprehensive Cancer Center Central (CCCG) Germany, Partner Site Leipzig, Leipzig, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany.
- Department of Radiation Oncology, University of Leipzig Medical Center, Leipzig, Germany.
- Comprehensive Cancer Center Central (CCCG) Germany, Partner Site Leipzig, Leipzig, Germany.
| |
Collapse
|
3
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Blitzer GC, Paz C, Glassey A, Ganz OR, Giri J, Pennati A, Meyers RO, Bates AM, Nickel KP, Weiss M, Morris ZS, Mattison RJ, McDowell KA, Croxford E, Chappell RJ, Glazer TA, Rogus-Pulia NM, Galipeau J, Kimple RJ. Functionality of bone marrow mesenchymal stromal cells derived from head and neck cancer patients - A FDA-IND enabling study regarding MSC-based treatments for radiation-induced xerostomia. Radiother Oncol 2024; 192:110093. [PMID: 38224919 PMCID: PMC10922976 DOI: 10.1016/j.radonc.2024.110093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
PURPOSE Salivary dysfunction is a significant side effect of radiation therapy for head and neck cancer (HNC). Preliminary data suggests that mesenchymal stromal cells (MSCs) can improve salivary function. Whether MSCs from HNC patients who have completed chemoradiation are functionally similar to those from healthy patients is unknown. We performed a pilot clinical study to determine whether bone marrow-derived MSCs [MSC(M)] from HNC patients could be used for the treatment of RT-induced salivary dysfunction. METHODS An IRB-approved pilot clinical study was undertaken on HNC patients with xerostomia who had completed treatment two or more years prior. Patients underwent iliac crest bone marrow aspirate and MSC(M) were isolated and cultured. Culture-expanded MSC(M) were stimulated with IFNγ and cryopreserved prior to reanimation and profiling for functional markers by flow cytometry and ELISA. MSC(M) were additionally injected into mice with radiation-induced xerostomia and the changes in salivary gland histology and salivary production were examined. RESULTS A total of six subjects were enrolled. MSC(M) from all subjects were culture expanded to > 20 million cells in a median of 15.5 days (range 8-20 days). Flow cytometry confirmed that cultured cells from HNC patients were MSC(M). Functional flow cytometry demonstrated that these IFNγ-stimulated MSC(M) acquired an immunosuppressive phenotype. IFNγ-stimulated MSC(M) from HNC patients were found to express GDNF, WNT1, and R-spondin 1 as well as pro-angiogenesis and immunomodulatory cytokines. In mice, IFNγ-stimulated MSC(M) injection after radiation decreased the loss of acinar cells, decreased the formation of fibrosis, and increased salivary production. CONCLUSIONS MSC (M) from previously treated HNC patients can be expanded for auto-transplantation and are functionally active. Furthermore IFNγ-stimulated MSC(M) express proteins implicated in salivary gland regeneration. This study provides preliminary data supporting the feasibility of using autologous MSC(M) from HNC patients to treat RT-induced salivary dysfunction.
Collapse
Affiliation(s)
- Grace C Blitzer
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Cristina Paz
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Annemarie Glassey
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Olga R Ganz
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Jayeeta Giri
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Andrea Pennati
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ross O Meyers
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Amber M Bates
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kwangok P Nickel
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Marissa Weiss
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Zachary S Morris
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ryan J Mattison
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kimberly A McDowell
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Emma Croxford
- Department of Biostatistics and Medical Informatics, 610 Walnut Street, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726 USA
| | - Richard J Chappell
- Department of Biostatistics and Medical Informatics, 610 Walnut Street, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Tiffany A Glazer
- Department of Surgery, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Nicole M Rogus-Pulia
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; Geriatric Research Education and Clinical Center, 2500 Overlook Terrace, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Jacques Galipeau
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Randall J Kimple
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA.
| |
Collapse
|
5
|
Baron M, Drohat P, Crawford B, Hornicek FJ, Best TM, Kouroupis D. Mesenchymal Stem/Stromal Cells: Immunomodulatory and Bone Regeneration Potential after Tumor Excision in Osteosarcoma Patients. Bioengineering (Basel) 2023; 10:1187. [PMID: 37892917 PMCID: PMC10604230 DOI: 10.3390/bioengineering10101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Osteosarcoma (OS) is a type of bone cancer that is derived from primitive mesenchymal cells typically affecting children and young adults. The current standard of treatment is a combination of neoadjuvant chemotherapy and surgical resection of the cancerous bone. Post-resection challenges in bone regeneration arise. To determine the appropriate amount of bone to be removed, preoperative imaging techniques such as bone and CT scans are employed. To prevent local recurrence, the current standard of care suggests maintaining bony and soft tissue margins from 3 to 7 cm beyond the tumor. The amount of bone removed in an OS patient leaves too large of a deficit for bone to form on its own and requires reconstruction with metal implants or allografts. Both methods require the bone to heal, either to the implant or across the allograft junction, often in the setting of marrow-killing chemotherapy. Therefore, the issue of bone regeneration within the surgically resected margins remains an important challenge for the patient, family, and treating providers. Mesenchymal stem/stromal cells (MSCs) are potential agents for enhancing bone regeneration post tumor resection. MSCs, used with scaffolds and growth factors, show promise in fostering bone regeneration in OS cases. We spotlight two MSC types-bone marrow-derived (BM-MSCs) and adipose tissue-derived (ASCs)-highlighting their bone regrowth facilitation and immunomodulatory effects on immune cells like macrophages and T cells, enhancing therapeutic outcomes. The objective of this review is two-fold: review work demonstrating any ability of MSCs to target the deranged immune system in the OS microenvironment, and synthesize the available literature on the use of MSCs as a therapeutic option for stimulating bone regrowth in OS patients post bone resection. When it comes to repairing bone defects, both MB-MSCs and ASCs hold great potential for stimulating bone regeneration. Research has showcased their effectiveness in reconstructing bone defects while maintaining a non-tumorigenic role following wide resection of bone tumors, underscoring their capability to enhance bone healing and regeneration following tumor excisions.
Collapse
Affiliation(s)
- Max Baron
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
| | - Philip Drohat
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
| | - Brooke Crawford
- Sarcoma Biology Laboratory, Department of Orthopedics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (B.C.); (F.J.H.)
| | - Francis J. Hornicek
- Sarcoma Biology Laboratory, Department of Orthopedics, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (B.C.); (F.J.H.)
| | - Thomas M. Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.B.); (P.D.); (T.M.B.)
- Diabetes Research Institute, Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
6
|
Altalhi SA, Eldin I Elbehairi S, Alfaifi MY, Al-Salmi FA, Shati AA, Alqahtani LS, Fayad E, F M Elshaarawy R, Nasr AM. Therapeutic potential and protection enhancement of mesenchymal stem cell against cisplatin-induced nephrotoxicity using hyaluronic acid-chitosan nanoparticles as an adjuvant. Int J Pharm 2023; 640:123023. [PMID: 37150270 DOI: 10.1016/j.ijpharm.2023.123023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
A newly synthesized nanoplatform of hyaluronic acid and chitosan nanoparticles (HA/CNPs) was applied to improve the therapeutic efficacy and protection of bone marrow mesenchymal stem cells (BM-MSCs) against cisplatin (CDDP)-induced nephrotoxicity in rats. CDDP administration causes significant increases in levels of serum creatinine (SCr), urea, and KIM-1 coupled with significant albumin level falls, as indicative of acute renal dysfunction. Moreover, the level of the antioxidant enzyme (GSH) was significantly decreased, while the levels of lipid peroxidation (MDA) and inflammatory (IL-6) and apoptotic (caspase-3) markers were significantly increased, indicating a decline in the kidney's antioxidant defense and increased inflammation. In contrast, when rats were pre-treated with either MSCs or MSCs-HA/CNPs before receiving CDDP, the levels of SCr, urea, KIM-1, MDA, IL-6, and caspase-3 were significantly decreased with simultaneous significant rises in GSH and albumin, impelling a great improvement in the antioxidant and anti-inflammatory defenses of the kidney as well as its functions. Intriguingly, MSCs-HA/CNPs were more effective against caspase-3 than MSCs alone, revealing the high anti-apoptotic capability of HA/CNPs. This finding suggests that HA/CNPs could effectively protect MSCs from oxidative stress and apoptosis and thus increase their stability and longevity.
Collapse
Affiliation(s)
- Sarah A Altalhi
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Serag Eldin I Elbehairi
- Biology Department, Faculty of Science, King Khalid University, 9004 Abha, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt.
| | - Mohammad Y Alfaifi
- Biology Department, Faculty of Science, King Khalid University, 9004 Abha, Saudi Arabia.
| | - Fawziah A Al-Salmi
- Biology Department, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ali A Shati
- Biology Department, Faculty of Science, King Khalid University, 9004 Abha, Saudi Arabia.
| | - Leena S Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 23445, Saudi Arabia.
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University, 43533 Suez, Egypt; Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf, Düsseldorf, Germany.
| | - Ali M Nasr
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, 42526 Port Said, Egypt.
| |
Collapse
|
7
|
Gebraad A, Ohlsbom R, Miettinen JJ, Emeh P, Pakarinen TK, Manninen M, Eskelinen A, Kuismanen K, Slipicevic A, Lehmann F, Nupponen NN, Heckman CA, Miettinen S. Growth Response and Differentiation of Bone Marrow-Derived Mesenchymal Stem/Stromal Cells in the Presence of Novel Multiple Myeloma Drug Melflufen. Cells 2022; 11:cells11091574. [PMID: 35563880 PMCID: PMC9103864 DOI: 10.3390/cells11091574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are self-renewing and multipotent progenitors, which constitute the main cellular compartment of the bone marrow stroma. Because MSCs have an important role in the pathogenesis of multiple myeloma, it is essential to know if novel drugs target MSCs. Melflufen is a novel anticancer peptide–drug conjugate compound for patients with relapsed refractory multiple myeloma. Here, we studied the cytotoxicity of melflufen, melphalan and doxorubicin in healthy human bone marrow-derived MSCs (BMSCs) and how these drugs affect BMSC proliferation. We established co-cultures of BMSCs with MM.1S myeloma cells to see if BMSCs increase or decrease the cytotoxicity of melflufen, melphalan, bortezomib and doxorubicin. We evaluated how the drugs affect BMSC differentiation into adipocytes and osteoblasts and the BMSC-supported formation of vascular networks. Our results showed that BMSCs were more sensitive to melflufen than to melphalan. The cytotoxicity of melflufen in myeloma cells was not affected by the co-culture with BMSCs, as was the case for melphalan, bortezomib and doxorubicin. Adipogenesis, osteogenesis and BMSC-mediated angiogenesis were all affected by melflufen. Melphalan and doxorubicin affected BMSC differentiation in similar ways. The effects on adipogenesis and osteogenesis were not solely because of effects on proliferation, seen from the differential expression of differentiation markers normalized by cell number. Overall, our results indicate that melflufen has a significant impact on BMSCs, which could possibly affect therapy outcome.
Collapse
Affiliation(s)
- Arjen Gebraad
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (R.O.); (P.E.); (S.M.)
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
- Correspondence:
| | - Roope Ohlsbom
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (R.O.); (P.E.); (S.M.)
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Juho J. Miettinen
- Institute for Molecular Medicine Finland-FIMM, HiLIFE–Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland; (J.J.M.); (C.A.H.)
| | - Promise Emeh
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (R.O.); (P.E.); (S.M.)
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Toni-Karri Pakarinen
- Department of Musculoskeletal Diseases, Tampere University Hospital, 33520 Tampere, Finland;
| | | | - Antti Eskelinen
- Coxa Hospital for Joint Replacement, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Kirsi Kuismanen
- Department of Obstetrics and Gynecology, Tampere University Hospital, 33520 Tampere, Finland;
| | - Ana Slipicevic
- Oncopeptides AB, 111 37 Stockholm, Sweden; (A.S.); (F.L.); (N.N.N.)
| | - Fredrik Lehmann
- Oncopeptides AB, 111 37 Stockholm, Sweden; (A.S.); (F.L.); (N.N.N.)
| | - Nina N. Nupponen
- Oncopeptides AB, 111 37 Stockholm, Sweden; (A.S.); (F.L.); (N.N.N.)
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland-FIMM, HiLIFE–Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00290 Helsinki, Finland; (J.J.M.); (C.A.H.)
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (R.O.); (P.E.); (S.M.)
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| |
Collapse
|
8
|
Liu H, Deng S, Han L, Ren Y, Gu J, He L, Liu T, Yuan ZX. Mesenchymal stem cells, exosomes and exosome-mimics as smart drug carriers for targeted cancer therapy. Colloids Surf B Biointerfaces 2021; 209:112163. [PMID: 34736220 DOI: 10.1016/j.colsurfb.2021.112163] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells with the capacity to differentiate into several cell types under appropriate conditions. They also possess remarkable antitumor features that make them a novel choice to treat cancers. Accumulating evidence suggest that the MSCs-derived extracellular vesicles, known as exosomes, play an essential role in the therapeutic effects of MSCs mainly by carrying biologically active factors. However, limitations such as low yield of exosomes and difficulty in isolation and purification hinder their clinical applications. To overcome these issues, research on development of exosome-mimics has attracted great attention. This systematic review represents, to the best of our knowledge, the first thorough evaluations of the innate antineoplastic features of MSCs-derived exosomes or exosome-mimics, the methods of drug loading, application as drug delivery system and their impacts on targeted cancer therapy. Importantly, we dissect the commonalities and differences as well as address the shortcomings of work accumulated over the last two decades and discuss how this information can serve as a guide map for optimal experimental design implementation ultimately aiding the effective transition into clinical trials.
Collapse
Affiliation(s)
- Hongmei Liu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Shichen Deng
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Yan Ren
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, Australia.
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
9
|
Lim SK, Khoo BY. An overview of mesenchymal stem cells and their potential therapeutic benefits in cancer therapy. Oncol Lett 2021; 22:785. [PMID: 34594426 PMCID: PMC8456491 DOI: 10.3892/ol.2021.13046] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
There has been increased interest in using stem cells for regenerative medicine and cancer therapy in the past decade. Mesenchymal stem cells (MSCs) are among the most studied stem cells due to their unique characteristics, such as self-renewal and developmental potency to differentiate into numerous cell types. MSC use has fewer ethical challenges compared with other types of stem cells. Although a number of studies have reported the beneficial effects of MSC-based therapies in treating various diseases, their contribution to cancer therapy remains controversial. The behaviour of MSCs is determined by the interaction between intrinsic transcriptional genes and extrinsic environmental factors. Numerous studies continue to emerge, as there is no denying the potential of MSCs to treat a wide variety of human afflictions. Therefore, the present review article provided an overview of MSCs and their differences compared with embryonic stem cells, and described the molecular mechanisms involved in maintaining their stemness. In addition, the article examined the therapeutic application of stem cells in the field of cancer. The present article also discussed the current divergent roles of MSCs in cancer therapy and the future potential in this field.
Collapse
Affiliation(s)
- Shern Kwok Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Boon Yin Khoo
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| |
Collapse
|
10
|
Karimi-Shahri M, Javid H, Sharbaf Mashhad A, Yazdani S, Hashemy SI. Mesenchymal stem cells in cancer therapy; the art of harnessing a foe to a friend. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1307-1323. [PMID: 35096289 PMCID: PMC8769515 DOI: 10.22038/ijbms.2021.58227.12934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/04/2021] [Indexed: 12/09/2022]
Abstract
For a long time, mesenchymal stem cells (MSCs) were discussed only as stem cells which could give rise to different types of cells. However, when it became clear that their presence in the tumor microenvironment (TME) was like a green light for tumorigenesis, they emerged from the ashes. This review was arranged to provide a comprehensive and precise description of MSCs' role in regulating tumorigenesis and to discuss the dark and the bright sides of cancer treatment strategies using MSCs. To gather the details about MSCs, we made an intensive literature review using keywords, including MSCs, tumor microenvironment, tumorigenesis, and targeted therapy. Through transferring cytokines, growth factors, and microRNAs, MSCs maintain the cancer stem cell population, increase angiogenesis, provide a facility for cancer metastasis, and shut down the anti-tumor activity of the immune system. Although MSCs progress tumorigenesis, there is a consensus that these cells could be used as a vehicle to transfer anti-cancer agents into the tumor milieu. This feature opened a new chapter in MSCs biology, this time from the therapeutic perspective. Although the data are not sufficient, the advent of new genetic engineering methods might make it possible to engage these cells as Trojan horses to eliminate the malignant population. So many years of investigation showed that MSCs are an important group of cells, residing in the TME, studying the function of which not only could add a delicate series of information to the process of tumorigenesis but also could revolutionize cancer treatment strategies.
Collapse
Affiliation(s)
- Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Sharbaf Mashhad
- Department of Medical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shaghayegh Yazdani
- Department of Medical Laboratory Sciences, Ilam Institute for Medical Sciences, Ilam, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Hamilton G, Teufelsbauer M. Adipose-derived stromal/stem cells and extracellular vesicles for cancer therapy. Expert Opin Biol Ther 2021; 22:67-78. [PMID: 34236014 DOI: 10.1080/14712598.2021.1954156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Mesenchymal stromal/stem cells (MSCs) hold great perspective for the therapy of a host of diseases due to regenerative and anti-inflammatory properties by differentiation into diverse cell populations, homing to damaged tissue regions, paracrine effects, and release of extracellular vesicles. AREAS COVERED This review describes the isolation, characterization, and potential use of MSCs and ADSCs for benign and malignant diseases. The MSCs may be administered as whole cells or in form of their secretome that is held responsible for most of their beneficial effects. A special constituent of the paracrine components are the extracellular vesicles (EVs) that carry a biologically potent cargo of proteins, cytokines, and RNA. EXPERT OPINION The applications of MSCs and ADSCs are amply documented and have been investigated in preclinical models and many unregulated and a few controlled trials. Larger numbers of MSCs and ADSCs can be obtained for allogeneic transfer but imply difficulties including perseverance of the cells in vivo and possible differentiation into harmful cell types. MSC-derived cell-free preparations are easier to handle and manufacture for various applications. Especially, with the help of bioreactors, EVs can be obtained in excessive numbers and preloaded or charged with proteins, cytokines, and regulatory RNA specimen to treat inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria.,Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Maryana Teufelsbauer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria.,Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Tsai SCS, Lin FCF, Chang KH, Li MC, Chou RH, Huang MY, Chen YC, Kao CY, Cheng CC, Lin HC, Hsu YC. The intravenous administration of skin-derived mesenchymal stem cells ameliorates hearing loss and preserves cochlear hair cells in cisplatin-injected mice: SMSCs ameliorate hearing loss and preserve outer hair cells in mice. Hear Res 2021; 413:108254. [PMID: 34020824 DOI: 10.1016/j.heares.2021.108254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 03/12/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) can be isolated from different tissue origins, such as the bone marrow, the placenta, the umbilical cord, adipose tissues, and skin tissues. MSCs can secrete anti-inflammatory molecules and growth factors for tissue repair and remodeling. However, the ability of skin-derived MSCs (SMSCs) to repair cochlear damage and ameliorate hearing loss remains unclear. Cisplatin is a commonly used chemotherapeutic agent that has the side effect of ototoxicity due to inflammation and oxidative stress. This study investigated the effects of SMSCs on cisplatin-induced hearing loss in mice. Two independent experiments were designed for modeling cisplatin-induced hearing loss in mice, one for chronic toxicity (4 mg/kg intraperitoneal [IP] injection once per day for 5 consecutive days) and the other for acute toxicity (25 mg/kg IP injection once on day one). Three days after cisplatin injection, 1 × 106 or 3 × 106 SMSCs were injected through the tail vein. Data on auditory brain responses suggested that SMSCs could significantly reduce the hearing threshold of cisplatin-injected mice. Furthermore, immunohistochemical staining data suggested that SMSCs could significantly ameliorate the loss of cochlear hair cells, TUNEL-positive cells and cleaved caspase 3-positive cells in cisplatin-injected mice. Neuropathological gene analyses revealed that SMSCs treatment could downregulate the expression of cochlear genes involved in apoptosis, autophagy, chromatin modification, disease association, matrix remodeling, oxidative stress, tissue integrity, transcription, and splicing and unfolded protein responses. Additionally, SMSCs treatment could upregulate the expression of cochlear genes affecting the axon and dendrite structures, cytokines, trophic factors, the neuronal skeleton and those involved in carbohydrate metabolism, growth factor signaling, myelination, neural connectivity, neural transmitter release, neural transmitter response and reuptake, neural transmitter synthesis and storage, and vesicle trafficking. Results from TUNEL and caspase 3 staining further confirmed that cisplatin-induced apoptosis in cochlear tissues of cisplatin-injected mice could be reduced by SMSCs treatment. In conclusion, the evidence of the effects of SMSCs in favor of ameliorating ototoxicity-induced hearing loss suggests a potential clinical application.
Collapse
Affiliation(s)
- Stella Chin-Shaw Tsai
- Department of Otolaryngology, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan
| | | | - Kuang-Hsi Chang
- Department of Medical Research, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; General Education Center, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Min-Chih Li
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Ruey-Hwang Chou
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Mei-Yue Huang
- Maria Von Med-Biotechnology Co. Ltd., Taipei, Taiwan
| | | | - Chien-Yu Kao
- Medical and Pharmaceutical Industry Technology and Development Center, Taipei, Taiwan
| | - Ching-Chang Cheng
- Laboratory Animal Service Center, Office of Research and Development, China Medical University, Taiwan
| | - Hung-Ching Lin
- Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City, Taiwan; Department of Otolaryngology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yi-Chao Hsu
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan; Department of Audiology and Speech-Language Pathology, Mackay Medical College, New Taipei City, Taiwan.
| |
Collapse
|
13
|
Chugh RM, Bhanja P, Norris A, Saha S. Experimental Models to Study COVID-19 Effect in Stem Cells. Cells 2021; 10:E91. [PMID: 33430424 PMCID: PMC7827246 DOI: 10.3390/cells10010091] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
The new strain of coronavirus (severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2)) emerged in 2019 and hence is often referred to as coronavirus disease 2019 (COVID-19). This disease causes hypoxic respiratory failure and acute respiratory distress syndrome (ARDS), and is considered as the cause of a global pandemic. Very limited reports in addition to ex vivo model systems are available to understand the mechanism of action of this virus, which can be used for testing of any drug efficacy against virus infectivity. COVID-19 induces tissue stem cell loss, resulting inhibition of epithelial repair followed by inflammatory fibrotic consequences. Development of clinically relevant models is important to examine the impact of the COVID-19 virus in tissue stem cells among different organs. In this review, we discuss ex vivo experimental models available to study the effect of COVID-19 on tissue stem cells.
Collapse
Affiliation(s)
- Rishi Man Chugh
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.M.C.); (P.B.)
| | - Payel Bhanja
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.M.C.); (P.B.)
| | - Andrew Norris
- BCN Bio Sciences, Pasadena, CA 91107, USA;
- David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Subhrajit Saha
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (R.M.C.); (P.B.)
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Pawitan JA, Bui TA, Mubarok W, Antarianto RD, Nurhayati RW, Dilogo IH, Oceandy D. Enhancement of the Therapeutic Capacity of Mesenchymal Stem Cells by Genetic Modification: A Systematic Review. Front Cell Dev Biol 2020; 8:587776. [PMID: 33195245 PMCID: PMC7661472 DOI: 10.3389/fcell.2020.587776] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022] Open
Abstract
Background The therapeutic capacity of mesenchymal stem cells (also known as mesenchymal stromal cells/MSCs) depends on their ability to respond to the need of the damaged tissue by secreting beneficial paracrine factors. MSCs can be genetically engineered to express certain beneficial factors. The aim of this systematic review is to compile and analyze published scientific literatures that report the use of engineered MSCs for the treatment of various diseases/conditions, to discuss the mechanisms of action, and to assess the efficacy of engineered MSC treatment. Methods We retrieved all published studies in PubMed/MEDLINE and Cochrane Library on July 27, 2019, without time restriction using the following keywords: “engineered MSC” and “therapy” or “manipulated MSC” and “therapy.” In addition, relevant articles that were found during full text search were added. We identified 85 articles that were reviewed in this paper. Results Of the 85 articles reviewed, 51 studies reported the use of engineered MSCs to treat tumor/cancer/malignancy/metastasis, whereas the other 34 studies tested engineered MSCs in treating non-tumor conditions. Most of the studies reported the use of MSCs in animal models, with only one study reporting a trial in human subjects. Thirty nine studies showed that the expression of beneficial paracrine factors would significantly enhance the therapeutic effects of the MSCs, whereas thirty three studies showed moderate effects, and one study in humans reported no effect. The mechanisms of action for MSC-based cancer treatment include the expression of “suicide genes,” induction of tumor cell apoptosis, and delivery of cytokines to induce an immune response against cancer cells. In the context of the treatment of non-cancerous diseases, the mechanism described in the reviewed papers included the expression of angiogenic, osteogenic, and growth factors. Conclusion The therapeutic capacity of MSCs can be enhanced by inducing the expression of certain paracrine factors by genetic modification. Genetically engineered MSCs have been used successfully in various animal models of diseases. However, the results should be interpreted cautiously because animal models might not perfectly represent real human diseases. Therefore, further studies are needed to explore the translational potential of genetically engineered MSCs.
Collapse
Affiliation(s)
- Jeanne Adiwinata Pawitan
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Stem Cell Medical Technology Integrated Service Unit, Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Thuy Anh Bui
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Wildan Mubarok
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Japan
| | - Radiana Dhewayani Antarianto
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Retno Wahyu Nurhayati
- Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ismail Hadisoebroto Dilogo
- Stem Cell Medical Technology Integrated Service Unit, Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Department of Orthopaedic and Traumatology, Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom.,Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
15
|
Babajani A, Soltani P, Jamshidi E, Farjoo MH, Niknejad H. Recent Advances on Drug-Loaded Mesenchymal Stem Cells With Anti-neoplastic Agents for Targeted Treatment of Cancer. Front Bioeng Biotechnol 2020; 8:748. [PMID: 32793565 PMCID: PMC7390947 DOI: 10.3389/fbioe.2020.00748] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs), as an undifferentiated group of adult multipotent cells, have remarkable antitumor features that bring them up as a novel choice to treat cancers. MSCs are capable of altering the behavior of cells in the tumor microenvironment, inducing an anti-inflammatory effect in tumor cells, inhibiting tumor angiogenesis, and preventing metastasis. Besides, MSCs can induce apoptosis and inhibit the proliferation of tumor cells. The ability of MSCs to be loaded with chemotherapeutic drugs and release them in the site of primary and metastatic neoplasms makes them a preferable choice as targeted drug delivery procedure. Targeted drug delivery minimizes unexpected side effects of chemotherapeutic drugs and improves clinical outcomes. This review focuses on recent advances on innate antineoplastic features of MSCs and the effect of chemotherapeutic drugs on viability, proliferation, and the regenerative capacity of various kinds of MSCs. It also discusses the efficacy and mechanisms of drug loading and releasing procedures along with in vivo and in vitro preclinical outcomes of antineoplastic effects of primed MSCs for clinical prospection.
Collapse
Affiliation(s)
- Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Soltani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Jamshidi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Farjoo
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
The Therapeutic Potential of Mesenchymal Stromal Cells in the Treatment of Chemotherapy-Induced Tissue Damage. Stem Cell Rev Rep 2020; 15:356-373. [PMID: 30937640 DOI: 10.1007/s12015-019-09886-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chemotherapy constitutes one of the key treatment modalities for solid and hematological malignancies. Albeit being an effective treatment, chemotherapy application is often limited by its damage to healthy tissues, and curative treatment options for chemotherapy-related side effects are largely missing. As mesenchymal stromal cells (MSCs) are known to exhibit regenerative capacity mainly by supporting a beneficial microenvironment for tissue repair, MSC-based therapies may attenuate chemotherapy-induced tissue injuries. An increasing number of animal studies shows favorable effects of MSC-based treatments; however, clinical trials for MSC therapies in the context of chemotherapy-related side effects are rare. In this concise review, we summarize the current knowledge of the effects of MSCs on chemotherapy-induced tissue toxicities. Both preclinical and early clinical trials investigating MSC-based treatments for chemotherapy-related side reactions are presented, and mechanistic explanations about the regenerative effects of MSCs in the context of chemotherapy-induced tissue damage are discussed. Furthermore, challenges of MSC-based treatments are outlined that need closer investigations before these multipotent cells can be safely applied to cancer patients. As any pro-tumorigenicity of MSCs needs to be ruled out prior to clinical utilization of these cells for cancer patients, the pro- and anti-tumorigenic activities of MSCs are discussed in detail.
Collapse
|
17
|
Rühle A, Thomsen A, Saffrich R, Voglstätter M, Bieber B, Sprave T, Wuchter P, Vaupel P, Huber PE, Grosu AL, Nicolay NH. Multipotent mesenchymal stromal cells are sensitive to thermic stress – potential implications for therapeutic hyperthermia. Int J Hyperthermia 2020; 37:430-441. [DOI: 10.1080/02656736.2020.1758350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Thomsen
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Maren Voglstätter
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Birgit Bieber
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanja Sprave
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter E. Huber
- Department of Molecular Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H. Nicolay
- Department of Radiation Oncology, Freiburg University Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Molecular Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
18
|
Lopez Perez R, Brauer J, Rühle A, Trinh T, Sisombath S, Wuchter P, Grosu AL, Debus J, Saffrich R, Huber PE, Nicolay NH. Human mesenchymal stem cells are resistant to UV-B irradiation. Sci Rep 2019; 9:20000. [PMID: 31882818 PMCID: PMC6934474 DOI: 10.1038/s41598-019-56591-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
Albeit being an effective therapy for various cutaneous conditions, UV-B irradiation can cause severe skin damage. While multipotent mesenchymal stem cells (MSCs) may aid the regeneration of UV-B-induced skin injuries, the influence of UV-B irradiation on MSCs remains widely unknown. Here, we show that human MSCs are relatively resistant to UV-B irradiation compared to dermal fibroblasts. MSCs exhibited higher clonogenic survival, proliferative activity and viability than dermal fibroblasts after exposure to UV-B irradiation. Cellular adhesion, morphology and expression of characteristic surface marker patterns remained largely unaffected in UV-irradiated MSCs. The differentiation ability along the adipogenic, osteogenic and chondrogenic lineages was preserved after UV-B treatment. However, UV-B radiation resulted in a reduced ability of MSCs and dermal fibroblasts to migrate. MSCs exhibited low apoptosis rates after UV-B irradiation and repaired UV-B-induced cyclobutane pyrimidine dimers more efficiently than dermal fibroblasts. UV-B irradiation led to prolonged p53 protein stability and increased p21 protein expression resulting in a prolonged G2 arrest and senescence induction in MSCs. The observed resistance may contribute to the ability of these multipotent cells to aid the regeneration of UV-B-induced skin injuries.
Collapse
Affiliation(s)
- Ramon Lopez Perez
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
| | - Jannek Brauer
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Alexander Rühle
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
- Department of Radiation Oncology, University of Freiburg - Medical Center, 79106, Freiburg, Germany
| | - Thuy Trinh
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University of Freiburg - Medical Center, 79106, Freiburg, Germany
| | - Jürgen Debus
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Peter E Huber
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular Radiation Oncology, German Cancer Research Center (dkfz), 69120, Heidelberg, Germany.
- Department of Radiation Oncology, University of Freiburg - Medical Center, 79106, Freiburg, Germany.
| |
Collapse
|
19
|
Bajelan B, Zaki-Dizaji M, Rahmani B, Darzi S, Darabi S, Rajaei F. Resistance of human primary mesenchymal stem cells to cytotoxic effects of nutlin-3 in vitro. J Cell Biochem 2019; 121:788-796. [PMID: 31452266 DOI: 10.1002/jcb.29324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/28/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND The small-molecule nutlin-3 was found to be an effective therapeutic compound and p53 activator, and acts as a murine double minute 2 antagonist, although these findings need to be clinically confirmed. The essential components of the bone marrow include mesenchymal stem cells (MSCs), which play a key role in protecting, regenerating, and proliferating hematopoietic stem cells (HSCs). This feature is vital for HSC after exposure to myelotoxic anticancer agents; nevertheless, the effects of nutlin-3 on MSCs remain to be disclosed. The present research study was conducted to examine the antiproliferative and proapoptotic effectiveness of nutlin-3 in bone marrow MSCs (BMSCs). MATERIALS AND METHODS Human-derived BMSCs were cultured for different durations, that is, 24, 48, and 72 hours, and treated using various concentrations of nutlin-3, including 5, 10, 25, 50, and 100 μΜ. To investigate the effect of nutlin-3 on the apoptosis, cell vitality and proliferation in BMSCs, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), thiazolyl blue tetrazolium bromide, propidium iodide (PI) and annexin V assay, as well as real-time polymerase chain reaction, were used. RESULTS BMSCs viability significantly decreased (P < .05) in the cells treated at concentrations of 50 and 100 μM for 24 hours and concentrations of 25, 50, and 100 μM for 48 hours and at all concentrations for 72 hours. The apoptosis of BMSCs (TUNEL positive) was significantly more visible at concentrations of 25 and 50 μM compared with that in the controls (P < .05), while this increased through dose-dependent processes. Annexin V/PI staining revealed negligible dose-dependent increases in all the apoptotic cells after 72 hours of incubation, and this apoptosis elevation was significant at 25 and 50 μM (P < .05). CONCLUSION Resistance to nutlin-3 was observed in human bone marrow-derived MSCs; nevertheless, further clinical data are required to be obtained with long-duration exposure to confirm the present findings.
Collapse
Affiliation(s)
- Babak Bajelan
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Majid Zaki-Dizaji
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Rahmani
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sina Darzi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
20
|
Bajelan B, Zaki‐Dizaji M, Darabi S, Rajaei F. The effects of Nutlin‐3 on morphology, cellular proliferation, and apoptosis in rat primary mesenchymal stem cells. J Cell Physiol 2018; 234:11424-11430. [DOI: 10.1002/jcp.27798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Babak Bajelan
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences Qazvin Iran
| | - Majid Zaki‐Dizaji
- Department of Medical Genetics School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences Qazvin Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences Qazvin Iran
- Department of Histology and Embryology, Faculty of Medicine Qazvin University of Medical Sciences Qazvin Iran
| |
Collapse
|
21
|
|
22
|
Rühle A, Huber PE, Saffrich R, Lopez Perez R, Nicolay NH. The current understanding of mesenchymal stem cells as potential attenuators of chemotherapy-induced toxicity. Int J Cancer 2018; 143:2628-2639. [PMID: 29931767 DOI: 10.1002/ijc.31619] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022]
Abstract
Chemotherapeutic agents are part of the standard treatment algorithms for many malignancies; however, their application and dosage are limited by their toxic effects to normal tissues. Chemotherapy-induced toxicities can be long-lasting and may be incompletely reversible; therefore, causative therapies for chemotherapy-dependent side effects are needed, especially considering the increasing survival rates of treated cancer patients. Mesenchymal stem cells (MSCs) have been shown to exhibit regenerative abilities for various forms of tissue damage. Preclinical data suggest that MSCs may also help to alleviate tissue lesions caused by chemotherapeutic agents, mainly by establishing a protective microenvironment for functional cells. Due to the systemic administration of most anticancer agents, the effects of these drugs on the MSCs themselves are of crucial importance to use stem cell-based approaches for the treatment of chemotherapy-induced tissue toxicities. Here, we present a concise review of the published data regarding the influence of various classes of chemotherapeutic agents on the survival, stem cell characteristics and physiological functions of MSCs. Molecular mechanisms underlying the effects are outlined, and resulting challenges of MSC-based treatments for chemotherapy-induced tissue injuries are discussed.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Rainer Saffrich
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany.,Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Chulpanova DS, Kitaeva KV, Tazetdinova LG, James V, Rizvanov AA, Solovyeva VV. Application of Mesenchymal Stem Cells for Therapeutic Agent Delivery in Anti-tumor Treatment. Front Pharmacol 2018; 9:259. [PMID: 29615915 PMCID: PMC5869248 DOI: 10.3389/fphar.2018.00259] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/08/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are non-hematopoietic progenitor cells, which can be isolated from different types of tissues including bone marrow, adipose tissue, tooth pulp, and placenta/umbilical cord blood. There isolation from adult tissues circumvents the ethical concerns of working with embryonic or fetal stem cells, whilst still providing cells capable of differentiating into various cell lineages, such as adipocytes, osteocytes and chondrocytes. An important feature of MSCs is the low immunogenicity due to the lack of co-stimulatory molecules expression, meaning there is no need for immunosuppression during allogenic transplantation. The tropism of MSCs to damaged tissues and tumor sites makes them a promising vector for therapeutic agent delivery to tumors and metastatic niches. MSCs can be genetically modified by virus vectors to encode tumor suppressor genes, immunomodulating cytokines and their combinations, other therapeutic approaches include MSCs priming/loading with chemotherapeutic drugs or nanoparticles. MSCs derived membrane microvesicles (MVs), which play an important role in intercellular communication, are also considered as a new therapeutic agent and drug delivery vector. Recruited by the tumor, MSCs can exhibit both pro- and anti-oncogenic properties. In this regard, for the development of new methods for cancer therapy using MSCs, a deeper understanding of the molecular and cellular interactions between MSCs and the tumor microenvironment is necessary. In this review, we discuss MSC and tumor interaction mechanisms and review the new therapeutic strategies using MSCs and MSCs derived MVs for cancer treatment.
Collapse
Affiliation(s)
- Daria S Chulpanova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Leysan G Tazetdinova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Albert A Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Valeriya V Solovyeva
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
24
|
Somaiah C, Kumar A, Sharma R, Sharma A, Anand T, Bhattacharyya J, Das D, Deka Talukdar S, Jaganathan BG. Mesenchymal stem cells show functional defect and decreased anti-cancer effect after exposure to chemotherapeutic drugs. J Biomed Sci 2018; 25:5. [PMID: 29351753 PMCID: PMC5774172 DOI: 10.1186/s12929-018-0407-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 01/08/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) are used for several therapeutic applications to improve the functions of bone, cardiac, nervous tissue as well as to facilitate the repopulation of hematopoietic stem cells. MSC give rise to the non-hematopoietic stromal cells of the bone marrow and are important for the maintenance of normal hematopoiesis. Chemotherapeutic drugs used for treatment of leukemia extensively damage the stromal cells and alter their gene expression profiles. METHODS We determined the changes in adipogenic, osteogenic differentiation, phenotypic and gene expression in MSC during treatment with chemotherapeutic drugs cytarabine, daunorubicin and vincristine. We also tested anti-cancer effects of drug treated MSC on leukemia cells. RESULTS Treatment with the chemotherapeutic drugs resulted in functional defects in MSC, leading to reduced proliferation, osteogenic and adipogenic differentiation. The drug treated MSC also showed decreased expression of cell surface receptors, and the changes in proliferation, phenotype and differentiation defect was partially reversible after withdrawing the drugs from the cells. The drug treated MSC showed increased expression of cytokines, IL6, FGF2 and TNFA but reduced levels of differentiation markers SOX9 and ACTC1. Drug treated MSC also contributed to reduced anti-cancer effects in leukemia cells. CONCLUSIONS Chemotherapeutic drug treatment altered the phenotype, osteogenic and adipogenic differentiation potential of MSC and modified the gene expression profile of the cells to render them more chemoprotective of the leukemic cells. Thus, additional therapeutic efforts to target the stromal cell population will help in preventing chemoresistance, disease relapse in leukemia and to maintain a healthy bone marrow stroma.
Collapse
Affiliation(s)
- Chinnapaka Somaiah
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Atul Kumar
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Renu Sharma
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Amit Sharma
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Trishna Anand
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Jina Bhattacharyya
- Department of Hematology, Gauhati Medical College and Hospital, Guwahati, India
| | - Damodar Das
- Department of Hematology, Gauhati Medical College and Hospital, Guwahati, India
| | | | - Bithiah Grace Jaganathan
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
25
|
Münz F, Lopez Perez R, Trinh T, Sisombath S, Weber KJ, Wuchter P, Debus J, Saffrich R, Huber PE, Nicolay NH. Human mesenchymal stem cells lose their functional properties after paclitaxel treatment. Sci Rep 2018; 8:312. [PMID: 29321693 PMCID: PMC5762916 DOI: 10.1038/s41598-017-18862-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are an integral part of the bone marrow niche and aid in the protection, regeneration and proliferation of hematopoietic stem cells after exposure to myelotoxic taxane anti-cancer agents, but the influence of taxane compounds on MSCs themselves remains incompletely understood. Here, we show that bone marrow-derived MSCs are highly sensitive even to low concentrations of the prototypical taxane compound paclitaxel. While MSCs remained metabolically viable, they were strongly impaired regarding both their proliferation and their functional capabilities after exposure to paclitaxel. Paclitaxel treatment resulted in reduced cell migration, delays in cellular adhesion and significant dose-dependent inhibition of the stem cells’ characteristic multi-lineage differentiation potential. Cellular morphology and expression of the defining surface markers remained largely unaltered. Paclitaxel only marginally increased apoptosis in MSCs, but strongly induced premature senescence in these stem cells, thereby explaining the preservation of the metabolic activity of functionally inactivated MSCs. The reported sensitivity of MSC function to paclitaxel treatment may help to explain the severe bone marrow toxicities commonly caused by taxane-based anti-cancer treatments.
Collapse
Affiliation(s)
- Franziska Münz
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ramon Lopez Perez
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Thuy Trinh
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Klaus-Josef Weber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Friedrich-Ebert-Str. 107, 68167, Mannheim, Germany
| | - Jürgen Debus
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg - Hessen, Medical Faculty Mannheim, Friedrich-Ebert-Str. 107, 68167, Mannheim, Germany.,Department of Hematology and Oncology, Heidelberg University Hospital, Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter E Huber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Nils H Nicolay
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120, Heidelberg, Germany. .,Department of Radiation Oncology, Heidelberg University Hospital, Neuenheimer Feld 400, 69120, Heidelberg, Germany.
| |
Collapse
|
26
|
Rühle A, Xia O, Perez RL, Trinh T, Richter W, Sarnowska A, Wuchter P, Debus J, Saffrich R, Huber PE, Nicolay NH. The Radiation Resistance of Human Multipotent Mesenchymal Stromal Cells Is Independent of Their Tissue of Origin. Int J Radiat Oncol Biol Phys 2018; 100:1259-1269. [PMID: 29452769 DOI: 10.1016/j.ijrobp.2018.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/10/2017] [Accepted: 01/03/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE Human mesenchymal stromal cells (MSCs) may aid the regeneration of ionizing radiation (IR)-induced tissue damage. They can be harvested from different tissues for clinical purposes; however, the role of the tissue source on the radiation response of human MSCs remains unknown. METHODS AND MATERIALS Human MSCs were isolated from adipose tissue, bone marrow, and umbilical cord, and cellular survival, proliferation, and apoptosis were measured after irradiation. The influence of IR on the defining functions of MSCs was assessed, and cell morphology, surface marker expression, and the differentiation potential were examined. Western blot analyses were performed to assess the activation of DNA damage signaling and repair pathways. RESULTS MSCs from adipose tissue, bone marrow, and umbilical cord exhibited a relative radioresistance independent of their tissue of origin. Defining properties including cellular adhesion and surface marker expression were preserved, and irradiated MSCs maintained their potential for multilineage differentiation irrespective of their tissue source. Analysis of activated DNA damage recognition and repair pathways demonstrated an efficient repair of IR-induced DNA double-strand breaks in MSCs from different tissues, thereby influencing the induction of apoptosis. CONCLUSIONS These data show for the first time that MSCs are resistant to IR and largely preserve their defining functions after irradiation irrespective of their tissue of origin. Efficient repair of IR-induced DNA double-strand breaks and consecutive reduction of apoptosis induction may contribute to the tissue-independent radiation resistance of MSCs.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Xia
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna Sarnowska
- Translative Platform for Regenerative Medicine, Mossakowski Medical Research Center, Polish Academy of Sciences, Warsaw, Poland
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, German Red Cross Donor Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Saffrich
- Institute of Transfusion Medicine and Immunology, German Red Cross Donor Blood Service Baden-Württemberg-Hessen, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; Department of Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
27
|
Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin Exp Metastasis 2018; 35:269-284. [PMID: 29307118 DOI: 10.1007/s10585-017-9870-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Tumors often overcome the cytotoxic effects of chemotherapy through either acquired or environment-mediated drug resistance. In addition, signals from the microenvironment obfuscate the beneficial effects of chemotherapy and may facilitate progression and metastatic dissemination. Seminal mediators in chemotherapy-induced metastasis appear to be a wide range of hematopoietic, mesenchymal and immune progenitor cells, originating from the bone marrow. The actual purpose of these cells is to orchestrate the repair response to the cytotoxic damage of chemotherapy. However, these repair responses are exploited by tumor cells at every step of the metastatic cascade, ranging from tumor cell invasion, intravasation and hematogenous dissemination to extravasation and effective colonization at the metastatic site. A better understanding of the mechanistic underpinnings of chemotherapy-induced metastasis will allow us to better predict which patients are more likely to exhibit pro-metastatic responses to chemotherapy and will help develop new therapeutic strategies to neutralize chemotherapy-driven prometastatic changes.
Collapse
|
28
|
Real NE, Castro GN, Darío Cuello-Carrión F, Perinetti C, Röhrich H, Cayado-Gutiérrez N, Guerrero-Gimenez ME, Ciocca DR. Molecular markers of DNA damage and repair in cervical cancer patients treated with cisplatin neoadjuvant chemotherapy: an exploratory study. Cell Stress Chaperones 2017; 22:811-822. [PMID: 28608263 PMCID: PMC5655369 DOI: 10.1007/s12192-017-0811-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/10/2017] [Accepted: 05/12/2017] [Indexed: 12/27/2022] Open
Abstract
Neoadjuvant (or induction) chemotherapy can be used for cervical cancer patients with locally advanced disease; this treatment is followed by radical surgery and/or radiation therapy. Cisplatin is considered to be the most active platinum agent drug for this cancer, with a response rate of 20%. In order to understand how the cisplatin treatment affects the stress response, in this work, we performed an exploratory study to analyze a number of stress proteins before and after cisplatin neoadjuvant chemotherapy. The study involved 14 patients; the pre- and post-chemotherapy paired biopsies were examined by hematoxylin and eosin staining and by immunohistochemistry. The proteins evaluated were p53, P16/INK4A, MSH2, nuclear protein transcriptional regulator 1 (NUPR1), and HSPB1 (total: HSPB1/t and phosphorylated: HSPB1/p). These proteins were selected because there is previous evidence of their relationship with drug resistance. The formation of platinum-DNA adducts was also studied. There was a great variation in the expression levels of the mentioned proteins in the pre-chemotherapy biopsies. After chemotherapy, p53 was not significantly affected by cisplatin, as well as P16/INK4A and MSH2 while nuclear NUPR1 content tended to decrease (p = 0.056). Cytoplasmic HSPB1/t expression levels decreased significantly following cisplatin therapy while nuclear HSPB1/t and HSPB1/p tended to increase. Since the most significant changes following chemotherapy appeared in the HSPB1 expression levels, the changes were confirmed by Western blot. The platinum-DNA adducts were observed in HeLa cell in apoptosis; however, in the tumor samples, the platinum-DNA adducts were observed in morphologically healthy tumor cells; these cells displayed nuclear HSPB1/p. Further mechanistic studies should be performed to reveal how HSPB1/p is related with drug resistance. When the correlations of the markers with the response to neoadjuvant chemotherapy were examined, only high pre-chemotherapy levels of cytoplasmic HSPB1/p correlated with a poor clinical and pathological response to neoadjuvant cisplatin chemotherapy (p = 0.056) suggesting that this marker could be useful opening its study in a larger number of cases.
Collapse
Affiliation(s)
- Nilda E Real
- Oncology Department, Hospital Diego Paroissien of Maipú, Mendoza, Argentina
| | - Gisela N Castro
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500, Mendoza, Argentina
| | - F Darío Cuello-Carrión
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500, Mendoza, Argentina
| | - Claudia Perinetti
- Oncology Department, Hospital Diego Paroissien of Maipú, Mendoza, Argentina
| | | | - Niubys Cayado-Gutiérrez
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500, Mendoza, Argentina
| | - Martin E Guerrero-Gimenez
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500, Mendoza, Argentina
| | - Daniel R Ciocca
- Laboratory of Oncology, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Dr. Ruiz Leal s/n, Parque General San Martín, 5500, Mendoza, Argentina.
| |
Collapse
|
29
|
Rühle A, Perez RL, Glowa C, Weber KJ, Ho AD, Debus J, Saffrich R, Huber PE, Nicolay NH. Cisplatin radiosensitizes radioresistant human mesenchymal stem cells. Oncotarget 2017; 8:87809-87820. [PMID: 29152122 PMCID: PMC5675674 DOI: 10.18632/oncotarget.21214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022] Open
Abstract
Cisplatin-based chemo-radiotherapy is widely used to treat cancers with often severe therapy-associated late toxicities. While mesenchymal stem cells (MSCs) were shown to aid regeneration of cisplatin- or radiation-induced tissue lesions, the effect of the combined treatment on the stem cells remains unknown. Here we demonstrate that cisplatin treatment radiosensitized human bone marrow-derived MSCs in a dose-dependent manner and increased levels of radiation-induced apoptosis. However, the defining stem cell properties of MSCs remained largely intact after cisplatin-based chemo-radiation, and stem cell motility, adhesion, surface marker expression and the characteristic differentiation potential were not significantly influenced. The increased cisplatin-mediated radiosensitivity was associated with a cell cycle shift of MSCs towards the radiosensitive G2/M phase and increased residual DNA double-strand breaks. These data demonstrate for the first time a dose-dependent radiosensitization effect of MSCs by cisplatin. Clinically, the observed increase in radiation sensitivity and subsequent loss of regenerative MSCs may contribute to the often severe late toxicities observed after cisplatin-based chemo-radiotherapy in cancer patients.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany
| | - Christin Glowa
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany
| | - Klaus-Josef Weber
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Department of Hematology and Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Peter E Huber
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.,Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, 69120 Heidelberg, Germany.,Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Chang YH, Liu HW, Chu TY, Wen YT, Tsai RK, Ding DC. Cisplatin-Impaired Adipogenic Differentiation of Adipose Mesenchymal Stem Cells 1. Cell Transplant 2017; 26:1077-1087. [PMID: 28155807 DOI: 10.3727/096368917x694886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (ADSCs) are derived from adipose tissue and can be induced in vitro to differentiate into osteoblasts, chondroblasts, myocytes, neurons, and other cell types. Cisplatin is a commonly used chemotherapy drug for cancer patients. However, the effects of cisplatin on ADSCs remain elusive. This study found that a high concentration of cisplatin affects the viability of ADSCs. First, the IC50 concentration of cisplatin was evaluated. Proliferation of ADSCs, as assessed by the XTT method, decreased immediately after treatment with various concentrations of cisplatin. ADSCs maintained mesenchymal stem cell surface markers after cisplatin treatment, as determined by flow cytometry. Upon differentiation by adding specific reagents, a significant decrease in adipogenic differentiation (by Oil red O staining) and osteogenic differentiation (by Alizarin red staining), and significant chondrogenic differentiation (by Alcian blue staining) were found after cisplatin treatment. Quantitative RT-PCR was also used in evaluating expression of specific genes to confirm differentiation. Finally, ADSCs from one donor who had received cisplatin showed significantly decreased adipogenic differentiation but increased osteogenic differentiation compared with ADSCs derived from one healthy donor. In conclusion, cisplatin affects the viability, proliferation, and differentiation of ADSCs both in vitro and in vivo via certain signaling pathways, such as p53 and Fas/FasL. The differentiation abilities of ADSCs should be evaluated before their transplantation for repairing cisplatin-induced tissue damage.
Collapse
|
31
|
Maria OM, Shalaby M, Syme A, Eliopoulos N, Muanza T. Adipose mesenchymal stromal cells minimize and repair radiation-induced oral mucositis. Cytotherapy 2016; 18:1129-45. [PMID: 27424150 DOI: 10.1016/j.jcyt.2016.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/18/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) have been used to minimize and repair radiation-induced normal tissue injury in the intestine, salivary gland, liver, skin, lungs and cardiac muscle. This study investigated the ability of adipose tissue-derived MSCs (aMSCs) to minimize and/or repair single dose radiation-induced oral mucositis (RIOM). METHODS Syngenic phenotypically and functionally characterized BALB/c mouse aMSCs were implanted intraperitoneally in a RIOM mouse model with different dosing protocols. Response was quantified macroscopically, microscopically and by using different histological and clinically relevant parameters. RESULTS Irradiation at 18 Gy generated a self-resolved single-dose RIOM BALB/c mouse model with 5.6 ± 0.3 days mean duration (95% confidence interval (CI) 4.233-7.1 days) and 100% survival rate. Intraperitoneal implantation of 5 doses of 2.5 million freshly cultured syngenic aMSCs significantly and reproducibly reduced RIOM ulcer duration to 1.6 ± 0.3 days (95% CI 0.0233-3.1 days, a 72% reduction in RIOM ulcer duration), ulcer size and ulcer floor epithelial height. The therapeutic benefits were significantly dependent on dose size and frequency, number of doses, and therapy onset time. aMSCs therapy significantly minimized the RIOM-related weight loss, accelerated the weight gain and improved irradiated animals' hydration and nutritional status. aMSCs therapy did not potentiate head and neck cancer in vitro. CONCLUSIONS Syngenic freshly cultured aMSCs significantly minimized and repaired radiation-induced oral mucositis with a 72% reduction in ulcer duration. aMSCs dose size and frequency, number of doses and therapy onset time are the main keys for optimized therapeutic outcome. aMSCs therapy did not stimulate Head and Neck cancer cell growth in-vitro.
Collapse
Affiliation(s)
- Osama Muhammad Maria
- Experimental Medicine Department, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Surgery Department, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Radiation Oncology Department, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | - Alasdair Syme
- Radiation Oncology Department, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Medical Physics Unit, Montreal, Quebec, Canada; Oncology Department, McGill University, Montreal, Quebec, Canada
| | - Nicoletta Eliopoulos
- Surgery Department, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Thierry Muanza
- Experimental Medicine Department, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Radiation Oncology Department, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Oncology Department, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
32
|
Mesenchymal stem cells are sensitive to bleomycin treatment. Sci Rep 2016; 6:26645. [PMID: 27215195 PMCID: PMC4877675 DOI: 10.1038/srep26645] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/06/2016] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to attenuate pulmonary damage induced by bleomycin-based anticancer treatments, but the influence of bleomycin on the stem cells themselves remains largely unknown. Here, we demonstrate that human bone marrow-derived MSCs are relatively sensitive to bleomycin exposure compared to adult fibroblasts. MSCs revealed increased levels of apoptosis after bleomycin treatment, while cellular morphology, stem cell surface marker expression and the ability for adhesion and migration remained unchanged. Bleomycin treatment also resulted in a reduced adipogenic differentiation potential of these stem cells. MSCs were found to efficiently repair DNA double strand breaks induced by bleomycin, mostly through non-homologous end joining repair. Low mRNA and protein expression levels of the inactivating enzyme bleomycin hydrolase were detected in MSCs that may contribute to the observed bleomycin-sensitive phenotype of these cells. The sensitivity of MSCs against bleomycin needs to be taken into consideration for ongoing and future treatment protocols investigating these stem cells as a potential treatment option for bleomycin-induced pulmonary damage in the clinic.
Collapse
|
33
|
Nicolay NH, Rühle A, Perez RL, Trinh T, Sisombath S, Weber KJ, Schmezer P, Ho AD, Debus J, Saffrich R, Huber PE. Mesenchymal stem cells exhibit resistance to topoisomerase inhibition. Cancer Lett 2016; 374:75-84. [PMID: 26876302 DOI: 10.1016/j.canlet.2016.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Inhibition of cellular topoisomerases has been established as an effective way of treating certain cancers, albeit with often high levels of toxicity to the bone marrow. While the involvement of mesenchymal stem cells (MSCs) in bone marrow homeostasis and regeneration has been well established, the effects of topoisomerase-inhibiting anticancer agents remain largely unknown. MATERIALS AND METHODS Human bone marrow MSCs were treated with topoisomerase I inhibitor irinotecan or topoisomerase II inhibitor etoposide, and survival and apoptosis levels were measured. The influence of topoisomerase inhibition on cellular morphology, adhesion and migration potential and the ability to differentiate was assessed. Additionally, the role of individual DNA double-strand break repair pathways in MSCs was investigated as a potential cellular mechanism of resistance to topoisomerase inhibitors. RESULTS Human bone marrow MSCs were found relatively resistant to topoisomerase I and II inhibitors and show survival levels comparable to these of differentiated fibroblasts. Treatment with irinotecan or etoposide did not significantly influence cellular adhesion, migratory ability, surface marker expression or induction of apoptosis in human MSCs. The ability to differentiate was found preserved in MSCs after exposure to high doses of irinotecan or etoposide. MSCs were able to efficiently repair DNA double-strand breaks induced by topoisomerase inhibitors both by non-homologous end joining and homologous recombination pathways. CONCLUSION Our data demonstrate a topoisomerase-resistant phenotype of human MSCs that may at least in part be due to the stem cells' ability to efficiently remove DNA damage caused by these anticancer agents. The observed resistance of MSCs warrants further investigation of these cells as a potential therapeutic option for treating topoisomerase inhibitor-induced bone marrow damage.
Collapse
Affiliation(s)
- Nils H Nicolay
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Alexander Rühle
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Thuy Trinh
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Sonevisay Sisombath
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Peter Schmezer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anthony D Ho
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Rainer Saffrich
- Department of Hematology and Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|