1
|
Colombini A, Lopa S, Libonati F, Talò G, Mareschi K, Marini E, Mangiavini L, Raffo V, Moretti M, de Girolamo L. Low-density cultured cartilage cells expanded in platelet lysate present distinct features to develop an innovative clinical treatment for diffuse cartilage lesions. Knee Surg Sports Traumatol Arthrosc 2024; 32:2859-2873. [PMID: 38842036 DOI: 10.1002/ksa.12305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE Chondrocyte-based cell therapies are effective for the treatment of chondral lesions, but remain poorly indicated for diffuse lesions in the context of early osteoarthritis (OA). The aim of this study was to develop a protocol to obtain chondroprogenitor cells suitable for the treatment of diffuse chondral lesions within early OA. METHODS Cartilage cells were expanded at low density in human platelet lysate (hPL). A test was performed to exclude senescence. The expression of surface cluster of differentiation 146, cluster of differentiation 166, major histocompatibility complex (MHC)-I and MHC-II and of genes of interest were evaluated, as well as the trophic potential of these cells, by the assessment of lubricin and matrix production. The immunomodulatory potential was assessed through their co-culture with macrophages. RESULTS Cartilage cells expanded at low density in hPL showed higher proliferation rate than standard-density cells, no replicative senescence, low immunogenicity and expression of lubricin. Moreover, they presented an increased expression of chondrogenic and antihypertrophic markers, as well as a superior matrix deposition if compared to cells cultured at standard density. Cartilage cells induced on macrophages an upregulation of CD206, although a higher increase of CD163 expression was observed in the presence of low-density cells. CONCLUSIONS These findings lay the grounds to explore the clinical usefulness of low-density cultured cartilage cells to treat diffuse lesions in early OA joints for both autologous and allogenic use. LEVEL OF EVIDENCE Not applicable.
Collapse
Affiliation(s)
| | - Silvia Lopa
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Francesca Libonati
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Giuseppe Talò
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Turin, Italy
| | - Elena Marini
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Raffo
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Laura de Girolamo
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
2
|
Khandani B, Movahedin M. Learning Towards Maturation of Defined Feeder-free Pluripotency Culture Systems: Lessons from Conventional Feeder-based Systems. Stem Cell Rev Rep 2024; 20:484-494. [PMID: 38079087 DOI: 10.1007/s12015-023-10662-7] [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] [Accepted: 12/01/2023] [Indexed: 02/03/2024]
Abstract
Pluripotent stem cells (PSCs) are widely recognized as one of the most promising types of stem cells for applications in regenerative medicine, tissue engineering, disease modeling, and drug screening. This is due to their unique ability to differentiate into cells from all three germ layers and their capacity for indefinite self-renewal. Initially, PSCs were cultured using animal feeder cells, but these systems presented several limitations, particularly in terms of Good Manufacturing Practices (GMP) regulations. As a result, feeder-free systems were introduced as a safer alternative. However, the precise mechanisms by which feeder cells support pluripotency are not fully understood. More importantly, it has been observed that some aspects of the need for feeder cells like the optimal density and cell type can vary depending on conditions such as the developmental stage of the PSCs, phases of the culture protocol, the method used in culture for induction of pluripotency, and intrinsic variability of PSCs. Thus, gaining a better understanding of the divergent roles and necessity of feeder cells in various conditions would lead to the development of condition-specific defined feeder-free systems that resolve the failure of current feeder-free systems in some conditions. Therefore, this review aims to explore considerable feeder-related issues that can lead to the development of condition-specific feeder-free systems.
Collapse
Affiliation(s)
- Bardia Khandani
- Department of Stem Cells Technology and Tissue Regeneration, Faculty of Interdisciplinary Science and Technology, Tarbiat Modares University, Tehran, Iran
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran, 14115111, Iran.
| |
Collapse
|
3
|
Tan Kwan Zen N, Zeming KK, Teo KL, Loberas M, Lee J, Goh CR, Yang DH, Oh S, Hui Hoi Po J, Cool SM, Hou HW, Han J. Scalable mesenchymal stem cell enrichment from bone marrow aspirate using deterministic lateral displacement (DLD) microfluidic sorting. LAB ON A CHIP 2023; 23:4313-4323. [PMID: 37702123 DOI: 10.1039/d3lc00379e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
The growing interest in regenerative medicine has opened new avenues for novel cell therapies using stem cells. Bone marrow aspirate (BMA) is an important source of stromal mesenchymal stem cells (MSCs). Conventional MSC harvesting from BMA relies on archaic centrifugation methods, often leading to poor yield due to osmotic stress, high centrifugation force, convoluted workflow, and long experimental time (∼2-3 hours). To address these issues, we have developed a scalable microfluidic technology based on deterministic lateral displacement (DLD) for MSC isolation. This passive, label-free cell sorting method capitalizes on the morphological differences between MSCs and blood cells (platelets and RBCs) for effective separation using an inverted L-shaped pillar array. To improve throughput, we developed a novel multi-chip DLD system that can process 2.5 mL of raw BMA in 20 ± 5 minutes, achieving a 2-fold increase in MSC recovery compared to centrifugation methods. Taken together, we envision that the developed DLD platform will enable fast and efficient isolation of MSCs from BMA for effective downstream cell therapy in clinical settings.
Collapse
Affiliation(s)
- Nicholas Tan Kwan Zen
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology (SMART), 138602, Singapore
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
| | - Kerwin Kwek Zeming
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology (SMART), 138602, Singapore
| | - Kim Leng Teo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 138668, Singapore
| | - Mavis Loberas
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 117510, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 119288, Singapore
| | - Jialing Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 138668, Singapore
| | - Chin Ren Goh
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology (SMART), 138602, Singapore
| | - Da Hou Yang
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology (SMART), 138602, Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 138668, Singapore
| | - James Hui Hoi Po
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 117510, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 119288, Singapore
| | - Simon M Cool
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 119288, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138668, Singapore
- School of Chemical Engineering, University of Queensland, Brisbane, 4072, Australia
| | - Han Wei Hou
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology (SMART), 138602, Singapore
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology (SMART), 138602, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA.
| |
Collapse
|
4
|
Orso F, Virga F, Dettori D, Dalmasso A, Paradzik M, Savino A, Pomatto MAC, Quirico L, Cucinelli S, Coco M, Mareschi K, Fagioli F, Salmena L, Camussi G, Provero P, Poli V, Mazzone M, Pandolfi PP, Taverna D. Stroma-derived miR-214 coordinates tumor dissemination. J Exp Clin Cancer Res 2023; 42:20. [PMID: 36639824 PMCID: PMC9837925 DOI: 10.1186/s13046-022-02553-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/29/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Tumor progression is based on a close interaction between cancer cells and Tumor MicroEnvironment (TME). Here, we focus on the role that Cancer Associated Fibroblasts (CAFs), Mesenchymal Stem Cells (MSCs) and microRNAs (miRs) play in breast cancer and melanoma malignancy. METHODS We used public databases to investigate miR-214 expression in the stroma compartment of primary human samples and evaluated tumor formation and dissemination following tumor cell injections in miR-214 overexpressing (miR-214over) and knock out (miR-214ko) mice. In addition, we dissected the impact of Conditioned Medium (CM) or Extracellular Vesicles (EVs) derived from miR-214-rich or depleted stroma cells on cell metastatic traits. RESULTS We evidence that the expression of miR-214 in human cancer or metastasis samples mostly correlates with stroma components and, in particular, with CAFs and MSCs. We present data revealing that the injection of tumor cells in miR-214over mice leads to increased extravasation and metastasis formation. In line, treatment of cancer cells with CM or EVs derived from miR-214-enriched stroma cells potentiate cancer cell migration/invasion in vitro. Conversely, dissemination from tumors grown in miR-214ko mice is impaired and metastatic traits significantly decreased when CM or EVs from miR-214-depleted stroma cells are used to treat cells in culture. Instead, extravasation and metastasis formation are fully re-established when miR-214ko mice are pretreated with miR-214-rich EVs of stroma origin. Mechanistically, we also show that tumor cells are able to induce miR-214 production in stroma cells, following the activation of IL-6/STAT3 signaling, which is then released via EVs subsequently up-taken by cancer cells. Here, a miR-214-dependent pro-metastatic program becomes activated. CONCLUSIONS Our findings highlight the relevance of stroma-derived miR-214 and its release in EVs for tumor dissemination, which paves the way for miR-214-based therapeutic interventions targeting not only tumor cells but also the TME.
Collapse
Affiliation(s)
- Francesca Orso
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
- Dept. of Translational Medicine (DIMET), Università del Piemonte Orientale, Novara, Italy
| | - Federico Virga
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
- Lab of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Louvain, Belgium
- Present Address: Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Daniela Dettori
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Alberto Dalmasso
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Mladen Paradzik
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Aurora Savino
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | | | - Lorena Quirico
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Stefania Cucinelli
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Martina Coco
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Katia Mareschi
- Paediatric Onco-Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Turin, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Franca Fagioli
- Paediatric Onco-Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Turin, Italy
- Department of Public Health and Paediatrics, University of Turin, Turin, Italy
| | - Leonardo Salmena
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paolo Provero
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Valeria Poli
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy
| | - Massimiliano Mazzone
- Lab of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Louvain, Belgium
| | - Pier Paolo Pandolfi
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy.
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy.
- William N. Pennington Cancer Institute, Renown Health, Nevada System of Higher Education, Reno, NV, 89502, USA.
| | - Daniela Taverna
- Molecular Biotechnology Center (MBC) "Guido Tarone", Via Nizza, 52, 10126, Turin, Italy.
- Dept. Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza, 52, 10126, Turin, Italy.
| |
Collapse
|
5
|
Rizvi SFA, Wasim B, Usman S, Borges KJJ, Sahibdad I, Salim A, Khan I. Zinc and hypoxic preconditioning: a strategy to enhance the functionality and therapeutic potential of bone marrow-derived mesenchymal stem cells. Mol Cell Biochem 2022; 477:2735-2749. [PMID: 35610401 DOI: 10.1007/s11010-022-04468-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 05/04/2022] [Indexed: 11/09/2022]
Abstract
The therapeutic use of bone marrow mesenchymal stem cells (BM-MSCs) requires a large number of cells (1-100 × 106 cells/kg of body weight). Extensive in vitro growth is limited due to the aging of cultured BM-MSCs which leads to abnormal morphology and senescence. Hypoxia increases BM-MSC proliferation, but the question of whether hypoxia preconditioning is safe for clinical application of BM-MSCs remains to be answered. Zinc is essential for cell proliferation and differentiation, especially for the regulation of DNA synthesis and mitosis. It is a structural constituent of numerous proteins on a molecular level, including transcription factors and enzymes of cellular signaling machinery. All the tissues, fluids, and organs of the human body contain zinc. More than 95% of zinc is intracellular, of which 44% is involved in the transcription of DNA. We investigated the effects of ZnCl2 on proliferation, morphology, migration, population doubling time (PDT), and gene expression of BM-MSCs under hypoxic (1% O2) and normoxic (21% O2) environments. BM-MSCs were preconditioned with optimized concentrations of ZnCl2 under normoxic and hypoxic environments and further examined for morphology by the phase-contrast inverted microscope, cell proliferation by MTT assay, PDT, cell migration ability, and gene expression analysis. Zinc significantly enhanced the proliferation of BM-MSCs, and it decreases PDT under hypoxic and normoxic environments as compared to control cells. Migration of BM-MSCs toward the site of injury increased and expression of HIF1-α significantly decreased under hypoxic conditions as compared to non-treated hypoxic cells and control. At late passages (P9), the morphology of normoxic BM-MSCs was transformed into large, wide, and flat cells, and they became polygonal and lost their communication with other cells. Conversely, zinc-preconditioned BM-MSCs retained their spindle-shaped, fibroblast-like morphology at P9. The expression of proliferative genes was found significantly upregulated, while downregulation of genes OCT4 and CCNA2 was observed in zinc-treated BM-MSCs under both normoxic and hypoxic conditions. ZnCl2 treatment can be used for extensive expansion of BM-MSCs in aged populations to obtain a large number of cells required for systemic administration to produce therapeutic efficacy.
Collapse
Affiliation(s)
- Syed Faizan Ali Rizvi
- Ghulam Muhammad Mahar Medical College Sukkur at Shaheed Mohtarma Benazir Bhutto Medical University Larkana, Larkana, 77150, Pakistan
- Ziauddin University, Clifton, Karachi, 74700, Pakistan
| | - Bushra Wasim
- Ziauddin University, Clifton, Karachi, 74700, Pakistan
| | | | | | - Iqra Sahibdad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Centre for Chemical and Biological Sciences, Karachi, 75270, Pakistan
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Centre for Chemical and Biological Sciences, Karachi, 75270, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Centre for Chemical and Biological Sciences, Karachi, 75270, Pakistan.
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
6
|
Centeno CJ, Berger DR, Money BT, Dodson E, Urbanek CW, Steinmetz NJ. Percutaneous autologous bone marrow concentrate for knee osteoarthritis: patient-reported outcomes and progenitor cell content. INTERNATIONAL ORTHOPAEDICS 2022; 46:2219-2228. [PMID: 35932306 PMCID: PMC9492580 DOI: 10.1007/s00264-022-05524-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/18/2022] [Indexed: 12/03/2022]
Abstract
PURPOSE Knee osteoarthritis (OA) is a common, progressively debilitating joint disease, and the intra-articular injection of autologous bone marrow concentrate (BMC) may offer a minimally invasive method of harnessing the body's own connective tissue progenitor cells to counteract accompanying degenerative effects of the disease. However, the extent to which the progenitor cell content of BMC influences treatment outcomes is unclear. We sought to determine whether patient-reported outcome measures associated with BMC treatment for knee OA are related to the concentration of progenitor cells provided. METHODS In the present study, 65 patients (72 knees) underwent treatment for knee OA with autologous BMC and self-reported their outcomes for up to one year using follow-up questionnaires tracking function, pain, and percent improvement. A small fraction of each patient's BMC sample was reserved for quantification with a haematological analyzer and cryopreserved for subsequent analysis of potential connective tissue progenitor cells using a colony-forming unit fibroblast (CFU-F) assay. RESULTS Patients reported significant increases in function and overall percent improvement in addition to decreases in pain relative to baseline levels following treatment with autologous BMC that persisted through 12 months. Patients reporting improved outcomes (46 of 72 knees) received BMC injections having higher CFU-F concentrations than non-responding patients (21.1×103 ± 12.4×103 vs 14.3×103 ± 7.0 x103 CFU-F per mL). A progenitor cell concentration of 18×103 CFU-F per mL of BMC was found to best differentiate responders from non-responders. CONCLUSION This study provides supportive evidence for using autologous BMC in the minimally invasive treatment of knee OA and suggests that increased progenitor cell content leads to improved treatment outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03011398, 1/7/17.
Collapse
Affiliation(s)
- Christopher J Centeno
- Centeno-Schultz Clinic, Broomfield, CO, USA
- Regenexx, Research and Development, Broomfield, CO, USA
| | | | | | - Ehren Dodson
- Regenexx, Research and Development, Broomfield, CO, USA
| | | | | |
Collapse
|
7
|
Yoon DS, Lee KM, Choi Y, Ko EA, Lee NH, Cho S, Park KH, Lee JH, Kim HW, Lee JW. TLR4 downregulation by the RNA-binding protein PUM1 alleviates cellular aging and osteoarthritis. Cell Death Differ 2022; 29:1364-1378. [PMID: 35034101 DOI: 10.1038/s41418-021-00925-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 12/27/2022] Open
Abstract
Dysfunction of mRNA or RNA-binding proteins (RBPs) causes cellular aging and age-related degenerative diseases; however, information regarding the mechanism through which RBP-mediated posttranscriptional regulation affects cellular aging and related disease processes is limited. In this study, PUM1 was found to be associated with the self-renewal capacity and aging process of human mesenchymal stem cells (MSC). PUM1 interacted with the 3'-untranslated region of Toll-like receptor 4 (TLR4) to suppress TLR4 mRNA translation and regulate the activity of nuclear factor-κB (NF-κB), a master regulator of the aging process in MSCs. PUM1 overexpression protected MSCs against H2O2-induced cellular senescence by suppressing TLR4-mediated NF-κB activity. TLR4-mediated NF-κB activation is a key regulator in osteoarthritis (OA) pathogenesis. PUM1 overexpression enhanced the chondrogenic potential of MSCs even under the influence of inflammation-inducing factors, such as lipopolysaccharide (LPS) or interleukin-1β (IL-1β), whereas the chondrogenic potential was reduced following the PUM1 knockdown-mediated TLR4 activation. PUM1 levels decreased under inflammatory conditions in vitro and during OA progression in human and mouse disease models. PUM1 knockdown in human chondrocytes promoted chondrogenic phenotype loss, whereas PUM1 overexpression protected the cells from inflammation-mediated disruption of the chondrogenic phenotype. Gene therapy using a lentiviral vector encoding mouse PUM1 showed promise in preserving articular cartilage integrity in OA mouse models. In conclusion, PUM1 is a novel suppressor of MSC aging, and the PUM1-TLR4 regulatory axis represents a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Dong Suk Yoon
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yoorim Choi
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, South Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea
| | - Sehee Cho
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, South Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, South Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, South Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, South Korea. .,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea. .,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, South Korea. .,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, South Korea.
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
8
|
Mareschi K, Marini E, Niclot AGSB, Barone M, Pinnetta G, Adamini A, Spadea M, Labanca L, Lucania G, Ferrero I, Fagioli F. A New Human Platelet Lysate for Mesenchymal Stem Cell Production Compliant with Good Manufacturing Practice Conditions. Int J Mol Sci 2022; 23:3234. [PMID: 35328655 PMCID: PMC8953582 DOI: 10.3390/ijms23063234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are classified as advanced therapy medicinal products, a new category of GMP (good manufacturing practice)-compliant medicines for clinical use. We isolated MSCs from 5 bone marrow (BM) samples using human platelet lysate (HPL) instead of foetal bovine serum (FBS). We used a new method of HPL production consisting of treating platelet (PLTs) pools with Ca-Gluconate to form a gel clot, then mechanically squeezing to release growth factors. We compared the new HPL (HPL-S) with the standard (HPL-E) obtained by freezing/thawing cycles and by adding heparin. HPL-S had not PLTs and fibrinogen but the quantity of proteins and growth factors was comparable to HPL-E. Therefore, HPL-S needed fewer production steps to be in compliance with GMP conditions. The number of colonies forming unit-fibroblasts (CFU-F) and the maintenance of stem markers showed no significant differences between MSCs with HPL-E and HPL-S. The cumulative population doubling was higher in MSCs with HPL-E in the earlier passages, but we observed an inverted trend of cell growth at the fourth passage. Immunophenotypic analysis showed a significant lower expression of HLA-DR in the MSCs with HPL-S (1.30%) than HPL-E (14.10%). In conclusion, we demonstrated that HPL-S is an effective alternative for MSC production under GMP conditions.
Collapse
Affiliation(s)
- Katia Mareschi
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N.); (M.B.); (M.S.); (F.F.)
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (G.P.); (A.A.); (I.F.)
| | - Elena Marini
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N.); (M.B.); (M.S.); (F.F.)
| | - Alessia Giovanna Santa Banche Niclot
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N.); (M.B.); (M.S.); (F.F.)
| | - Marta Barone
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N.); (M.B.); (M.S.); (F.F.)
| | - Giuseppe Pinnetta
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (G.P.); (A.A.); (I.F.)
| | - Aloe Adamini
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (G.P.); (A.A.); (I.F.)
| | - Manuela Spadea
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N.); (M.B.); (M.S.); (F.F.)
| | - Luciana Labanca
- Blood Component Production and Validation Center, City of Health and Science of Turin, S. Anna Hospital, 10126 Turin, Italy; (L.L.); (G.L.)
| | - Graziella Lucania
- Blood Component Production and Validation Center, City of Health and Science of Turin, S. Anna Hospital, 10126 Turin, Italy; (L.L.); (G.L.)
| | - Ivana Ferrero
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (G.P.); (A.A.); (I.F.)
| | - Franca Fagioli
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N.); (M.B.); (M.S.); (F.F.)
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (G.P.); (A.A.); (I.F.)
| |
Collapse
|
9
|
Teufelsbauer M, Lang C, Plangger A, Rath B, Moser D, Staud C, Radtke C, Neumayer C, Hamilton G. Effects of metformin on human bone-derived mesenchymal stromal cell-breast cancer cell line interactions. Med Oncol 2022; 39:54. [PMID: 35150338 PMCID: PMC8840908 DOI: 10.1007/s12032-022-01655-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
Metformin is used to treat patients with type 2 diabetes mellitus and was found to lower the incidence of cancer. Bone metastasis is a common impairment associated with advanced breast cancer. The present study investigated the effects of metformin on human bone-derived mesenchymal stromal cells (BM-MSC)—breast cancer cell line interactions. BM-MSCs grown from box chisels were tested for growth-stimulating and migration-controlling activity on four breast cancer cell lines either untreated or after pretreatment with metformin. Growth stimulation was tested in MTT tests and migration in scratch assays. Furthermore, the expression of adipokines of BM-MSCs in response to metformin was assessed using Western blot arrays. Compared to breast cancer cell lines (3.6 ± 1.4% reduction of proliferation), 500 µM metformin significantly inhibited the proliferation of BM-MSC lines (mean 12.3 ± 2.2 reduction). Pretreatment of BM-MSCs with metformin showed variable effects of the resulting conditioned media (CM) on breast cancer cell lines depending on the specific BM-MSC—cancer line combination. Metformin significantly reduced the migration of breast cancer cell lines MDA-MB-231 and MDA-MB-436 in response to CM of drug-pretreated BM-MSCs. Assessment of metformin-induced alterations in the expression of adipokines by BM-MSC CM indicated increased osteogenic signaling and possibly impairment of metastasis. In conclusion, the anticancer activities of metformin are the result of a range of direct and indirect mechanisms that lower tumor proliferation and progression. A lower metformin-induced protumor activity of BM-MSCs in the bone microenvironment seem to contribute to the positive effects of the drug in selected breast cancer patients.
Collapse
Affiliation(s)
- Maryana Teufelsbauer
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Clemens Lang
- Department of Trauma Surgery, Sozialmedizinisches Zentrum Ost, Donauspital, Vienna, Austria
| | - Adelina Plangger
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Doris Moser
- Department of Cranio, Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Neumayer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
10
|
Canosa S, Mareschi K, Marini E, Carosso AR, Castiglia S, Rustichelli D, Ferrero I, Gennarelli G, Bussolati B, Nocifora A, Asnaghi V, Bergallo M, Isidoro C, Benedetto C, Revelli A, Fagioli F. A Novel Xeno-Free Method to Isolate Human Endometrial Mesenchymal Stromal Cells (E-MSCs) in Good Manufacturing Practice (GMP) Conditions. Int J Mol Sci 2022; 23:ijms23041931. [PMID: 35216052 PMCID: PMC8876308 DOI: 10.3390/ijms23041931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/05/2022] [Accepted: 02/06/2022] [Indexed: 11/16/2022] Open
Abstract
The cyclic regeneration of human endometrium is guaranteed by the proliferative capacity of endometrial mesenchymal stromal cells (E-MSCs). Due to this, the autologous infusion of E-MSCs has been proposed to support endometrial growth in a wide range of gynecological diseases. We aimed to compare two different endometrial sampling methods, surgical curettage and vacuum aspiration biopsy random assay (VABRA), and to validate a novel xeno-free method to culture human E-MSCs. Six E-MSCs cell samples were isolated after mechanical tissue homogenization and cultured using human platelet lysate. E-MSCs were characterized for the colony formation capacity, proliferative potential, and multilineage differentiation. The expression of mesenchymal and stemness markers were tested by FACS analysis and real-time PCR, respectively. Chromosomal alterations were evaluated by karyotype analysis, whereas tumorigenic capacity and invasiveness were tested by soft agar assay. Both endometrial sampling techniques allowed efficient isolation and expansion of E-MSCs using a xeno-free method, preserving their mesenchymal and stemness phenotype, proliferative potential, and limited multi-lineage differentiation ability during the culture. No chromosomal alterations and invasive/tumorigenic capacity were observed. Herein, we report the first evidence of efficient E-MSCs isolation and culture in Good Manufacturing Practice compliance conditions, suggesting VABRA endometrial sampling as alternative to surgical curettage.
Collapse
Affiliation(s)
- Stefano Canosa
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, S. Anna Hospital, Department of Surgical Sciences, University of Torino, 10126 Torino, Italy; (S.C.); (A.R.C.); (G.G.); (C.B.); (A.R.)
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Torino, 10126 Torino, Italy; (E.M.); (M.B.); (F.F.)
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy; (S.C.); (D.R.); (I.F.)
- Correspondence: ; Tel.: +39-(011)-313-5420
| | - Elena Marini
- Department of Public Health and Paediatrics, University of Torino, 10126 Torino, Italy; (E.M.); (M.B.); (F.F.)
| | - Andrea Roberto Carosso
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, S. Anna Hospital, Department of Surgical Sciences, University of Torino, 10126 Torino, Italy; (S.C.); (A.R.C.); (G.G.); (C.B.); (A.R.)
| | - Sara Castiglia
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy; (S.C.); (D.R.); (I.F.)
| | - Deborah Rustichelli
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy; (S.C.); (D.R.); (I.F.)
| | - Ivana Ferrero
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy; (S.C.); (D.R.); (I.F.)
| | - Gianluca Gennarelli
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, S. Anna Hospital, Department of Surgical Sciences, University of Torino, 10126 Torino, Italy; (S.C.); (A.R.C.); (G.G.); (C.B.); (A.R.)
| | - Benedetta Bussolati
- Molecular Biotechnology Centre, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Alberto Nocifora
- Department of Oncology, Pathology Unit, University of Torino, 10126 Torino, Italy;
| | - Valentina Asnaghi
- Department of Laboratory Medicine, Medical Genetics Division, City of Health and Science of Torino, 10124 Torino, Italy;
| | - Massimiliano Bergallo
- Department of Public Health and Paediatrics, University of Torino, 10126 Torino, Italy; (E.M.); (M.B.); (F.F.)
- Paediatric Laboratory Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Ciro Isidoro
- Department of Health Sciences, University of Piemonte Orientale, 13100 Novara, Italy;
| | - Chiara Benedetto
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, S. Anna Hospital, Department of Surgical Sciences, University of Torino, 10126 Torino, Italy; (S.C.); (A.R.C.); (G.G.); (C.B.); (A.R.)
| | - Alberto Revelli
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, S. Anna Hospital, Department of Surgical Sciences, University of Torino, 10126 Torino, Italy; (S.C.); (A.R.C.); (G.G.); (C.B.); (A.R.)
| | - Franca Fagioli
- Department of Public Health and Paediatrics, University of Torino, 10126 Torino, Italy; (E.M.); (M.B.); (F.F.)
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy; (S.C.); (D.R.); (I.F.)
| |
Collapse
|
11
|
Jeyaraman M, Bingi SK, Muthu S, Jeyaraman N, Packkyarathinam RP, Ranjan R, Sharma S, Jha SK, Khanna M, Rajendran SNS, Rajendran RL, Gangadaran P. Impact of the Process Variables on the Yield of Mesenchymal Stromal Cells from Bone Marrow Aspirate Concentrate. Bioengineering (Basel) 2022; 9:57. [PMID: 35200410 PMCID: PMC8869489 DOI: 10.3390/bioengineering9020057] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Human bone marrow (BM) has been highlighted as a promising source of mesenchymal stromal cells (MSCs) containing various growth factors and cytokines that can be potentially utilized in regenerative procedures involving cartilage and bone. However, the proportion of MSCs in the nucleated cell population of BM is only around 0.001% to 0.01% thereby making the harvesting and processing technique crucial for obtaining optimal results upon its use in various regenerative processes. Although several studies in the literature have given encouraging results on the utility of BM aspiration concentrate (BMAC) in various regenerative procedures, there is a lack of consensus concerning the harvesting variables such as choice of anesthetic agent to be used, site of harvest, size of the syringe to be used, anticoagulant of choice, and processing variables such as centrifugation time, and speed. In this review article, we aim to discuss the variables in the harvesting and processing technique of BMAC and their impact on the yield of MSCs in the final concentrate obtained from them.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine, Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India;
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India;
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, India; (S.K.B.); (M.K.)
| | - Shiva Kumar Bingi
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, India; (S.K.B.); (M.K.)
- Fellow in Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow 226010, India
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India;
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, India; (S.K.B.); (M.K.)
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, India
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, India; (S.K.B.); (M.K.)
- Fellow in Orthopaedic Rheumatology, Dr. RML National Law University, Lucknow 226010, India
- Fellow in Joint Replacement, Department of Orthopaedics, Atlas Hospitals, Tiruchirappalli 620002, India
| | | | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201310, India;
| | - Shilpa Sharma
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India;
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, India; (S.K.B.); (M.K.)
- Department of Orthopaedics, Prasad Institute of Medical Sciences, Lucknow 226401, India
| | - Sree Naga Sowndary Rajendran
- Department of Medicine, Sri Venkateshwaraa Medical College Hospital and Research Centre, Puducherry 605102, India;
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
12
|
Potapnev MP. Analysis of approaches to increase the efficacy of cell therapy based on mesenchymal stromal cells. GENES & CELLS 2021; 16:22-28. [DOI: 10.23868/202112003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The review considers the main stages of isolating, processing and clinical use of human mesenchymal stromal cells (MSCs). They included: donor selection, selection of the source of MSCs, methods of isolation of cellular suspension from tissue, culturing in vitro for cell biomass propagation, priming of the resulting cell product, timing and ways of its clinical application, selection of the recipient of MSCs. The analysis of the stages of MSCs preparation and conditions for their use was carried out from the position of the influence on the final therapeutic effect of cell therapy in patients (or experimental animals - in preclinical studies). The optimal parameters of work with MSCs at each stage, the possibility to improve their quality / biological activity in order to increase their therapeutic efficacy were determined. The analysis and ways of avoiding the influence of adverse factors associated with the manufacturing and use of MSCs on the effectiveness of cell therapy in patients were given.
Collapse
|
13
|
Yigitturk G, Erbas O, Karabay Yavasoglu NU, Acikgoz E, Buhur A, Gokhan A, Gurel C, Gunduz C, Yavasoglu A. The neuro-restorative effect of adipose-derived mesenchymal stem cell transplantation on a mouse model of diabetic neuropathy. Neurol Res 2021; 44:156-164. [PMID: 34410214 DOI: 10.1080/01616412.2021.1967679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Diabetic neuropathy (DN) is the most common degenerative complication associated with Diabetes Mellitus. Despite widespread awareness about DN, the only effective treatments are blood glucose control and pain management. The aim of the current study was to determine the effect of intramuscular adipose-derived mesenchymal stem cell (AMSC) transplantation on sciatic nerves in DN using EMG and histological analyses. A total of 27 mice were randomly divided into three groups: control group, DN group and AMSC group. In EMG, CMAP amplitude in the sciatic nerves was lower, but distal latency was higher in the DN group compared with the control group. CMAP amplitude in the sciatic nerves was higher in the AMSC group compared with the DN group. Distal latency in the sciatic nerve was lower in the AMSC group compared with the DN group. Histologic examination of the tissues in the animals treated with AMSC showed a remarkable improvement in microscopic morphology. Fluorescence microscopy analyses demonstrated that intramuscularly transplanted AMSC was selectively localized in the sciatic nerves. Transplantation of AMSC increased protein expression of S100, cdk2, NGF and DHH, all of which, interfered with DN onset in sciatic nerves. The findings of the present study suggest that AMSC transplantation improved DN through a signal-regulatory effect on Schwann cells, neurotrophic actions and restoration of myelination.
Collapse
Affiliation(s)
- Gurkan Yigitturk
- Department Of Histology And Embryology, Faculty Of Medicine, Muğla Sıtkı Koçman University, Mugla, Turkey
| | - Oytun Erbas
- Department Of Physiology, Faculty Of Medicine, Bilim University, Istanbul, Turkey
| | | | - Eda Acikgoz
- Department Of Histology And Embryology, Faculty Of Medicine, Van Yüzüncü Yıl University, Izmir, Turkey
| | - Aylin Buhur
- Department Of Histology And Embryology, Faculty Of Medicine, Ege University, Izmir, Turkey
| | - Aylin Gokhan
- Department Of Histology And Embryology, Faculty Of Medicine, Ege University, Izmir, Turkey
| | - Cevik Gurel
- Department Of Histology And Embryology, Faculty Of Medicine, Ege University, Izmir, Turkey
| | - Cumhur Gunduz
- Department Of Medical Biology, Faculty Of Medicine, Ege University, Izmir, Turkey
| | - Altug Yavasoglu
- Department Of Histology And Embryology, Faculty Of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
14
|
Spohn G, Witte AS, Kretschmer A, Seifried E, Schäfer R. More Human BM-MSC With Similar Subpopulation Composition and Functional Characteristics Can Be Produced With a GMP-Compatible Fabric Filter System Compared to Density Gradient Technique. Front Cell Dev Biol 2021; 9:638798. [PMID: 33869188 PMCID: PMC8044851 DOI: 10.3389/fcell.2021.638798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs), multipotent progenitors that can be isolated from a variety of different tissues, are becoming increasingly important as cell therapeutics targeting immunopathologies and tissue regeneration. Current protocols for MSC isolation from bone marrow (BM) rely on density gradient centrifugation (DGC), and the production of sufficient MSC doses is a critical factor for conducting clinical MSC trials. Previously, a Good Manufacturing Practice (GMP)–compatible non-woven fabric filter device system to isolate MSCs was developed to increase the MSC yield from the BM. The aim of our study was to compare high-resolution phenotypic and functional characteristics of BM-MSCs isolated with this device and with standard DGC technology. Methods Human BM samples from 5 donors were analyzed. Each sample was divided equally, processing by DGC, and with the filter device. Stem cell content was assessed by quantification of colony-forming units fibroblasts (CFU-F). Immunophenotype was analyzed by multicolor flow cytometry. In vitro trilineage differentiation potential, trophic factors, and IDO-1 production were assessed. Functionally, immunomodulatory potential, wound healing, and angiogenesis were assayed in vitro. Results The CFU-F yield was 15-fold higher in the MSC preparations isolated with the device compared to those isolated by DGC. Consequently, the MSC yield that could be manufactured at passage 3 per mL collected BM was more than 10 times higher in the device group compared to DGC (1.65 × 109 vs. 1.45 × 108). The immunomodulatory potential and IDO-1 production showed donor-to-donor variabilities without differences between fabric filter-isolated and DGC-isolated MSCs. The results from the wound closure assays, the tube formation assays, and the trilineage differentiation assays were similar between the groups with respect to the isolation method. Sixty-four MSC subpopulations could be quantified with CD140a+CD119+CD146+ as most common phenotype group, and CD140a+CD119+CD146+MSCA-1–CD106–CD271– and CD140a+CD119+CD146–MSCA-1–CD106–CD271– as most frequent MSC subpopulations. As trophic factors hepatocyte growth factor, epidermal growth factor, brain-derived neurotrophic factor, angiopoietin-1, and vascular endothelial growth factor A could be detected in both groups with considerable variability between donors, but independent of the respective MSC isolation technique. Conclusion The isolation of MSCs using a GMP-compatible fabric filter system device resulted in higher yield of CFU-F, producing substantially more MSCs with similar subpopulation composition and functional characteristics as MSCs isolated by DGC.
Collapse
Affiliation(s)
- Gabriele Spohn
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, Frankfurt am Main, Germany
| | - Anne-Sophie Witte
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, Frankfurt am Main, Germany
| | - Anja Kretschmer
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, Frankfurt am Main, Germany
| | - Erhard Seifried
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, Frankfurt am Main, Germany
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, Frankfurt am Main, Germany
| |
Collapse
|
15
|
Cryopreserved bone marrow aspirate concentrate as a cell source for the colony-forming unit fibroblast assay. Cytotherapy 2020; 22:486-493. [DOI: 10.1016/j.jcyt.2020.04.091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 03/31/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022]
|
16
|
Mesenchymal Stem/Progenitor Cells: The Prospect of Human Clinical Translation. Stem Cells Int 2020; 2020:8837654. [PMID: 33953753 PMCID: PMC8063852 DOI: 10.1155/2020/8837654] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/19/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) are key players in regenerative medicine, relying principally on their differentiation/regeneration potential, immunomodulatory properties, paracrine effects, and potent homing ability with minimal if any ethical concerns. Even though multiple preclinical and clinical studies have demonstrated remarkable properties for MSCs, the clinical applicability of MSC-based therapies is still questionable. Several challenges exist that critically hinder a successful clinical translation of MSC-based therapies, including but not limited to heterogeneity of their populations, variability in their quality and quantity, donor-related factors, discrepancies in protocols for isolation, in vitro expansion and premodification, and variability in methods of cell delivery, dosing, and cell homing. Alterations of MSC viability, proliferation, properties, and/or function are also affected by various drugs and chemicals. Moreover, significant safety concerns exist due to possible teratogenic/neoplastic potential and transmission of infectious diseases. Through the current review, we aim to highlight the major challenges facing MSCs' human clinical translation and shed light on the undergoing strategies to overcome them.
Collapse
|
17
|
Mareschi K, Castiglia S, Adamini A, Rustichelli D, Marini E, Banche Niclot AGS, Bergallo M, Labanca L, Ferrero I, Fagioli F. Inactivated Platelet Lysate Supports the Proliferation and Immunomodulant Characteristics of Mesenchymal Stromal Cells in GMP Culture Conditions. Biomedicines 2020; 8:biomedicines8070220. [PMID: 32708843 PMCID: PMC7400095 DOI: 10.3390/biomedicines8070220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) isolated from bone marrow (BM-MSCs) are considered advanced therapy medicinal products (ATMPs) and need to be produced according to good manufacturing practice (GMP) in their clinical use. Human platelet lysate (HPL) is a good GMP-compliant alternative to animal serum, and we have demonstrated that after pathogen inactivation with psoralen, it was safer and more efficient to use psoralen in the production of MSCs following GMP guidelines. In this study, the MSCs cultivated in fetal bovine serum (FBS-MSC) or inactivated HPL (iHPL-MSC) were compared for their immunomodulatory properties. We studied the effects of MSCs on (1) the proliferation of total lymphocytes (Ly) and on naïve T Ly subsets induced to differentiate in Th1 versus Th2 Ly; (2) the immunophenotype of different T-cell subsets; (3) and the cytokine release to verify Th1, Th2, and Th17 polarization. These were analyzed by using an in vitro co-culture system. We observed that iHPL-MSCs showed the same immunomodulatory properties observed in the FBS-MSC co-cultures. Furthermore, a more efficient effect on the increase of naïve T- cells and in the Th1 cytokine release from iHPL was observed. This study confirms that iHPL, used as a medium supplement, may be considered a good alternative to FBS for a GMP-compliant MSC expansion, and also to preserve their immunomodulatory proprieties.
Collapse
Affiliation(s)
- Katia Mareschi
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N); (M.B.); (F.F.)
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (S.C.); (A.A.); (D.R.); (I.F.)
- Correspondence: ; Tel.: +39-11-3135420
| | - Sara Castiglia
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (S.C.); (A.A.); (D.R.); (I.F.)
| | - Aloe Adamini
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (S.C.); (A.A.); (D.R.); (I.F.)
| | - Deborah Rustichelli
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (S.C.); (A.A.); (D.R.); (I.F.)
| | - Elena Marini
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N); (M.B.); (F.F.)
| | - Alessia Giovanna Santa Banche Niclot
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N); (M.B.); (F.F.)
| | - Massimiliano Bergallo
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N); (M.B.); (F.F.)
| | - Luciana Labanca
- Blood Component Production and Validation Center, City of Health and Science of Turin, S. Anna Hospital, 10126 Turin, Italy;
| | - Ivana Ferrero
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (S.C.); (A.A.); (D.R.); (I.F.)
| | - Franca Fagioli
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, 10126 Torino, Italy; (E.M.); (A.G.S.B.N); (M.B.); (F.F.)
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children’s Hospital, City of Health and Science of Turin, 10126 Torino, Italy; (S.C.); (A.A.); (D.R.); (I.F.)
| |
Collapse
|
18
|
Kiiskinen J, Merivaara A, Hakkarainen T, Kääriäinen M, Miettinen S, Yliperttula M, Koivuniemi R. Nanofibrillar cellulose wound dressing supports the growth and characteristics of human mesenchymal stem/stromal cells without cell adhesion coatings. Stem Cell Res Ther 2019; 10:292. [PMID: 31547864 PMCID: PMC6757411 DOI: 10.1186/s13287-019-1394-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/15/2019] [Accepted: 08/22/2019] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND In the field of regenerative medicine, delivery of human adipose-derived mesenchymal stem/stromal cells (hASCs) has shown great promise to promote wound healing. However, a hostile environment of the injured tissue has shown considerably to limit the survival rate of the transplanted cells, and thus, to improve the cell survival and retention towards successful cell transplantation, an optimal cell scaffold is required. The objective of this study was to evaluate the potential use of wood-derived nanofibrillar cellulose (NFC) wound dressing as a cell scaffold material for hASCs in order to develop a cell transplantation method free from animal-derived components for wound treatment. METHODS Patient-derived hASCs were cultured on NFC wound dressing without cell adhesion coatings. Cell characteristics, including cell viability, morphology, cytoskeletal structure, proliferation potency, and mesenchymal cell and differentiation marker expression, were analyzed using cell viability assays, electron microscopy, immunocytochemistry, and quantitative or reverse transcriptase PCR. Student's t test and one-way ANOVA followed by a Tukey honestly significant difference post hoc test were used to determine statistical significance. RESULTS hASCs were able to adhere to NFC dressing and maintained high cell survival without cell adhesion coatings with a cell density-dependent manner for the studied period of 2 weeks. In addition, NFC dressing did not induce any remarkable cytotoxicity towards hASCs or alter the morphology, proliferation potency, filamentous actin structure, the expression of mesenchymal vimentin and extracellular matrix (ECM) proteins collagen I and fibronectin, or the undifferentiated state of hASCs. CONCLUSIONS As a result, NFC wound dressing offers a functional cell culture platform for hASCs to be used further for in vivo wound healing studies in the future.
Collapse
Affiliation(s)
- Jasmi Kiiskinen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Arto Merivaara
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Tiina Hakkarainen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Minna Kääriäinen
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Marjo Yliperttula
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland
| | - Raili Koivuniemi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014, Helsinki, Finland.
| |
Collapse
|
19
|
Das R, Roosloot R, van Pel M, Schepers K, Driessen M, Fibbe WE, de Bruijn JD, Roelofs H. Preparing for cell culture scale-out: establishing parity of bioreactor- and flask-expanded mesenchymal stromal cell cultures. J Transl Med 2019; 17:241. [PMID: 31340829 PMCID: PMC6657181 DOI: 10.1186/s12967-019-1989-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Cell-based therapies have the potential to become treatment options for many diseases, but efficient scale-out of these therapies has proven to be a major hurdle. Bioreactors can be used to overcome this hurdle, but changing the culture method can introduce unwanted changes to the cell product. Therefore, it is important to establish parity between products generated using traditional methods versus those generated using a bioreactor. Methods Mesenchymal stromal cells (MSCs) are cultured in parallel using either traditional culture flasks, spinner vessels or a new bioreactor system. To investigate parity between the cells obtained from different methods, harvested cells are compared in terms of yield, phenotype and functionality. Results Bioreactor-based expansion yielded high cell numbers (222–510 million cells). Highest cell expansion was observed upon culture in flasks [average 5.0 population doublings (PDL)], followed by bioreactor (4.0 PDL) and spinner flasks (3.3 PDL). Flow cytometry confirmed MSC identity (CD73+, CD90+ and CD105+) and lack of contaminating hematopoietic cell populations. Cultured MSCs did not display genetic aberrations and no difference in differentiation and immunomodulatory capacity was observed between culture conditions. The response to IFNγ stimulation was similar for cells obtained from all culture conditions, as was the capacity to inhibit T cell proliferation. Conclusions The new bioreactor technology can be used to culture large amounts of cells with characteristics equivalent to those cultured using traditional, flask based, methods. Electronic supplementary material The online version of this article (10.1186/s12967-019-1989-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ruud Das
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands.
| | - Rens Roosloot
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands
| | - Melissa van Pel
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Koen Schepers
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Marijn Driessen
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands
| | - Willem E Fibbe
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Joost Dick de Bruijn
- Scinus Cell Expansion BV, Professor Bronkhorstlaan 10, Building 48, 3723 MB, Bilthoven, The Netherlands.,Twente University, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands.,Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Helene Roelofs
- Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
20
|
Antebi B, Walker KP, Mohammadipoor A, Rodriguez LA, Moore RK, Cancio LC, Batchinsky AI. Bench-to-bedside optimization of mesenchymal stem cell isolation, processing, and expansion for in vivo administration. Regen Med 2019; 14:279-293. [PMID: 31070521 DOI: 10.2217/rme-2018-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: In this study, we aimed at identifying the optimal conditions for isolation, processing and expansion of mesenchymal stem cells (MSCs). Methods: Porcine bone marrow was obtained from either small- or large-volume bone marrow aspirate (BMA). Next, three BMA processing methods were compared. Finally, the best condition was selected from various culture parameters, including basal media, supplementation and seeding density. Results: Our results demonstrate that a small-volume BMA and direct plating yields significantly higher concentration of MSCs. Basal media supplementation with 10% platelet lysate and seeding density of 1000 cells/cm2 can generate large numbers of multipotent MSCs with augmented function and low population doublings. Conclusion: This work provides guidance for preparation of robust MSCs for future clinical trials.
Collapse
Affiliation(s)
- Ben Antebi
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Kerfoot P Walker
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.,Oak Ridge Institute for Science & Education, Oak Ridge, TN 37831-0117, USA
| | - Arezoo Mohammadipoor
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.,Oak Ridge Institute for Science & Education, Oak Ridge, TN 37831-0117, USA
| | - Luis A Rodriguez
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Robbie K Moore
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Leopoldo C Cancio
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA
| | - Andriy I Batchinsky
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.,The Geneva Foundation, Tacoma, WA 98402, USA
| |
Collapse
|
21
|
Sfougataki I, Grafakos I, Varela I, Mitrakos A, Karagiannidou A, Tzannoudaki M, Poulou M, Mertzanian A, Roubelakis G. M, Stefanaki K, Traeger-Synodinos J, Kanavakis E, Kitra V, Tzetis M, Goussetis E. Reprogramming of bone marrow derived mesenchymal stromal cells to human induced pluripotent stem cells from pediatric patients with hematological diseases using a commercial mRNA kit. Blood Cells Mol Dis 2019; 76:32-39. [DOI: 10.1016/j.bcmd.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 02/01/2023]
|
22
|
McCutchan A, Dobson GP, Stewart N, Letson HL, Grant AL, Jovanovic IA, Hazratwala K, Wilkinson M, McEwen P, Morris J. Absence of cytotoxic and inflammatory effects following in vitro exposure of chondrogenically-differentiated human mesenchymal stem cells to adenosine, lidocaine and Mg 2+ solution. J Exp Orthop 2019; 6:16. [PMID: 30989345 PMCID: PMC6465392 DOI: 10.1186/s40634-019-0185-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background ALM solution, a combination of adenosine, lidocaine and Mg2+, is an emerging small volume therapy that has been shown to prevent and correct coagulopathy and surgery-related inflammation in preclinical models, though its application in orthopaedic surgery is yet to be demonstrated. The effect of ALM solution on chondrocytes is unknown. The aim of this preliminary study was to investigate the effect of ALM solution on viability and inflammatory responses of chondrogenically-differentiated human bone marrow-derived mesenchymal stem cells (chondro-MSC), in vitro. Methods Chondro-MSC were exposed to media only, saline (0.9% NaCl or 1.3% NaCl) only, or saline containing ALM (1 mM adenosine, 3 mM lidocaine, 2.5 mM Mg2+) or tranexamic acid (TXA, 100 mg/ml) for 1 or 4 h. Responses to ALM solutions containing higher lidocaine concentrations were also compared. Chondrocyte viability was determined using WST-8 colorimetric assays and inflammatory cytokine (TNF-α, IL-1β, IL-8) and matrix metalloproteinases (MMP-3, MMP-12, MMP-13) concentrations using multiplex bead arrays. Results The viability of chondro-MSC was significantly greater after 1 h treatment with ALM compared to saline (96.2 ± 7.9 versus 75.6 ± 7.3%). Extension of exposure times to 4 h had no significant adverse effect on cell viability after treatment with ALM (1 h, 85.4 ± 5.6 v 4 h, 74.0 ± 15.2%). Cytotoxicity was evident following exposure to solutions containing lidocaine concentrations greater than 30 mM. There were no significant differences in viability (80 ± 5.4 v 57.3 ± 16.2%) or secretion of IL-8 (60 ± 20 v 160 ± 50 pg/ml), MMP-3 (0.95 ± 0.6 v 3.4 ± 1.6 ng/ml), and MMP-13 (4.2 ± 2.4 v 9.2 ± 4.3 ng/ml) in chondro-MSC exposed to saline, ALM or TXA. Conclusions Short-term, in vitro exposure to clinically-relevant concentrations of ALM solution had no adverse inflammatory or chondrotoxic effects on human chondro-MSC, with responses comparable to saline and TXA. These findings provide support for continued evaluation of ALM solution as a possible therapeutic to improve outcomes following orthopaedic procedures.
Collapse
Affiliation(s)
- Andrew McCutchan
- Department of Haematology and Bone Marrow Transplantation, Townsville Hospital, Townsville, Australia
| | - Geoffrey P Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Natalie Stewart
- Department of Haematology and Bone Marrow Transplantation, Townsville Hospital, Townsville, Australia
| | - Hayley L Letson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia
| | - Andrea L Grant
- The Orthopaedic Research Institute of Queensland, 7 Turner St, Pimlico, Townsville, Q 4812, Australia
| | | | - Kaushik Hazratwala
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia.,The Orthopaedic Research Institute of Queensland, 7 Turner St, Pimlico, Townsville, Q 4812, Australia
| | - Matthew Wilkinson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia.,The Orthopaedic Research Institute of Queensland, 7 Turner St, Pimlico, Townsville, Q 4812, Australia
| | - Peter McEwen
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia.,The Orthopaedic Research Institute of Queensland, 7 Turner St, Pimlico, Townsville, Q 4812, Australia
| | - Jodie Morris
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, Australia. .,The Orthopaedic Research Institute of Queensland, 7 Turner St, Pimlico, Townsville, Q 4812, Australia.
| |
Collapse
|
23
|
Experimental Strategies of Mesenchymal Stem Cell Propagation: Adverse Events and Potential Risk of Functional Changes. Stem Cells Int 2019; 2019:7012692. [PMID: 30956673 PMCID: PMC6431404 DOI: 10.1155/2019/7012692] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/28/2018] [Accepted: 01/13/2019] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are attractive candidates for cell-based tissue repair approaches. Hundreds of clinical trials using MSCs have been completed and many others are still being investigated. For most therapeutic applications, MSC propagation in vitro is often required. However, ex vivo culture condition is not fully physiological and may affect biological properties of MSCs including their regenerative potential. Moreover, both cell cryopreservation and labelling procedure prior to infusion may have the negative impact on their expected effect in vivo. The incidence of MSC transformation during in vitro culture should be also taken into consideration before using cells in stem cell therapy. In our review, we focused on different aspects of MSC propagation that might influence their regenerative properties of MSC. We also discussed the influence of different factors that might abolish MSC proliferation and differentiation as well as potential impact of stem cell senescence and aging. Despite of many positive therapeutic effects of MSC therapy, one has to be conscious about potential cell changes that could appear during manufacturing of MSCs.
Collapse
|
24
|
In Vitro Mesenchymal Progenitor Cell Expansion is a Predictor of Transplant-related Mortality and acute GvHD III-IV After Bone Marrow Transplantation in Univariate Analysis: A Large Single-Center Experience. J Pediatr Hematol Oncol 2019; 41:42-46. [PMID: 30113355 DOI: 10.1097/mph.0000000000001281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent stem cells able to differentiate into mesenchymal origin tissue and support the growth of hematopoietic stem cells. In order to understand the role of MSCs infused in bone marrow grafts, 53 consecutive patients were analyzed for engraftment, acute and chronic graft-versus-host disease (GvHD), transplant-related mortality (TRM), relapse incidence, and overall survival. The MSC content was measured as MSC expansion at the second passage. When in vitro-expanded MSC (cumulative population doubling at second passage, cPDp2) values were stratified according to the median value (2.2-fold increase), the univariate analysis showed a significant difference in TRM (23% vs. 3.8%, P=0.05.) and in acute GvHD III-IV incidence (12% vs. 4%, P=0.04), while the multivariate analysis did not confirm its independent role. No clinical parameters in donors and recipients were identified as predictors of cPDp2 expansion. Our study suggests a role for short-term ex vivo-expanded MSCs in reduced aGVHD III-IV incidence and TRM in univariate analysis. A multicenter, larger study is warranted to confirm these data.
Collapse
|
25
|
Wu C, Chen L, Huang YZ, Huang Y, Parolini O, Zhong Q, Tian X, Deng L. Comparison of the Proliferation and Differentiation Potential of Human Urine-, Placenta Decidua Basalis-, and Bone Marrow-Derived Stem Cells. Stem Cells Int 2018; 2018:7131532. [PMID: 30651734 PMCID: PMC6311712 DOI: 10.1155/2018/7131532] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/05/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023] Open
Abstract
Human multipotent stem cell-based therapies have shown remarkable potential in regenerative medicine and tissue engineering applications due to their abilities of self-renewal and differentiation into multiple adult cell types under appropriate conditions. Presently, human multipotent stem cells can be isolated from different sources, but variation among their basic biology can result in suboptimal selection of seed cells in preclinical and clinical research. Thus, the goal of this study was to compare the biological characteristics of multipotent stem cells isolated from human bone marrow, placental decidua basalis, and urine, respectively. First, we found that urine-derived stem cells (USCs) displayed different morphologies compared with other stem cell types. USCs and placenta decidua basalis-derived mesenchymal stem cells (PDB-MSCs) had superior proliferation ability in contrast to bone marrow-derived mesenchymal stem cells (BMSCs); these cells grew to have the highest colony-forming unit (CFU) counts. In phenotypic analysis using flow cytometry, similarity among all stem cell marker expression was found, excluding CD29 and CD105. Regarding stem cell differentiation capability, USCs were observed to have better adipogenic and endothelial abilities as well as vascularization potential compared to BMSCs and PDB-MSCs. As for osteogenic and chondrogenic induction, BMSCs were superior to all three stem cell types. Future therapeutic indications and clinical applications of BMSCs, PDB-MSCs, and USCs should be based on their characteristics, such as growth kinetics and differentiation capabilities.
Collapse
Affiliation(s)
- Chengguang Wu
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich 8091, Switzerland
| | - Long Chen
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Yi-zhou Huang
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Orthopaedic Research Center, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Ornella Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia 25124, Italy
- Istituto di Anatomia Umana e Biologia Cellulare, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Roma 00168, Italy
| | - Qing Zhong
- Children's Medical Research Institute, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Xiaobin Tian
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Li Deng
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| |
Collapse
|
26
|
Huynh NPT, Zhang B, Guilak F. High-depth transcriptomic profiling reveals the temporal gene signature of human mesenchymal stem cells during chondrogenesis. FASEB J 2018; 33:358-372. [PMID: 29985644 DOI: 10.1096/fj.201800534r] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) provide an attractive cell source for cartilage repair and cell therapy; however, the underlying molecular pathways that drive chondrogenesis of these populations of adult stem cells remain poorly understood. We generated a rich data set of high-throughput RNA sequencing of human MSCs throughout chondrogenesis at 6 different time points. Our data consisted of 18 libraries with 3 individual donors as biologic replicates, with each library possessing a sequencing depth of 100 million reads. Computational analyses with differential gene expression, gene ontology, and weighted gene correlation network analysis identified dynamic changes in multiple biologic pathways and, most importantly, a chondrogenic gene subset, whose functional characterization promises to further harness the potential of MSCs for cartilage tissue engineering. Furthermore, we created a graphic user interface encyclopedia built with the goal of producing an open resource of transcriptomic regulation for additional data mining and pathway analysis of the process of MSC chondrogenesis.-Huynh, N. P. T., Zhang, B., Guilak, F. High-depth transcriptomic profiling reveals the temporal gene signature of human mesenchymal stem cells during chondrogenesis.
Collapse
Affiliation(s)
- Nguyen P T Huynh
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; and.,Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; and
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; and
| |
Collapse
|
27
|
Mohamed-Ahmed S, Fristad I, Lie SA, Suliman S, Mustafa K, Vindenes H, Idris SB. Adipose-derived and bone marrow mesenchymal stem cells: a donor-matched comparison. Stem Cell Res Ther 2018; 9:168. [PMID: 29921311 PMCID: PMC6008936 DOI: 10.1186/s13287-018-0914-1] [Citation(s) in RCA: 332] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/12/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023] Open
Abstract
Background Adipose-derived stem cells (ASCs) have been introduced as an alternative to bone marrow mesenchymal stem cells (BMSCs) for cell-based therapy. However, different studies comparing ASCs and BMSCs have shown conflicting results. In fact, harvesting ASCs and BMSCs from different individuals might influence the results, making comparison difficult. Therefore, this study aimed to characterize donor-matched ASCs and BMSCs in order to investigate proliferation, differentiation potential and possible effects of donor variation on these mesenchymal stem cells (MSCs). Methods Human bone marrow and adipose tissue samples were obtained from nine donors aged 8–14. ASCs and BMSCs were isolated and characterized based on expression of surface markers using flow cytometry. The proliferation up to 21 days was investigated. Multi-lineage differentiation was induced using osteogenic, chondrogenic and adipogenic differentiation media. Alkaline phosphatase (ALP) activity was monitored and collagen type I formation was evaluated by immunofluorescence staining. In vitro multi-potency was studied using tissue-specific stains and lineage-specific gene expression. In addition, the osteogenic lineage was evaluated at protein level. Results Isolated ASCs and BMSCs from all donors demonstrated morphologic and immunophenotypic characteristics of MSCs, with expression of MSCs markers and negative expression of hematopoietic markers. Unlike BMSCs, ASCs showed high expression of CD49d and low expression of Stro-1. In general, ASCs showed significantly higher proliferation and adipogenic capacity with more lipid vesicle formation and expression of the adipogenesis-related genes than BMSCs. In contrast, BMSCs showed significantly higher osteogenic and chondrogenic capacity compared to ASCs. BMSCs had earlier and higher ALP activity, calcium deposition, and expression of the osteogenesis- and chondrogenesis-related genes and the osteogenesis-related protein osteopontin. Proliferation and differentiation capacity of ASCs and BMSCs varied significantly among the donors. Conclusions ASCs and BMSCs showed tissue-specific differentiation abilities, but with significant variation between donors. The similarities and differences in the properties of ASCs and BMSCs should be taken into consideration when planning stem cell-based therapy.
Collapse
Affiliation(s)
- Samih Mohamed-Ahmed
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Inge Fristad
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Stein Atle Lie
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Salwa Suliman
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Hallvard Vindenes
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway.,Department for Plastic, Hand and Reconstructive Surgery, National Fire Damage Center, Bergen, Norway
| | - Shaza B Idris
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
28
|
Mesenchymal Stromal Cells: From Discovery to Manufacturing and Commercialization. Stem Cells Int 2018; 2018:4083921. [PMID: 30057622 PMCID: PMC6051015 DOI: 10.1155/2018/4083921] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/01/2018] [Accepted: 03/11/2018] [Indexed: 02/07/2023] Open
Abstract
Over the last decades, mesenchymal stromal cells (MSC) have been the focus of intense research by academia and industry due to their unique features. MSC can be easily isolated and expanded through in vitro culture by taking full advantage of their self-renewing capacity. In addition, MSC exert immunomodulatory effects and can be differentiated into various lineages, which makes them highly attractive for clinical applications in cell-based therapies. In this review, we attempt to provide a brief historical overview of MSC discovery, characterization, and the first clinical studies conducted. The current MSC manufacturing platforms are reviewed with special attention regarding the use of bioreactors for the production of GMP-compliant clinically relevant cell numbers. The first commercial MSC-based products are also addressed, as well as the remaining challenges to the widespread use of MSC-derived products.
Collapse
|
29
|
Detela G, Bain OW, Kim HW, Williams DJ, Mason C, Mathur A, Wall IB. Donor Variability in Growth Kinetics of Healthy hMSCs Using Manual Processing: Considerations for Manufacture of Cell Therapies. Biotechnol J 2018; 13. [PMID: 29334181 DOI: 10.1002/biot.201700085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 12/04/2017] [Indexed: 12/28/2022]
Abstract
Human mesenchymal stromal cells (hMSCs) are excellent candidates for cell therapy but their expansion to desired clinical quantities can be compromised by ex vivo processing, due to differences between donor material and process variation. The aim of this article is to characterize growth kinetics of healthy baseline "reference" hMSCs using typical manual processing. Bone-marrow derived hMSCs from ten donors are isolated based on plastic adherence, expanded, and analyzed for their growth kinetics until passage 4. Results indicate that hMSC density decreases with overall time in culture (p < 0.001) but no significant differences are observed between successive passages after passage 1. In addition, fold increase in cell number dropped between passage 1 and 2 for three batches, which correlated to lower performance in total fold increase and expansion potential of these batches, suggesting that proliferative ability of hMSCs can be predicted at an early stage. An indicative bounded operating window is determined between passage 1 and 3 (PDL < 10), despite the high inter-donor variability present under standardized hMSC expansion conditions used. hMSC growth profile analysis will be of benefit to cell therapy manufacturing as a tool to predict culture performance and attainment of clinically-relevant yields, therefore stratifying the patient population based on early observation.
Collapse
Affiliation(s)
- Giulia Detela
- Department of Biochemical Engineering, University College London, Gordon Street, WC1H 0AH, London, United Kingdom
| | - Owen W Bain
- Department of Biochemical Engineering, University College London, Gordon Street, WC1H 0AH, London, United Kingdom
| | - Hae-Won Kim
- Department of Nanobiomedical Science and BK21 Plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea.,College of Dentistry and Institute of Tissue Regeneration Engineering (ITREN),, Dankook University, Cheonan 31116, Republic of Korea
| | - David J Williams
- Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, United Kingdom
| | - Chris Mason
- Department of Biochemical Engineering, University College London, Gordon Street, WC1H 0AH, London, United Kingdom
| | - Anthony Mathur
- Barts Health NIHR Biomedical Research Unit, Department of Cardiology, London Chest Hospital, London E2 9JX, United Kingdom
| | - Ivan B Wall
- Department of Biochemical Engineering, University College London, Gordon Street, WC1H 0AH, London, United Kingdom.,Department of Nanobiomedical Science and BK21 Plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea.,School of Life and Health Sciences, Aston University, Aston Triangle, B4 7ET, Birmingham, United Kingdom
| |
Collapse
|
30
|
Quantitative Assessment of Optimal Bone Marrow Site for the Isolation of Porcine Mesenchymal Stem Cells. Stem Cells Int 2017; 2017:1836960. [PMID: 28539939 PMCID: PMC5429955 DOI: 10.1155/2017/1836960] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/28/2017] [Indexed: 12/13/2022] Open
Abstract
Background. One of the most plentiful sources for MSCs is the bone marrow; however, it is unknown whether MSC yield differs among different bone marrow sites. In this study, we quantified cellular yield and evaluated resident MSC population from five bone marrow sites in the porcine model. In addition, we assessed the feasibility of a commercially available platelet concentrator (Magellan® MAR01™ Arteriocyte Medical Systems, Hopkinton, MA) as a bedside stem cell concentration device. Methods. Analyses of bone marrow aspirate (BMA) and concentrated bone marrow aspirate (cBMA) included bone marrow volume, platelet and nucleated cell yield, colony-forming unit fibroblast (CFU-F) number, flow cytometry, and assessment of differentiation potential. Results. Following processing, the concentration of platelets and nucleated cells significantly increased but was not significantly different between sites. The iliac crest had significantly less bone marrow volume; however, it yielded significantly more CFUs compared to the other bone marrow sites. Culture-expanded cells from all tested sites expressed high levels of MSC surface markers and demonstrated adipogenic and osteogenic differentiation potential. Conclusions. All anatomical bone marrow sites contained MSCs, but the iliac crest was the most abundant source of MSCs. Additionally, the Magellan can function effectively as a bedside stem cell concentrator.
Collapse
|
31
|
The exciting prospects of new therapies with mesenchymal stromal cells. Cytotherapy 2016; 19:1-8. [PMID: 27769637 DOI: 10.1016/j.jcyt.2016.09.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/26/2016] [Accepted: 09/10/2016] [Indexed: 12/25/2022]
Abstract
From the outset, it was apparent that developing new therapies with mesenchymal stem/stromal cells (MSCs) was not a simple or easy task. Among the earliest experiments was administration of MSCs from normal mice to transgenic mice that developed brittle bones because they expressed a mutated gene for type 1 collagen isolated from a patient with osteogenesis imperfecta. The results prompted a clinical trial of MSCs in patients with severe osteogenesis imperfecta. Subsequent work by large numbers of scientists and clinicians has established that, with minor exceptions, MSCs do not engraft or differentiate to a large extent in vivo. Instead the cells produce beneficial effects in a large number of animal models and some clinical trials by secreting paracrine factors and extracellular vesicles in a "hit and run" scenario. The field faces a number of challenges, but the results indicate that we are on the way to effective therapies for millions of patients who suffer from devastating diseases.
Collapse
|
32
|
Characterization and Expression of Senescence Marker in Prolonged Passages of Rat Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:8487264. [PMID: 27579045 PMCID: PMC4989133 DOI: 10.1155/2016/8487264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
The present study is aimed at optimizing the in vitro culture protocol for generation of rat bone marrow- (BM-) derived mesenchymal stem cells (MSCs) and characterizing the culture-mediated cellular senescence. The initial phase of generation and characterization was conducted using the adherent cells from Sprague Dawley (SD) rat's BM via morphological analysis, growth kinetics, colony forming unit capacity, immunophenotyping, and mesodermal lineage differentiation. Mesenchymal stem cells were successfully generated and characterized as delineated by the expressions of CD90.1, CD44H, CD29, and CD71 and lack of CD11b/c and CD45 markers. Upon induction, rBM-MSCs differentiated into osteocytes and adipocytes and expressed osteocytes and adipocytes genes. However, a decline in cell growth was observed at passage 4 onwards and it was further deciphered through apoptosis, cell cycle, and senescence assays. Despite the enhanced cell viability at later passages (P4-5), the expression of senescence marker, β-galactosidase, was significantly increased at passage 5. Furthermore, the cell cycle analysis has confirmed the in vitro culture-mediated cellular senescence where cells were arrested at the G0/G1 phase of cell cycle. Although the currently optimized protocols had successfully yielded rBM-MSCs, the culture-mediated cellular senescence limits the growth of rBM-MSCs and its potential use in rat-based MSC research.
Collapse
|
33
|
Kennedy DE, Witte PL, Knight KL. Bone marrow fat and the decline of B lymphopoiesis in rabbits. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 58:30-9. [PMID: 26577994 PMCID: PMC4775299 DOI: 10.1016/j.dci.2015.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/05/2015] [Indexed: 05/03/2023]
Abstract
B lymphopoiesis is necessary to generate a diverse pool of naïve B cells that are able to respond to a broad spectrum of antigens during immune responses to pathogens and to vaccination. Rabbits have been utilized for many years to generate high affinity monoclonal and polyclonal antibodies. Specific antibodies generated in rabbits have greatly advanced scientific discoveries, but the unique qualities of rabbit B cell development have been underappreciated. Unlike in humans and mice, where B lymphopoiesis declines in mid to late life, B lymphopoiesis in rabbits arrests early in life, between 2 and 4 months of age. This review focuses on the early loss of B cell development in rabbits and the contribution of the bone marrow microenvironment to this process. We also propose directions for future research in this area, and discuss how the rabbit can be used as a model to understand the decline of B lymphopoiesis that occurs in humans late in life. Such studies will be important for developing therapeutics targeted to prevent and/or reverse declining B lymphopoiesis in the elderly, as well as boosting immunity and antibody responses after infection or vaccination.
Collapse
Affiliation(s)
- Domenick E Kennedy
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Pamela L Witte
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Katherine L Knight
- Loyola University Chicago, Department of Microbiology and Immunology, USA.
| |
Collapse
|
34
|
Ferlin KM, Kaplan DS, Fisher JP. Separation of Mesenchymal Stem Cells Through a Strategic Centrifugation Protocol. Tissue Eng Part C Methods 2016; 22:348-59. [PMID: 26797048 DOI: 10.1089/ten.tec.2015.0408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite great promise surrounding mesenchymal stem cells (MSCs), their implementation for tissue engineering strategies remains in the development phases. Many of the concerns regarding the clinical use of MSCs originate from population heterogeneity, during both isolation and differentiation. In this study, we utilize our previously developed centrifugation cell adhesion protocol for the separation of MSCs. Our findings reveal that MSCs can be isolated from whole bone marrow using a 200 g (700 pN) centrifugal force after 24 h of culture on polystyrene with cell surface marker expression equivalent to positive controls. During differentiation, a centrifugation protocol with identical force parameters could be applied 14 days into chondrogenic differentiation to isolate differentiated chondrocytes, which exhibited increased expression of chondrogenic markers compared to controls. In summary, the use of our developed centrifugation cell adhesion protocol has proven to be an effective means to separate MSC populations, decreasing the heterogeneity of subsequent cell therapy products.
Collapse
Affiliation(s)
- Kimberly M Ferlin
- 1 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | - David S Kaplan
- 2 Food and Drug Administration, Center for Devices and Radiological Health (FDA, CDRH) , Silver Spring, Maryland
| | - John P Fisher
- 1 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| |
Collapse
|
35
|
Ferlin KM, Prendergast ME, Miller ML, Kaplan DS, Fisher JP. Influence of 3D printed porous architecture on mesenchymal stem cell enrichment and differentiation. Acta Biomater 2016; 32:161-169. [PMID: 26773464 DOI: 10.1016/j.actbio.2016.01.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/25/2015] [Accepted: 01/06/2016] [Indexed: 10/22/2022]
Abstract
The interactions between cells and an underlying biomaterial are important for the promotion of cell adhesion, proliferation, and function. Mesenchymal stem cells (MSCs) have great clinical potential as they are an adult stem cell population capable of multilineage differentiation. The relationship between MSC behavior and several material properties including substrate stiffness and pore size are well investigated, but there has been little research on the influence of porous architecture in a three-dimensional scaffold with a well-controlled architecture. Here, we investigate the impact of two different three-dimensionally printed, pore geometries on the enrichment and differentiation of MSCs. 3D printed scaffolds with ordered cubic pore geometry were supportive of MSC enrichment from unprocessed bone marrow, resulting in cell surface marker expression that was comparable to typical adhesion to tissue culture polystyrene, the gold standard for MSC culture. Results also show that scaffolds fabricated with ordered cubic pores significantly increase the gene expression of MSCs undergoing adipogenesis and chondrogenesis, when compared to scaffolds with ordered cylindrical pores. However, at the protein expression level, these differences were modest. For MSCs undergoing osteogenesis, gene expression results suggest that cylindrical pores may initially increase early osteogenic marker expression, while protein level expression at later timepoints is increased for scaffolds with ordered cubic pores. Taken together, these results suggest that 3D printed scaffolds with ordered cubic pores could be a suitable culture system for single-step MSC enrichment and differentiation. STATEMENT OF SIGNIFICANCE Mesenchymal stem cells (MSCs) have great therapeutic potential, as they are capable of multilineage differentiation. MSC behavior, including lineage commitment, may be influenced by biomaterial properties including substrate stiffness and pore size. With three-dimensional (3D) printing, we can investigate these relationships in 3D culture systems. Here, we fabricated scaffolds with two different well-controlled pore geometries, and investigated the impact on MSC enrichment and differentiation. Results show that scaffolds with ordered cubic pore geometry were supportive of both MSC enrichment from unprocessed bone marrow as well as MSC differentiation, resulting in increased gene expression during adipogenesis and chondrogenesis. These results suggest that 3D printed scaffolds with ordered cubic pores could be a suitable culture system for single-step MSC enrichment and differentiation.
Collapse
|
36
|
Prockop DJ. Inflammation, fibrosis, and modulation of the process by mesenchymal stem/stromal cells. Matrix Biol 2016; 51:7-13. [PMID: 26807758 DOI: 10.1016/j.matbio.2016.01.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fibrosis and scarring are the end stage of many disease processes. In effect, the collagen fibers that initially provide a necessary strength during the repair of injured tissues are frequently synthesized in excessive amounts and become irreversible fibrotic deposits that limit regeneration of the endogenous cells of a tissue. This review will focus on the potential of mesenchymal stem/stromal cells for treatment of fibrotic diseases, with emphasis on the role of TSG-6 as a mediator of anti-inflammatory effects.
Collapse
Affiliation(s)
- Darwin J Prockop
- Institute for Regenerative Medicine, Texas A&M University, College of Medicine, Temple, TX, USA.
| |
Collapse
|
37
|
Panchalingam KM, Jung S, Rosenberg L, Behie LA. Bioprocessing strategies for the large-scale production of human mesenchymal stem cells: a review. Stem Cell Res Ther 2015; 6:225. [PMID: 26597928 PMCID: PMC4657237 DOI: 10.1186/s13287-015-0228-5] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs), also called mesenchymal stromal cells, have been of great interest in regenerative medicine applications because of not only their differentiation potential but also their ability to secrete bioactive factors that can modulate the immune system and promote tissue repair. This potential has initiated many early-phase clinical studies for the treatment of various diseases, disorders, and injuries by using either hMSCs themselves or their secreted products. Currently, hMSCs for clinical use are generated through conventional static adherent cultures in the presence of fetal bovine serum or human-sourced supplements. However, these methods suffer from variable culture conditions (i.e., ill-defined medium components and heterogeneous culture environment) and thus are not ideal procedures to meet the expected future demand of quality-assured hMSCs for human therapeutic use. Optimizing a bioprocess to generate hMSCs or their secreted products (or both) promises to improve the efficacy as well as safety of this stem cell therapy. In this review, current media and methods for hMSC culture are outlined and bioprocess development strategies discussed.
Collapse
Affiliation(s)
- Krishna M Panchalingam
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Sunghoon Jung
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Lawrence Rosenberg
- Department of Surgery, McGill University Health Centre, 845 Rue Sherbrooke Quest, Montreal, QC, H3G 1A4, Canada.,Jewish General Hospital, 3755 Chemin de la Côte-Ste-Catherine Road, Montreal, QC, H3T 1E2, Canada
| | - Leo A Behie
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
38
|
Kennedy DE, Witte PL, Knight KL. Withdrawn: Bone marrow fat and the decline of B lymphopoiesis in rabbits. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015:S0145-305X(15)30071-9. [PMID: 26550685 DOI: 10.1016/j.dci.2015.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/03/2015] [Indexed: 06/05/2023]
Affiliation(s)
- Domenick E Kennedy
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Pamela L Witte
- Loyola University Chicago, Department of Microbiology and Immunology, USA
| | - Katherine L Knight
- Loyola University Chicago, Department of Microbiology and Immunology, USA.
| |
Collapse
|
39
|
Torre ML, Lucarelli E, Guidi S, Ferrari M, Alessandri G, De Girolamo L, Pessina A, Ferrero I. Ex Vivo Expanded Mesenchymal Stromal Cell Minimal Quality Requirements for Clinical Application. Stem Cells Dev 2015; 24:677-85. [DOI: 10.1089/scd.2014.0299] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
| | - Enrico Lucarelli
- Osteoarticolar Regeneration Laboratory, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Simona Guidi
- CTP Tecnologie di Processo S.p.A. Advanced Therapy Division, Poggibonsi, Siena, Italy
| | - Maura Ferrari
- Cell Culture Centre, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna, Brescia, Italy
| | - Giulio Alessandri
- Laboratory of Cellular Neurobiology, Department of Cerebrovascular Disease, IRCCS Neurological Institute, Carlo Besta, Milan, Italy
| | - Laura De Girolamo
- Orthopedic Biotechnology Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Ivana Ferrero
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Health and Science of Turin, Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | | |
Collapse
|
40
|
Roseti L, Serra M, Bassi A. Standard operating procedure for the good manufacturing practice-compliant production of human bone marrow mesenchymal stem cells. Methods Mol Biol 2015; 1283:171-186. [PMID: 25092055 DOI: 10.1007/7651_2014_103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
According to the European Regulation (EC 1394/2007), Mesenchymal Stem Cells expanded in culture for clinical use are considered as Advanced Therapy Medicinal Products. As a consequence, they must be produced in compliance with Good Manufacturing Practice in order to ensure safety, reproducibility, and efficacy. Here, we report a Standard Operating Procedure describing the Good Manufacturing Practice-compliant production of Bone Marrow-derived Mesenchymal Stem Cells suitable for autologous implantation in humans. This procedure can be considered as a template for the development of investigational medicinal Mesenchymal Stem Cells-based product protocols to be enclosed in the dossier required for a clinical trial approval. Possible clinical applications concern local uses in the regeneration of bone tissue in nonunion fractures or in orthopedic and maxillofacial diseases characterized by a bone loss.
Collapse
Affiliation(s)
- Livia Roseti
- Dipartimento Rizzoli RIT-Research, Innovation and Technology, Cell Factory, Musculoskeletal Tissue Bank, PROMETEO, RAMSES Laboratory, Istituto Ortopedico Rizzoli, Via Pupilli 1, 40136, Bologna, Italy,
| | | | | |
Collapse
|
41
|
Naung NY, Suttapreyasri S, Kamolmatyakul S, Nuntanaranont T. Comparative study of different centrifugation protocols for a density gradient separation media in isolation of osteoprogenitors from bone marrow aspirate. J Oral Biol Craniofac Res 2014; 4:160-8. [PMID: 25737938 DOI: 10.1016/j.jobcr.2014.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/11/2014] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Human bone marrow contains osteoprogenitors capable of differentiating into osteoblasts. Density gradient centrifugation (DGC) is a commonly used method to isolate osteoprogenitors from bone marrow. Numerous studies used different dilution and centrifugation protocols, which might affect cell yields and quality. Moreover, the relative isolation efficiencies of the different separation protocols have not been investigated. This study compares the enrichment efficacy of the two different centrifugation protocols for a commonly used DGC media in isolation of osteoprogenitors. MATERIAL AND METHOD Bone marrow was aspirated from human anterior iliac crests. Osteoprogenitors are isolated with Ficoll DGC media. A centrifugal force of 400 g and 1:1 dilution was compared with the centrifugal force of 1000 g after three dilution times with a buffer. RESULTS The average numbers of isolated cells were significantly higher when using lower centrifugal force with 1:1 dilution, however, there was no detectable difference between Colony-forming unit-fibroblast (CFU-F) forming capacity, STRO-1 positivity, osteogenic differentiation or mineralization abilities between protocols. CONCLUSION Both protocols could isolate competent and functional osteoprogenitors, while a lower centrifugal force (400 g) with 1:1 dilution produced recovery of more osteoprogenitors.
Collapse
Affiliation(s)
- Noel Ye Naung
- Department of Oral Sciences, School of Dentistry, University of Otago, Dunedin, New Zealand
| | | | | | | |
Collapse
|
42
|
Castiglia S, Mareschi K, Labanca L, Lucania G, Leone M, Sanavio F, Castello L, Rustichelli D, Signorino E, Gunetti M, Bergallo M, Bordiga AM, Ferrero I, Fagioli F. Inactivated human platelet lysate with psoralen: a new perspective for mesenchymal stromal cell production in Good Manufacturing Practice conditions. Cytotherapy 2014; 16:750-63. [PMID: 24529555 PMCID: PMC7185570 DOI: 10.1016/j.jcyt.2013.12.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 12/20/2013] [Accepted: 12/22/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) are ideal candidates for regenerative and immunomodulatory therapies. The use of xenogeneic protein-free Good Manufacturing Practice-compliant growth media is a prerequisite for clinical MSC isolation and expansion. Human platelet lysate (HPL) has been efficiently implemented into MSC clinical manufacturing as a substitute for fetal bovine serum (FBS). Because the use of human-derived blood materials alleviates immunologic risks but not the transmission of blood-borne viruses, the aim of our study was to test an even safer alternative than HPL to FBS: HPL subjected to pathogen inactivation by psoralen (iHPL). METHODS Bone marrow samples were plated and expanded in α-minimum essential medium with 10% of three culture supplements: HPL, iHPL and FBS, at the same time. MSC morphology, growth and immunophenotype were analyzed at each passage. Karyotype, tumorigenicity and sterility were analyzed at the third passage. Statistical analyses were performed. RESULTS The MSCs cultivated in the three different culture conditions showed no significant differences in terms of fibroblast colony-forming unit number, immunophenotype or in their multipotent capacity. Conversely, the HPL/iHPL-MSCs were smaller, more numerous, had a higher proliferative potential and showed a higher Oct-3/4 and NANOG protein expression than did FBS-MSCs. Although HPL/iHPL-MSCs exhibit characteristics that may be attributable to a higher primitive stemness than FBS-MSCs, no tumorigenic mutations or karyotype modifications were observed. CONCLUSIONS We demonstrated that iHPL is safer than HPL and represents a good, Good Manufacturing Practice-compliant alternative to FBS for MSC clinical production that is even more advantageous in terms of cellular growth and stemness.
Collapse
Affiliation(s)
- Sara Castiglia
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Katia Mareschi
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy; Department of Public Health and Pediatrics, University of Turin, Turin, Italy.
| | - Luciana Labanca
- Blood Component Production and Validation Center, City of Science and Health of Turin, S. Anna Hospital, Turin, Italy
| | - Graziella Lucania
- Blood Component Production and Validation Center, City of Science and Health of Turin, S. Anna Hospital, Turin, Italy
| | - Marco Leone
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Fiorella Sanavio
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Laura Castello
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Deborah Rustichelli
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Elena Signorino
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | - Monica Gunetti
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| | | | - Anna Maria Bordiga
- Blood Component Production and Validation Center, City of Science and Health of Turin, S. Anna Hospital, Turin, Italy
| | - Ivana Ferrero
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy; Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Franca Fagioli
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Turin, Italy
| |
Collapse
|
43
|
Secunda R, Vennila R, Mohanashankar AM, Rajasundari M, Jeswanth S, Surendran R. Isolation, expansion and characterisation of mesenchymal stem cells from human bone marrow, adipose tissue, umbilical cord blood and matrix: a comparative study. Cytotechnology 2014; 67:793-807. [PMID: 24798808 DOI: 10.1007/s10616-014-9718-z] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 03/20/2014] [Indexed: 12/21/2022] Open
Abstract
The multipotent and immunosuppressive capacities of mesenchymal stem cells (MSCs) attract several scientists worldwide towards translational research focusing on treatment of diseases including liver failure. Though MSC's have been isolated from different sources, researchers do not concur on the best source for expansion and clinical translation. In this study, we have compared the isolation, proliferation and expansion of MSCs from umbilical cord blood (UCB), Wharton's Jelly (WJ), bone marrow (BM) and adipose tissue (AT). MSCs were isolated by density gradient separation from UCB, BM and AT and by both enzymatic and explant method for WJ. The MSCs are characterized by their ability to adhere to plastic, expression of positive (CD105, CD73, CD90, CD29, CD44) and negative (CD45, CD14, CD34) markers by flow cytometry and also by their in vitro adipogenic, osteogenic and chondrogenic differentiation. This comprehensive study clearly shows that WJ is better than UCB both in terms of rapidity, yield and ease of procedure. AT and BM are autologous sources for MSC's but the specimen collection involves cumbersome and painful procedures and an invasive approach. However being autologous, they are safe and probable candidates for therapeutic future applications.
Collapse
Affiliation(s)
- R Secunda
- Centre for Advanced Research (Stem Cells), Government Stanley Hospital, Chennai, 600 001, Tamilnadu, India
| | | | | | | | | | | |
Collapse
|
44
|
Boido M, Piras A, Valsecchi V, Spigolon G, Mareschi K, Ferrero I, Vizzini A, Temi S, Mazzini L, Fagioli F, Vercelli A. Human mesenchymal stromal cell transplantation modulates neuroinflammatory milieu in a mouse model of amyotrophic lateral sclerosis. Cytotherapy 2014; 16:1059-72. [PMID: 24794182 DOI: 10.1016/j.jcyt.2014.02.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/03/2014] [Accepted: 02/09/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs), after intraparenchymal, intrathecal and endovenous administration, have been previously tested for cell therapy in amyotrophic lateral sclerosis in the SOD1 (superoxide dismutase 1) mouse. However, every administration route has specific pros and cons. METHODS We administrated human MSCs (hMSCs) in the cisterna lumbaris, which is easily accessible and could be used in outpatient surgery, in the SOD1 G93A mouse, at the earliest onset of symptoms. Control animals received saline injections. Motor behavior was checked starting from 2 months of age until the mice were killed. Animals were killed 2 weeks after transplantation; lumbar motoneurons were stereologically counted, astrocytes and microglia were analyzed and quantified after immunohistochemistry and cytokine expression was assayed by means of real-time polymerase chain reaction. RESULTS We provide evidence that this route of administration can exert strongly positive effects. Motoneuron death and motor decay were delayed, astrogliosis was reduced and microglial activation was modulated. In addition, hMSC transplantation prevented the downregulation of the anti-inflammatory interleukin-10, as well as that of vascular endothelial growth factor observed in saline-treated transgenic mice compared with wild type, and resulted in a dramatic increase in the expression of the anti-inflammatory interleukin-13. CONCLUSIONS Our results suggest that hMSCs, when intracisternally administered, can exert their paracrine potential, influencing the inflammatory response of the host.
Collapse
Affiliation(s)
- Marina Boido
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy.
| | - Antonio Piras
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| | - Valeria Valsecchi
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| | - Giada Spigolon
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| | - Katia Mareschi
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Department of Public Health and Paediatrics, University of Torino, Torino, Italy
| | - Ivana Ferrero
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Department of Public Health and Paediatrics, University of Torino, Torino, Italy
| | - Andrea Vizzini
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| | - Santa Temi
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| | - Letizia Mazzini
- ALS Centre Department of Neurology, University of Eastern Piedmont, Novara, Italy
| | - Franca Fagioli
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Department of Public Health and Paediatrics, University of Torino, Torino, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| |
Collapse
|
45
|
Hoch AI, Leach JK. Concise review: optimizing expansion of bone marrow mesenchymal stem/stromal cells for clinical applications. Stem Cells Transl Med 2014; 3:643-52. [PMID: 24682286 DOI: 10.5966/sctm.2013-0196] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bone marrow-derived mesenchymal stem/stromal cells (MSCs) have demonstrated success in the clinical treatment of hematopoietic pathologies and cardiovascular disease and are the focus of treating other diseases of the musculoskeletal, digestive, integumentary, and nervous systems. However, during the requisite two-dimensional (2D) expansion to achieve a clinically relevant number of cells, MSCs exhibit profound degeneration in progenitor potency. Proliferation, multilineage potential, and colony-forming efficiency are fundamental progenitor properties that are abrogated by extensive monolayer culture. To harness the robust therapeutic potential of MSCs, a consistent, rapid, and minimally detrimental expansion method is necessary. Alternative expansion efforts have exhibited promise in the ability to preserve MSC progenitor potency better than the 2D paradigm by mimicking features of the native bone marrow niche. MSCs have been successfully expanded when stimulated by growth factors, under reduced oxygen tension, and in three-dimensional bioreactors. MSC therapeutic value can be optimized for clinical applications by combining system inputs to tailor culture parameters for recapitulating the niche with probes that nondestructively monitor progenitor potency. The purpose of this review is to explore how modulations in the 2D paradigm affect MSC progenitor properties and to highlight recent efforts in alternative expansion techniques.
Collapse
Affiliation(s)
- Allison I Hoch
- Department of Biomedical Engineering and Department of Orthopaedic Surgery, School of Medicine, University of California, Davis, Sacramento, California, USA
| | | |
Collapse
|
46
|
Mesenchymal stem cells for regenerative therapy: optimization of cell preparation protocols. BIOMED RESEARCH INTERNATIONAL 2014; 2014:951512. [PMID: 24511552 PMCID: PMC3912818 DOI: 10.1155/2014/951512] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/08/2013] [Indexed: 12/14/2022]
Abstract
Administration of bone marrow-derived mesenchymal stem cells (MSCs) is an innovative approach for the treatment of a range of diseases that are not curable by current therapies including heart failure. A number of clinical trials have been completed and many others are ongoing; more than 2,000 patients worldwide have been administered with culture-expanded allogeneic or autologous MSCs for the treatment of various diseases, showing feasibility and safety (and some efficacy) of this approach. However, protocols for isolation and expansion of donor MSCs vary widely between these trials, which could affect the efficacy of the therapy. It is therefore important to develop international standards of MSC production, which should be evidence-based, regulatory authority-compliant, of good medical practice grade, cost-effective, and clinically practical, so that this innovative approach becomes an established widely adopted treatment. This review article summarizes protocols to isolate and expand bone marrow-derived MSCs in 47 recent clinical trials of MSC-based therapy, which were published after 2007 onwards and provided sufficient methodological information. Identified issues and possible solutions associated with the MSC production methods, including materials and protocols for isolation and expansion, are discussed with reference to relevant experimental evidence with aim of future clinical success of MSC-based therapy.
Collapse
|
47
|
Mazzini L, Vercelli A, Ferrero I, Boido M, Cantello R, Fagioli F. Transplantation of mesenchymal stem cells in ALS. PROGRESS IN BRAIN RESEARCH 2013. [PMID: 23186722 DOI: 10.1016/b978-0-444-59544-7.00016-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating incurable, neurodegenerative disease that targets motor neurons (MNs) in the primary motor cortex, brainstem, and spinal cord, leading to muscle atrophy, paralysis, and death due to respiratory failure within 2-5 years. Currently, there is no cure for ALS. The development of a therapy that can support or restore MN function and attenuate toxicity in the spinal cord provides the most comprehensive approach for treating ALS. Mesenchymal stem cells might be suitable for cell therapy in ALS because of their immunomodulatory and protective properties. In this review, the authors discuss the major challenges to the translation of in vitro and animal studies of MSCs therapy in the clinical setting.
Collapse
Affiliation(s)
- Letizia Mazzini
- ALS Centre, Department of Neurology, Eastern Piedmont University, "Maggiore della Carità" Hospital, Novara, Italy.
| | | | | | | | | | | |
Collapse
|
48
|
Rustichelli D, Castiglia S, Gunetti M, Mareschi K, Signorino E, Muraro M, Castello L, Sanavio F, Leone M, Ferrero I, Fagioli F. Validation of analytical methods in compliance with good manufacturing practice: a practical approach. J Transl Med 2013; 11:197. [PMID: 23981284 PMCID: PMC3765465 DOI: 10.1186/1479-5876-11-197] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 08/21/2013] [Indexed: 11/10/2022] Open
Abstract
Background The quality and safety of cell therapy products must be maintained throughout their production and quality control cycle, ensuring their final use in the patient. We validated the Lymulus Amebocyte Lysate (LAL) test and immunophenotype according to International Conference on Harmonization Q2 Guidelines and the EU Pharmacopoeia, considering accuracy, precision, repeatability, linearity and range. Methods For the endotoxin test we used a kinetic chromogenic LAL test. As this is a limit test for the control of impurities, in compliance with International Conference on Harmonization Q2 Guidelines and the EU Pharmacopoeia, we evaluated the specificity and detection limit. For the immunophenotype test, an identity test, we evaluated specificity through the Fluorescence Minus One method and we repeated all experiments thrice to verify precision. The immunophenotype validation required a performance qualification of the flow cytometer using two types of standard beads which have to be used daily to check cytometer reproducibly set up. The results were compared together. Collected data were statistically analyzed calculating mean, standard deviation and coefficient of variation percentage (CV%). Results The LAL test is repeatable and specific. The spike recovery value of each sample was between 0.25 EU/ml and 1 EU/ml with a CV% < 10%. The correlation coefficient (≥ 0.980) and CV% (< 10%) of the standard curve tested in duplicate showed the test's linearity and a minimum detectable concentration value of 0.005 EU/ml. The immunophenotype method performed thrice on our cell therapy products is specific and repeatable as showed by CV% inter -experiment < 10%. Conclusions Our data demonstrated that validated analytical procedures are suitable as quality controls for the batch release of cell therapy products. Our paper could offer an important contribution for the scientific community in the field of CTPs, above all to small Cell Factories such as ours, where it is not always possible to have CFR21 compliant software.
Collapse
Affiliation(s)
- Deborah Rustichelli
- Paediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, P.zza Polonia 94, Turin 10126, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Myogenic potential of whole bone marrow mesenchymal stem cells in vitro and in vivo for usage in urinary incontinence. PLoS One 2012; 7:e45538. [PMID: 23029081 PMCID: PMC3448658 DOI: 10.1371/journal.pone.0045538] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 08/23/2012] [Indexed: 12/21/2022] Open
Abstract
Urinary incontinence, defined as the complaint of any involuntary loss of urine, is a pathological condition, which affects 30% females and 15% males over 60, often following a progressive decrease of rhabdosphincter cells due to increasing age or secondary to damage to the pelvic floor musculature, connective tissue and/or nerves. Recently, stem cell therapy has been proposed as a source for cell replacement and for trophic support to the sphincter. To develop new therapeutic strategies for urinary incontinence, we studied the interaction between mesenchymal stem cells (MSCs) and muscle cells in vitro; thereafter, aiming at a clinical usage, we analyzed the supporting role of MSCs for muscle cells in vitro and in in vivo xenotransplantation. MSCs can express markers of the myogenic cell lineages and give rise, under specific cell culture conditions, to myotube-like structures. Nevertheless, we failed to obtain mixed myotubes both in vitro and in vivo. For in vivo transplantation, we tested a new protocol to collect human MSCs from whole bone marrow, to get larger numbers of cells. MSCs, when transplanted into the pelvic muscles close to the external urethral sphincter, survived for a long time in absence of immunosuppression, and migrated into the muscle among fibers, and towards neuromuscular endplates. Moreover, they showed low levels of cycling cells, and did not infiltrate blood vessels. We never observed formation of cell masses suggestive of tumorigenesis. Those which remained close to the injection site showed an immature phenotype, whereas those in the muscle had more elongated morphologies. Therefore, MSCs are safe and can be easily transplanted without risk of side effects in the pelvic muscles. Further studies are needed to elucidate their integration into muscle fibers, and to promote their muscular transdifferentiation either before or after transplantation.
Collapse
|
50
|
Gunetti M, Castiglia S, Rustichelli D, Mareschi K, Sanavio F, Muraro M, Signorino E, Castello L, Ferrero I, Fagioli F. Validation of analytical methods in GMP: the disposable Fast Read 102® device, an alternative practical approach for cell counting. J Transl Med 2012; 10:112. [PMID: 22650233 PMCID: PMC3502295 DOI: 10.1186/1479-5876-10-112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 05/11/2012] [Indexed: 12/04/2022] Open
Abstract
Background The quality and safety of advanced therapy products must be maintained throughout their production and quality control cycle to ensure their final use in patients. We validated the cell count method according to the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use and European Pharmacopoeia, considering the tests’ accuracy, precision, repeatability, linearity and range. Methods As the cell count is a potency test, we checked accuracy, precision, and linearity, according to ICH Q2. Briefly our experimental approach was first to evaluate the accuracy of Fast Read 102® compared to the Bürker chamber. Once the accuracy of the alternative method was demonstrated, we checked the precision and linearity test only using Fast Read 102®. The data were statistically analyzed by average, standard deviation and coefficient of variation percentages inter and intra operator. Results All the tests performed met the established acceptance criteria of a coefficient of variation of less than ten percent. For the cell count, the precision reached by each operator had a coefficient of variation of less than ten percent (total cells) and under five percent (viable cells). The best range of dilution, to obtain a slope line value very similar to 1, was between 1:8 and 1:128. Conclusions Our data demonstrated that the Fast Read 102® count method is accurate, precise and ensures the linearity of the results obtained in a range of cell dilution. Under our standard method procedures, this assay may thus be considered a good quality control method for the cell count as a batch release quality control test. Moreover, the Fast Read 102® chamber is a plastic, disposable device that allows a number of samples to be counted in the same chamber. Last but not least, it overcomes the problem of chamber washing after use and so allows a cell count in a clean environment such as that in a Cell Factory. In a good manufacturing practice setting the disposable cell counting devices will allow a single use of the count chamber they can then be thrown away, thus avoiding the waste disposal of vital dye (e.g. Trypan Blue) or lysing solution (e.g. Tuerk solution).
Collapse
Affiliation(s)
- Monica Gunetti
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin 10126, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|