1
|
Kan SA, Ali A, Kao SW, Tsai BCK, Lin YM, Hsieh DJY, Kuo CH, Kuo WW, Lin SZ, Huang CY. A Novel Therapeutic Strategy for Ameliorating Hyperglycemia-Induced Liver Injury via Overexpression of the Carboxyl Terminus of HSP70-Interacting Protein in Wharton's Jelly Mesenchymal Stem Cells. Biotechnol Appl Biochem 2025:e2771. [PMID: 40350690 DOI: 10.1002/bab.2771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/13/2025] [Indexed: 05/14/2025]
Abstract
Diabetes is a widespread metabolic disorder that significantly affects modern society. The liver plays a vital role in metabolism; however, hyperglycemia can induce liver damage and disrupt its normal functions. Wharton's jelly mesenchymal stem cells (WJMSCs) engineered to express the carboxyl terminus of HSP70-interacting protein (CHIP) have demonstrated protective effects against hyperglycemia-induced damage in various organs. Nonetheless, the potential hepatoprotective effects and underlying mechanisms of these modified stem cells in diabetic livers remain unclear. Therefore, this study aimed to evaluate the efficacy of CHIP-transfected WJMSCs in mitigating hyperglycemia-induced hepatic injury in diabetic rats and to elucidate the associated protective mechanisms. Diabetic rats received tail vein injections of WJMSCs either overexpressing or silenced for CHIP. Seven weeks post-transplantation, all rats were sacrificed, and liver tissues were harvested for histological staining and Western blot analysis. The findings indicated that CHIP-overexpressing WJMSCs significantly reversed hyperglycemia-induced liver damage, reducing tissue injury, fibrosis, and glycogen deposition. These cells also alleviated hepatic inflammation and apoptosis. Moreover, they regulated oxidative stress pathways by lowering gp91-phox, Rac1, and phosphorylated PKCζ levels, while enhancing phosphorylated Nrf2 and SOD-2 expression. Additionally, the modified WJMSCs suppressed STAT3 activation and downregulated FOXO3a, suggesting a role in attenuating fibrosis and triglyceride accumulation in diabetic livers. Overall, CHIP-overexpressing WJMSCs reversed hyperglycemia-induced hepatic alterations by mitigating inflammation and oxidative stress while also modulating pathways related to fibrosis and lipid metabolism. These results highlight the therapeutic potential of CHIP-modified WJMSCs in managing diabetic liver complications and offer promising avenues for future treatment strategies.
Collapse
Affiliation(s)
- Shun-An Kan
- Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ayaz Ali
- Cardiovascular and Mitochondrial related Diseases Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Shih-Wen Kao
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial related Diseases Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yueh-Min Lin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Dennis Jine-Yuan Hsieh
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- School of Physical Education and Sports Science, Soochow University, Suzhou, China
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial related Diseases Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Tzu Chi University, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
2
|
Dupuy S, Salvador J, Morille M, Noël D, Belamie E. Control and interplay of scaffold-biomolecule interactions applied to cartilage tissue engineering. Biomater Sci 2025; 13:1871-1900. [PMID: 40052975 DOI: 10.1039/d5bm00049a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cartilage tissue engineering based on the combination of biomaterials, adult or stem cells and bioactive factors is a challenging approach for regenerative medicine with the aim of achieving the formation of a functional neotissue stable in the long term. Various 3D scaffolds have been developed to mimic the extracellular matrix environment and promote cartilage repair. In addition, bioactive factors have been extensively employed to induce and maintain the cartilage phenotype. However, the spatiotemporal control of bioactive factor release remains critical for maximizing the regenerative potential of multipotent cells, such as mesenchymal stromal cells (MSCs), and achieving efficient chondrogenesis and sustained tissue homeostasis, which are essential for the repair of hyaline cartilage. Despite advances, the effective delivery of bioactive factors is limited by challenges such as insufficient retention at the site of injury and the loss of therapeutic efficacy due to uncontrolled drug release. These limitations have prompted research on biomolecule-scaffold interactions to develop advanced delivery systems that provide sustained release and controlled bioavailability of biological factors, thereby improving therapeutic outcomes. This review focuses specifically on biomaterials (natural, hybrid and synthetic) and biomolecules (molecules, proteins, nucleic acids) of interest for cartilage engineering. Herein, we review in detail the approaches developed to maintain the biomolecules in scaffolds and control their release, based on their chemical nature and structure, through steric, non-covalent and/or covalent interactions, with a view to their application in cartilage repair.
Collapse
Affiliation(s)
- Silouane Dupuy
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Jérémy Salvador
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- EPHE, PSL Research University, 75014 Paris, France
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Marie Morille
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Emmanuel Belamie
- ICGM, University of Montpellier, CNRS, ENSCM, Montpellier, France.
- EPHE, PSL Research University, 75014 Paris, France
| |
Collapse
|
3
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
4
|
Duysens J, Graide H, Niesten A, Mouithys-Mickalad A, Deby-Dupont G, Franck T, Ceusters J, Serteyn D. Culture and Immunomodulation of Equine Muscle-Derived Mesenchymal Stromal Cells: A Comparative Study of Innovative 2D versus 3D Models Using Equine Platelet Lysate. Cells 2024; 13:1290. [PMID: 39120320 PMCID: PMC11312061 DOI: 10.3390/cells13151290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Muscle-derived mesenchymal stromal cells (mdMSCs) hold great promise in regenerative medicine due to their immunomodulatory properties, multipotent differentiation capacity and ease of collection. However, traditional in vitro expansion methods use fetal bovine serum (FBS) and have numerous limitations including ethical concerns, batch-to-batch variability, immunogenicity, xenogenic contamination and regulatory compliance issues. This study investigates the use of 10% equine platelet lysate (ePL) obtained by plasmapheresis as a substitute for FBS in the culture of mdMSCs in innovative 2D and 3D models. Using muscle microbiopsies as the primary cell source in both models showed promising results. Initial investigations indicated that small variations in heparin concentration in 2D cultures strongly influenced medium coagulation with an optimal proliferation observed at final heparin concentrations of 1.44 IU/mL. The two novel models investigated showed that expansion of mdMSCs is achievable. At the end of expansion, the 3D model revealed a higher total number of cells harvested (64.60 ± 5.32 million) compared to the 2D culture (57.20 ± 7.66 million). Trilineage differentiation assays confirmed the multipotency (osteoblasts, chondroblasts and adipocytes) of the mdMSCs generated in both models with no significant difference observed. Immunophenotyping confirmed the expression of the mesenchymal stem cell (MSC) markers CD-90 and CD-44, with low expression of CD-45 and MHCII markers for mdMSCs derived from the two models. The generated mdMSCs also had great immunomodulatory properties. Specific immunological extraction followed by enzymatic detection (SIEFED) analysis demonstrated that mdMSCs from both models inhibited myeloperoxidase (MPO) activity in a strong dose-dependent manner. Moreover, they were also able to reduce reactive oxygen species (ROS) activity, with mdMSCs from the 3D model showing significantly higher dose-dependent inhibition compared to the 2D model. These results highlighted for the first time the feasibility and efficacy of using 10% ePL for mdMSC expansion in novel 2D and 3D approaches and also that mdMSCs have strong immunomodulatory properties that can be exploited to advance the field of regenerative medicine and cell therapy instead of using FBS with all its drawbacks.
Collapse
Affiliation(s)
- J. Duysens
- Revatis SA, Rue de la Science 8, 6900 Marche-En-Famenne, Belgium; (H.G.); (J.C.); (D.S.)
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| | - H. Graide
- Revatis SA, Rue de la Science 8, 6900 Marche-En-Famenne, Belgium; (H.G.); (J.C.); (D.S.)
| | - A. Niesten
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| | - A. Mouithys-Mickalad
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| | - G. Deby-Dupont
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| | - T. Franck
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| | - J. Ceusters
- Revatis SA, Rue de la Science 8, 6900 Marche-En-Famenne, Belgium; (H.G.); (J.C.); (D.S.)
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| | - D. Serteyn
- Revatis SA, Rue de la Science 8, 6900 Marche-En-Famenne, Belgium; (H.G.); (J.C.); (D.S.)
- Centre of Oxygen Research and Development (CORD), University of Liege, 4000 Liege, Belgium; (A.N.); (A.M.-M.); (G.D.-D.); (T.F.)
| |
Collapse
|
5
|
Farzamfar S, Garcia LM, Rahmani M, Bolduc S. Navigating the Immunological Crossroads: Mesenchymal Stem/Stromal Cells as Architects of Inflammatory Harmony in Tissue-Engineered Constructs. Bioengineering (Basel) 2024; 11:494. [PMID: 38790361 PMCID: PMC11118848 DOI: 10.3390/bioengineering11050494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic landscape of tissue engineering, the integration of tissue-engineered constructs (TECs) faces a dual challenge-initiating beneficial inflammation for regeneration while avoiding the perils of prolonged immune activation. As TECs encounter the immediate reaction of the immune system upon implantation, the unique immunomodulatory properties of mesenchymal stem/stromal cells (MSCs) emerge as key navigators. Harnessing the paracrine effects of MSCs, researchers aim to craft a localized microenvironment that not only enhances TEC integration but also holds therapeutic promise for inflammatory-driven pathologies. This review unravels the latest advancements, applications, obstacles, and future prospects surrounding the strategic alliance between MSCs and TECs, shedding light on the immunological symphony that guides the course of regenerative medicine.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Luciana Melo Garcia
- Department of Medicine, Université Laval, Québec, QC G1V 0A6, Canada;
- Hematology-Oncology Service, CHU de Québec—Université Laval, Québec, QC G1V 0A6, Canada
| | - Mahya Rahmani
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
| | - Stephane Bolduc
- Centre de Recherche en Organogénèse Expérimentale/LOEX, Regenerative Medicine Division, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada; (S.F.); (M.R.)
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
6
|
Knoedler L, Dean J, Diatta F, Thompson N, Knoedler S, Rhys R, Sherwani K, Ettl T, Mayer S, Falkner F, Kilian K, Panayi AC, Iske J, Safi AF, Tullius SG, Haykal S, Pomahac B, Kauke-Navarro M. Immune modulation in transplant medicine: a comprehensive review of cell therapy applications and future directions. Front Immunol 2024; 15:1372862. [PMID: 38650942 PMCID: PMC11033354 DOI: 10.3389/fimmu.2024.1372862] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
Balancing the immune response after solid organ transplantation (SOT) and vascularized composite allotransplantation (VCA) remains an ongoing clinical challenge. While immunosuppressants can effectively reduce acute rejection rates following transplant surgery, some patients still experience recurrent acute rejection episodes, which in turn may progress to chronic rejection. Furthermore, these immunosuppressive regimens are associated with an increased risk of malignancies and metabolic disorders. Despite significant advancements in the field, these IS related side effects persist as clinical hurdles, emphasizing the need for innovative therapeutic strategies to improve transplant survival and longevity. Cellular therapy, a novel therapeutic approach, has emerged as a potential pathway to promote immune tolerance while minimizing systemic side-effects of standard IS regiments. Various cell types, including chimeric antigen receptor T cells (CAR-T), mesenchymal stromal cells (MSCs), regulatory myeloid cells (RMCs) and regulatory T cells (Tregs), offer unique immunomodulatory properties that may help achieve improved outcomes in transplant patients. This review aims to elucidate the role of cellular therapies, particularly MSCs, T cells, Tregs, RMCs, macrophages, and dendritic cells in SOT and VCA. We explore the immunological features of each cell type, their capacity for immune regulation, and the prospective advantages and obstacles linked to their application in transplant patients. An in-depth outline of the current state of the technology may help SOT and VCA providers refine their perioperative treatment strategies while laying the foundation for further trials that investigate cellular therapeutics in transplantation surgery.
Collapse
Affiliation(s)
- Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fortunay Diatta
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Noelle Thompson
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Samuel Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Richmond Rhys
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Khalil Sherwani
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Tobias Ettl
- Department of Dental, Oral and Maxillofacial Surgery, Regensburg, Germany
| | - Simon Mayer
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Florian Falkner
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Katja Kilian
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Adriana C. Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ali-Farid Safi
- Faculty of Medicine, University of Bern, Bern, Switzerland
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
| | - Stefan G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Siba Haykal
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bohdan Pomahac
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Martin Kauke-Navarro
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
7
|
Xu Z, Arkudas A, Munawar MA, Schubert DW, Fey T, Weisbach V, Mengen LM, Horch RE, Cai A. Schwann Cells Do Not Promote Myogenic Differentiation in the EPI Loop Model. Tissue Eng Part A 2024; 30:244-256. [PMID: 38063005 DOI: 10.1089/ten.tea.2023.0215] [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] [Indexed: 01/12/2024] Open
Abstract
In skeletal muscle tissue engineering, innervation and vascularization play an essential role in the establishment of functional skeletal muscle. For adequate three-dimensional assembly, biocompatible aligned nanofibers are beneficial as matrices for cell seeding. The aim of this study was to analyze the impact of Schwann cells (SC) on myoblast (Mb) and adipogenic mesenchymal stromal cell (ADSC) cocultures on poly-ɛ-caprolactone (PCL)-collagen I-nanofibers in vivo. Human Mb/ADSC cocultures, as well as Mb/ADSC/SC cocultures, were seeded onto PCL-collagen I-nanofiber scaffolds and implanted into the innervated arteriovenous loop model (EPI loop model) of immunodeficient rats for 4 weeks. Histological staining and gene expression were used to compare their capacity for vascularization, immunological response, myogenic differentiation, and innervation. After 4 weeks, both Mb/ADSC and Mb/ADSC/SC coculture systems showed similar amounts and distribution of vascularization, as well as immunological activity. Myogenic differentiation could be observed in both groups through histological staining (desmin, myosin heavy chain) and gene expression (MYOD, MYH3, ACTA1) without significant difference between groups. Expression of CHRNB and LAMB2 also implied neuromuscular junction formation. Our study suggests that the addition of SC did not significantly impact myogenesis and innervation in this model. The implanted motor nerve branch may have played a more significant role than the presence of SC.
Collapse
Affiliation(s)
- Zhou Xu
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Thyroid and Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Andreas Arkudas
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Muhammad Azeem Munawar
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Dirk W Schubert
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tobias Fey
- Department of Materials Science and Engineering, Institute of Glass and Ceramics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lilly M Mengen
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Aijia Cai
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
8
|
Sattar MA, Lingens LF, Guillaume VGJ, Goetzl R, Beier JP, Ruhl T. Association between Donor Age and Osteogenic Potential of Human Adipose Stem Cells in Bone Tissue Engineering. Curr Issues Mol Biol 2024; 46:1424-1436. [PMID: 38392210 PMCID: PMC10887920 DOI: 10.3390/cimb46020092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Adipose stem cells (ASCs) have multilineage differentiation capacity and hold great potential for regenerative medicine. Compared to bone marrow-derived mesenchymal stem cells (bmMSCs), ASCs are easier to isolate from abundant sources with significantly higher yields. It is generally accepted that bmMSCs show age-related changes in their proliferation and differentiation potentials, whereas this aspect is still controversial in the case of ASCs. In this review, we evaluated the existing data on the effect of donor age on the osteogenic potential of human ASCs. Overall, a poor agreement has been achieved because of inconsistent findings in the previous studies. Finally, we attempted to delineate the possible reasons behind the lack of agreements reported in the literature. ASCs represent a heterogeneous cell population, and the osteogenic potential of ASCs can be influenced by donor-related factors such as age, but also gender, lifestyle, and the underlying health and metabolic state of donors. Furthermore, future studies should consider experimental factors in in vitro conditions, including passaging, cryopreservation, culture conditions, variations in differentiation protocols, and readout methods.
Collapse
Affiliation(s)
- Md Abdus Sattar
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Lara F Lingens
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Vincent G J Guillaume
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Rebekka Goetzl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital, Rheinisch-Westfälische Technische Hochschule Aachen, 52074 Aachen, Germany
| |
Collapse
|
9
|
Miceli V, Zito G, Bulati M, Gallo A, Busà R, Iannolo G, Conaldi PG. Different priming strategies improve distinct therapeutic capabilities of mesenchymal stromal/stem cells: Potential implications for their clinical use. World J Stem Cells 2023; 15:400-420. [PMID: 37342218 PMCID: PMC10277962 DOI: 10.4252/wjsc.v15.i5.400] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 05/26/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) have shown significant therapeutic potential, and have therefore been extensively investigated in preclinical studies of regenerative medicine. However, while MSCs have been shown to be safe as a cellular treatment, they have usually been therapeutically ineffective in human diseases. In fact, in many clinical trials it has been shown that MSCs have moderate or poor efficacy. This inefficacy appears to be ascribable primarily to the heterogeneity of MSCs. Recently, specific priming strategies have been used to improve the therapeutic properties of MSCs. In this review, we explore the literature on the principal priming approaches used to enhance the preclinical inefficacy of MSCs. We found that different priming strategies have been used to direct the therapeutic effects of MSCs toward specific pathological processes. Particularly, while hypoxic priming can be used primarily for the treatment of acute diseases, inflammatory cytokines can be used mainly to prime MSCs in order to treat chronic immune-related disorders. The shift in approach from regeneration to inflammation implies, in MSCs, a shift in the production of functional factors that stimulate regenerative or anti-inflammatory pathways. The opportunity to fine-tune the therapeutic properties of MSCs through different priming strategies could conceivably pave the way for optimizing their therapeutic potential.
Collapse
Affiliation(s)
- Vitale Miceli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy.
| | - Giovanni Zito
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Matteo Bulati
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Alessia Gallo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Rosalia Busà
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Gioacchin Iannolo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Pier Giulio Conaldi
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| |
Collapse
|
10
|
Zhang Y, Dong Y, Wei Q, Zhuang Z, Liu Y, Yuan Q, He W, Jing Z, Li J, Li P, Zhang L, Hong Z, Zhang N, Wang H, Li W. miR-126 mitigates the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells by targeting the ERK1/2 and Bcl-2 pathways. Acta Biochim Biophys Sin (Shanghai) 2023; 55:449-459. [PMID: 36942990 PMCID: PMC10160225 DOI: 10.3724/abbs.2023016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (hBMMSCs) are a promising cell source for bone engineering owing to their high potential to differentiate into osteoblasts. The objective of the present study is to assess microRNA-126 (miR-126) and examine its effects on the osteogenic differentiation of hBMMSCs. In this study, we investigate the role of miR-126 in the progression of osteogenic differentiation (OD) as well as the apoptosis and inflammation of hBMMSCs during OD induction. OD is induced in hBMMSCs, and matrix mineralization along with other OD-associated markers are evaluated by Alizarin Red S (AR) staining and quantitative PCR (qPCR). Gain- and loss-of-function studies are performed to demonstrate the role of miR-126 in the OD of hBMMSCs. Flow cytometry and qPCR-based cytokine expression studies are performed to investigate the effect of miR-126 on the apoptosis and inflammation of hBMMSCs. The results indicate that miR-126 expression is downregulated during the OD of hBMMSCs. Gain- and loss-of function assays reveal that miR-126 upregulation inhibits the differentiation of hBMMSCs into osteoblasts, whereas the downregulation of miR-126 promotes hBMMSC differentiation, as assessed by the determination of osteogenic genes and alkaline phosphatase activity. Furthermore, the miR-126 level is positively correlated with the production of inflammatory cytokines and apoptotic cell death. Additionally, our results suggest that miR-126 negatively regulates not only B-cell lymphoma 2 (Bcl-2) expression but also the phosphorylation of extracellular signal‑regulated protein kinase (ERK) 1/2. Moreover, restoring ERK1/2 activity and upregulating Bcl-2 expression counteract the miR-126-mediated suppression of OD in hBMMSCs by promoting inflammation and apoptosis, respectively. Overall, our findings suggest a novel molecular mechanism relevant to the differentiation of hBMMSCs into osteoblasts, which can potentially facilitate bone formation by counteracting miR-126-mediated suppression of ERK1/2 activity and Bcl-2 expression.
Collapse
Affiliation(s)
- Ying Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yiping Dong
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Qiushi Wei
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
| | - Zhikun Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Youwen Liu
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Qiang Yuan
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wei He
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
| | - Zhenhao Jing
- Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jitian Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Peifeng Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Leilei Zhang
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| | - Zhinan Hong
- Institute of Orthopaedics of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510240, China
| | - Ning Zhang
- Hunan University of Chinese Medicine, Changsha 410208, China
| | - Haibin Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wuyin Li
- Medical Center of Hip, Luoyang Orthopedic-Traumatological Hospital (Orthopedics Hospital of Henan Province), Luoyang 471002, China
| |
Collapse
|
11
|
Singh S, Kumar Paswan K, Kumar A, Gupta V, Sonker M, Ashhar Khan M, Kumar A, Shreyash N. Recent Advancements in Polyurethane-based Tissue Engineering. ACS APPLIED BIO MATERIALS 2023; 6:327-348. [PMID: 36719800 DOI: 10.1021/acsabm.2c00788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In tissue engineering, polyurethane-based implants have gained significant traction because of their high compatibility and inertness. The implants therefore show fewer side effects and lasts longer. Also, the mechanical properties can be tuned and morphed into a particular shape, owing to which polyurethanes show immense versatility. In the last 3 years, scientists have devised methods to enhance the strength of and induce dynamic properties in polyurethanes, and these developments offer an immense opportunity to use them in tissue engineering. The focus of this review is on applications of polyurethane implants for biomedical application with detailed analysis of hard tissue implants like bone tissues and soft tissues like cartilage, muscles, skeletal tissues, and blood vessels. The synthetic routes for the preparation of scaffolds have been discussed to gain a better understanding of the issues that arise regarding toxicity. The focus here is also on concerns regarding the biocompatibility of the implants, given that the precursors and byproducts are poisonous.
Collapse
Affiliation(s)
- Sukriti Singh
- Department of Chemical and Biochemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Karan Kumar Paswan
- Department of Chemical and Biochemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Alok Kumar
- Department of Chemical and Biochemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Vishwas Gupta
- Department of Petroleum Engineering, Rajiv Gandhi Institute of Petroleum Technology, Mubarakpur Mukhatiya, Uttar Pradesh 229304, India
| | - Muskan Sonker
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Mohd Ashhar Khan
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Amrit Kumar
- Indian Oil Corporation Limited, Panipat Refinery, Panipat, Odisha 132140, India
| | - Nehil Shreyash
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
12
|
Qi P, Ning Z, Zhang X. Synergistic effects of 3D chitosan-based hybrid scaffolds and mesenchymal stem cells in orthopaedic tissue engineering. IET Nanobiotechnol 2023; 17:41-48. [PMID: 36708277 PMCID: PMC10116017 DOI: 10.1049/nbt2.12103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 01/29/2023] Open
Abstract
Restoration of damaged bone and cartilage tissue with biomaterial scaffolds is an area of interest in orthopaedics. Chitosan is among the low-cost biomaterials used as scaffolds with considerable biocompability to almost every human tissue. Considerable osteoconductivity, porosity, and appropriate pore size distribution have made chitosan an appropriate scaffold for loading of stem cells and a good homing place for differentiation of stem cells to bone tissue. Moreover, the similarity of chitosan to glycosaminoglycans and its potential to be used as soft gels, which could be lasting more than 1 week in mobile chondral defects, has made chitosan a polymer of interest in repairing bone and cartilage defects. Different types of scaffolds using chitosan in combination with mesenchymal stem cells (MSCs) are discussed. MSCs are widely used in regenerative medicine because of their regenerative ability, and recent line evidence reviewed demonstrated that the combination of MSCs with a combination of chitosan with different materials, including collagen type 1, hyaluronic acid, Poly(L-lacticacid)/gelatin/β-tricalcium phosphate, gamma-poly[glutamic acid] polyelectrolyte/titanium alloy, modified Poly(L-Lactide-co-Epsilon-Caprolactone), calcium phosphate, β-glycerophosphate hydrogel/calcium phosphate cement (CPC), and CPC-Chitosan-RGD, can increase the efficacy of using MSCs, and chitosan-based stem cell delivery can be a promising method in restoration of damaged bone and cartilage tissue.
Collapse
Affiliation(s)
- Ping Qi
- Department of General Practice, Linyi People's Hospital, Linyi, Shandong, China
| | - Zhaohui Ning
- Department of Traditional Chinese Medicine, Taian Central Hospital, Taian, Shandong, China
| | - Xiuju Zhang
- Department of General Practice, Linyi People's Hospital, Linyi, Shandong, China
| |
Collapse
|
13
|
Mobasher MA, Ahmed EI, Hakami NY, Germoush MO, Awad NS, Khodeer DM. The Combined Effect of Licorice Extract and Bone Marrow Mesenchymal Stem Cells on Cisplatin-Induced Hepatocellular Damage in Rats. Metabolites 2023; 13:94. [PMID: 36677019 PMCID: PMC9861302 DOI: 10.3390/metabo13010094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Drug-induced liver damage is a life-threatening disorder, and one major form of it is the hepatotoxicity induced by the drug cisplatin. In folk medicine, Licorice (Glycyrrhiza glabra (is used for detoxification and is believed to be a potent antioxidant. Currently, the magically self-renewable potential of bone marrow mesenchymal stem cells (BM-MSCs) has prompted us to explore their hepatoregenerative capability. The impact of G. glabra extract (GGE) and BM-MSCs alone and, in combination, on protecting against hepatotoxicity was tested on cisplatin-induced liver injury in rats. Hepatic damage, as revealed by liver histopathology and increased levels of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), and malondialdehyde (MDA), was elevated in rats by received 7 mg/kg of cisplatin intraperitoneally. The combination of GGE and BM-MSCs returned the enzyme levels to near the normal range. It also improved levels of liver superoxide dismutase (SOD) and glutathione (GSH) and reduced MDA levels. Additionally, it was found that when GGE and BM-MSCs were used together, they significantly downregulated caspase9 (Casp9), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), and interleukin-1β (IL-1β), which are involved in severe proinflammatory and apoptotic signaling cascades in the liver. Moreover, combining GGE and BM-MSCs led to the normal result of hepatocytes in several examined liver histological sections. Therefore, our findings suggest that GGE may have protective effects against oxidative liver damage and the promising regenerative potential of BM-MSCs.
Collapse
Affiliation(s)
- Maysa A. Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Eman Ibrahim Ahmed
- Pharmacology and Therapeutics Department, College of Medicine, Jouf University, Sakaka 72346, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum 63511, Egypt
| | - Nora Y. Hakami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
| | - Mousa O. Germoush
- Biology Department, College of Science, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nabil S Awad
- Department of Genetics, Faculty of Agriculture and Natural Resources, Aswan University, Aswan 81528, Egypt
- College of Biotechnology, Misr University for Science and Technology, Giza 12563, Egypt
| | - Dina M. Khodeer
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
14
|
Baek S, Park H, Igci FD, Lee D. Electrical Stimulation of Human Adipose-Derived Mesenchymal Stem Cells on O2 Plasma-Treated ITO Glass Promotes Osteogenic Differentiation. Int J Mol Sci 2022; 23:ijms232012490. [PMID: 36293347 PMCID: PMC9604346 DOI: 10.3390/ijms232012490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Electrical signals represent an essential form of cellular communication. For decades, electrical stimulation has been used effectively in clinical practice to enhance bone healing. However, the detailed mechanisms between electrical stimulation and bone healing are not well understood. In addition, there have been many difficulties in setting up a stable and efficient electrical stimulation system within the in vitro environment. Therefore, various conductive materials and electrical stimulation methods have been tested to establish an effective electrical stimulation system. Through these systems, many studies have been conducted on the effects of electrical stimulation on bone healing and osteogenic differentiation. However, previous studies were limited by the use of opaque conductive materials that obscure the cells; fluorescent observations and staining are known to be two of the critical methods to confirm the states of the cells. Indium tin oxide (ITO) glass is known to have excellent transparency and conductivity, but it is challenging to cultivate cells due to low cell adhesion characteristics. Therefore, we used O2 plasma treatment to increase the hydrophilicity and wettability of ITO glass. This enhanced cell affinity to the glass, providing a stable surface for the cells to attach. Then, electrical stimulation was applied with an amplitude range of 10 to 200 µA at a frequency of 10 Hz. Our results demonstrated that the osteogenic differentiation efficiency was maximized under the amplitude conditions of 10 µA and 50 µA. Accordingly, the results of our study suggest the development of an excellent platform in the field of biological research as a good tool to elucidate various mechanisms of cell bioactivity under electrical conditions.
Collapse
Affiliation(s)
- Seungho Baek
- PCL Inc., 128, Beobwon-ro, Songpa-gu, Seoul 08510, Korea
| | - Heekyung Park
- Department of Biomedical Engineering, School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-gu, Seoul 06974, Korea
| | - Fatma Dilara Igci
- Department of Biomedical Engineering, School of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-gu, Seoul 06974, Korea
| | - Donghyun Lee
- PCL Inc., 128, Beobwon-ro, Songpa-gu, Seoul 08510, Korea
- Correspondence:
| |
Collapse
|
15
|
Salvador J, Berthelot J, Bony C, Robin B, Him JLK, Noël D, Belamie E, Morille M. Size-tunable lipid vectors for controlled local delivery of siRNA from gene activated matrix. Acta Biomater 2022; 153:97-107. [PMID: 36113724 DOI: 10.1016/j.actbio.2022.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022]
Abstract
Tissue engineering aims to restore or replace different types of biological tissues through the association of cells, biologic factors and biomaterials. Currently, stem cells arise as a major cell source for many therapeutic indications, and their association with 3D scaffolds allow increasing regenerative medicine efficiency. In this context, the use of RNA interference to enhance or control stem cell differentiation into the desired phenotype appears as a promising strategy. However, achieving high transfection efficiency of cells in a 3D structure requires the use of a vector allowing for the spatiotemporally controlled release of the genetic material from these scaffolds. In this study, we report a new siRNA nanovector, called solvent exchange lipoplexe formulation (SELF), which has a tunable size, is stable over time in cell culture conditions and possess a high efficiency to transfect primary human mesenchymal stromal cells (hMSC). We associated SELFs with porous 3D collagen microspheres and demonstrated that the loading capacity and release kinetics were different depending on the size of the associated SELF. Interestingly, these different release profiles resulted in differences in the transfection kinetics of hMSCs. This original and unique type of gene activated matrix, with adaptable release kinetics, could be of interest for long-term and/or sequential transfection profiles of stem cells in 3D culture. STATEMENT OF SIGNIFICANCE: This work combines the use of human mesenchymal stromal cell (hMSC) and gene therapy for tissue engineering. Here, a gene-activated matrix was elaborated with collagen microspheres supporting hMSCs and acting as a reservoir for transfection vectors. This injectable GAM allows for the local and sustained delivery of nucleic acids, hence long-lasting transfection of the supported cells. With the original synthesis protocol presented herein, the size of the nanocarriers can be easily adapted, resulting in different siRNA release profiles from the microspheres. Most interestingly, different siRNA release profiles gave rise to different cell transfection profiles as assessed by the downregulation of a target gene. This highlights the versatility of the system and its suitability for various pathophysiological needs in regenerative medicine.
Collapse
Affiliation(s)
- Jeremy Salvador
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; IRMB, University of Montpellier, INSERM, Montpellier, France; EPHE, PSL Research University, 75014 Paris, France
| | - Jade Berthelot
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; EPHE, PSL Research University, 75014 Paris, France
| | - Claire Bony
- IRMB, University of Montpellier, INSERM, Montpellier, France
| | - Baptiste Robin
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Josephine Lai Kee Him
- Centre de Biologie Structurale (CBS), Univ Montpellier, INSERM, CNRS, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France.
| | - Emmanuel Belamie
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; EPHE, PSL Research University, 75014 Paris, France.
| | - Marie Morille
- ICGM, Univ Montpellier, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
16
|
Rhodes ADY, Duran-Mota JA, Oliva N. Current progress in bionanomaterials to modulate the epigenome. Biomater Sci 2022; 10:5081-5091. [PMID: 35880652 DOI: 10.1039/d2bm01027e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent advances in genomics during the 1990s have made it possible to study and identify genetic and epigenetic responses of cells and tissues to various drugs and environmental factors. This has accelerated the number of targets available to treat a range of diseases from cancer to wound healing disorders. Equally interesting is the understanding of how bio- and nanomaterials alter gene expression through epigenetic mechanisms, and whether they have the potential to elicit a positive therapeutic response without requiring additional biomolecule delivery. In fact, from a cell's perspective, a biomaterial is nothing more than an environmental factor, and so it has the power to epigenetically modulate gene expression of cells in contact with it. Understanding these epigenetic interactions between biomaterials and cells will open new avenues in the development of technologies that can not only provide biological signals (i.e. drugs, growth factors) necessary for therapy and regeneration, but also intimately interact with cells to promote the expression of genes of interest. This review article aims to summarise the current state-of-the-art and progress on the development of bio- and nanomaterials to modulate the epigenome.
Collapse
Affiliation(s)
- Anna D Y Rhodes
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK.
| | - Jose Antonio Duran-Mota
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK. .,Materials Engineering Group (GEMAT), IQS Barcelona, Barcelona 08017, Spain
| | - Nuria Oliva
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK.
| |
Collapse
|
17
|
Improved osteogenic differentiation by extremely low electromagnetic field exposure: possible application for bone engineering. Histochem Cell Biol 2022; 158:369-381. [PMID: 35751679 PMCID: PMC9512759 DOI: 10.1007/s00418-022-02126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 11/21/2022]
Abstract
Human periodontal ligament mesenchymal stem cells (hPDLSCs) are a promising cell type model for regenerative medicine applications due to their anti-inflammatory, immunomodulatory and non-tumorigenic potentials. Extremely low-frequency electromagnetic fields (ELF-EMF) are reported to affect biological properties such as cell proliferation and differentiation and modulate gene expression profile. In this study, we investigated the effects of an intermittent ELF-EMF exposure (6 h/day) for the standard differentiation period (28 days) and for 10 days in hPDLSCs in the presence or not of osteogenic differentiation medium (OM). We evaluated cell proliferation, de novo calcium deposition and osteogenic differentiation marker expression in sham and ELF-EMF-exposed cells. After ELF-EMF exposure, compared with sham-exposed, an increase in cell proliferation rate (p < 0.001) and de novo calcium deposition (p < 0.001) was observed after 10 days of exposure. Real-time PCR and Western blot results showed that COL1A1 and RUNX-2 gene expression and COL1A1, RUNX-2 and OPN protein expression were upregulated respectively in the cells exposed to ELF-EMF exposure along with or without OM for 10 days. Altogether, these results suggested that the promotion of osteogenic differentiation is more efficient in ELF-EMF-exposed hPDLSCs. Moreover, our analyses indicated that there is an early induction of hPDLSC differentiation after ELF-EMF application.
Collapse
|
18
|
Fayon A, Helle D, Francius G, Vincourt JB, Regnault V, Dumas D, Menu P, El Omar R. Characterization of an Innovative Biomaterial Derived From Human Wharton’s Jelly as a New Promising Coating for Tissue Engineering Applications. Front Bioeng Biotechnol 2022; 10:884069. [PMID: 35769101 PMCID: PMC9234273 DOI: 10.3389/fbioe.2022.884069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
The extracellular matrix (ECM) offers the opportunity to create a biomaterial consisting of a microenvironment with interesting biological and biophysical properties for improving and regulating cell functions. Animal-derived ECM are the most widely used as an alternative to human tissues that are of very limited availability. However, incomplete decellularization of these tissues presents a high risk of immune rejection and disease transmission. In this study, we present an innovative method to extract human ECM derived from the Wharton’s jelly (WJ-ECMaa) of umbilical cords as a novel biomaterial to be used in tissue engineering. WJ-ECMaa was very efficiently decellularized, suggesting its possible use in allogeneic conditions. Characterization of its content allowed the identification of type I collagen as its main component. Various other matrix proteins, playing an important role in cell adhesion and proliferation, were also detected. WJ-ECMaa applied as a surface coating was analyzed by fluorescent labeling and atomic force microscopy. The results revealed a particular arrangement of collagen fibers not previously described in the literature. This biomaterial also presented better cytocompatibility compared to the conventional collagen coating. Moreover, it showed adequate hemocompatibility, allowing its use as a surface with direct contact with blood. Application of WJ-ECMaa as a coating of the luminal surface of umbilical arteries for a use in vascular tissue engineering, has improved significantly the cellularization of this surface by allowing a full and homogeneous cell coverage. Taking these results together, our novel extraction method of human ECM offers a very promising biomaterial with many potential applications in tissue engineering such as the one presented direct in vascular tissue engineering. Further characterization of the composition and functionality will help explore the ways it can be used in tissue engineering applications, especially as a scaffold or a surface coating.
Collapse
Affiliation(s)
- Adrien Fayon
- Université de Lorraine, CNRS, IMoPA, Nancy, France
| | | | - Gregory Francius
- CNRS, Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l’Environnement, Université de Lorraine, Nancy, France
| | - Jean-Baptiste Vincourt
- Université de Lorraine, CNRS, IMoPA, Nancy, France
- Université de Lorraine, CNRS, INSERM, IBSLor (UMS2008/US40), Nancy, France
| | | | | | - Patrick Menu
- Université de Lorraine, CNRS, IMoPA, Nancy, France
- *Correspondence: Patrick Menu,
| | | |
Collapse
|
19
|
Ashammakhi N, GhavamiNejad A, Tutar R, Fricker A, Roy I, Chatzistavrou X, Hoque Apu E, Nguyen KL, Ahsan T, Pountos I, Caterson EJ. Highlights on Advancing Frontiers in Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:633-664. [PMID: 34210148 PMCID: PMC9242713 DOI: 10.1089/ten.teb.2021.0012] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/15/2021] [Indexed: 01/05/2023]
Abstract
The field of tissue engineering continues to advance, sometimes in exponential leaps forward, but also sometimes at a rate that does not fulfill the promise that the field imagined a few decades ago. This review is in part a catalog of success in an effort to inform the process of innovation. Tissue engineering has recruited new technologies and developed new methods for engineering tissue constructs that can be used to mitigate or model disease states for study. Key to this antecedent statement is that the scientific effort must be anchored in the needs of a disease state and be working toward a functional product in regenerative medicine. It is this focus on the wildly important ideas coupled with partnered research efforts within both academia and industry that have shown most translational potential. The field continues to thrive and among the most important recent developments are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies that warrant special attention. Developments in the aforementioned areas as well as future directions are highlighted in this article. Although several early efforts have not come to fruition, there are good examples of commercial profitability that merit continued investment in tissue engineering. Impact statement Tissue engineering led to the development of new methods for regenerative medicine and disease models. Among the most important recent developments in tissue engineering are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies. These technologies and an understanding of them will have impact on the success of tissue engineering and its translation to regenerative medicine. Continued investment in tissue engineering will yield products and therapeutics, with both commercial importance and simultaneous disease mitigation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, Michigan, USA
| | - Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Annabelle Fricker
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Ehsanul Hoque Apu
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Taby Ahsan
- RoosterBio, Inc., Frederick, Maryland, USA
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
| | - Edward J. Caterson
- Division of Plastic Surgery, Department of Surgery, Nemours/Alfred I. du Pont Hospital for Children, Wilmington, Delaware, USA
| |
Collapse
|
20
|
Salemi S, Prange JA, Baumgartner V, Mohr-Haralampieva D, Eberli D. Adult stem cell sources for skeletal and smooth muscle tissue engineering. Stem Cell Res Ther 2022; 13:156. [PMID: 35410452 PMCID: PMC8996587 DOI: 10.1186/s13287-022-02835-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/24/2022] [Indexed: 12/13/2023] Open
Abstract
INTRODUCTION Tissue engineering is an innovative field with enormous developments in recent years. These advances are not only in the understanding of how stem cells can be isolated, cultured and manipulated but also in their potential for clinical applications. Thus, tissue engineering when applied to skeletal and smooth muscle cells is an area that bears high benefit for patients with muscular diseases or damage. Most of the recent research has been focused on use of adult stem cells. These cells have the ability to rejuvenate and repair damaged tissues and can be derived from different organs and tissue sources. Recently there are several different types of adult stem cells, which have the potential to function as a cell source for tissue engineering of skeletal and smooth muscles. However, to build neo-tissues there are several challenges which have to be addressed, such as the selection of the most suitable stem cell type, isolation techniques, gaining control over its differentiation and proliferation process. CONCLUSION The usage of adult stem cells for muscle engineering applications is promising. Here, we summarize the status of research on the use of adult stem cells for cell transplantation in experimental animals and humans. In particular, the application of skeletal and smooth muscle engineering in pre-clinical and clinical trials will be discussed.
Collapse
Affiliation(s)
- Souzan Salemi
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Jenny A. Prange
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Valentin Baumgartner
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Deana Mohr-Haralampieva
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| | - Daniel Eberli
- grid.412004.30000 0004 0478 9977Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, Wagistrasse 21, 4.OG, 8952 Schlieren, Switzerland
| |
Collapse
|
21
|
Pisani S, Croce S, Mauramati S, Marmonti M, Cobianchi L, Herman I, Dorati R, Avanzini MA, Genta I, Benazzo M, Conti B. Engineered Full Thickness Electrospun Scaffold for Esophageal Tissue Regeneration: From In Vitro to In Vivo Approach. Pharmaceutics 2022; 14:pharmaceutics14020252. [PMID: 35213985 PMCID: PMC8876746 DOI: 10.3390/pharmaceutics14020252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Acquired congenital esophageal malformations, such as malignant esophageal cancer, require esophagectomy resulting in full thickness resection, which cannot be left untreated. The proposed approach is a polymeric full-thickness scaffold engineered with mesenchymal stem cells (MSCs) to promote and speed up the regeneration process, ensuring adequate support and esophageal tissue reconstruction and avoiding the use of autologous conduits. Copolymers poly-L-lactide-co-poly-ε-caprolactone (PLA-PCL) 70:30 and 85:15 ratio were chosen to prepare electrospun tubular scaffolds. Electrospinning apparatus equipped with two different types of tubular mandrels: cylindrical (∅ 10 mm) and asymmetrical (∅ 10 mm and ∅ 8 mm) were used. Tubular scaffolds underwent morphological, mechanical (uniaxial tensile stress) and biological (MTT and Dapi staining) characterization. Asymmetric tubular geometry resulted in the best properties and was selected for in vivo surgical implantation. Anesthetized pigs underwent full thickness circumferential resection of the mid-lower thoracic esophagus, followed by implantation of the asymmetric scaffold. Preliminary in vivo results demonstrated that detached stitch suture achieved better results in terms of animal welfare and scaffold integration; thus, it is to be preferred to continuous suture.
Collapse
Affiliation(s)
- Silvia Pisani
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.M.); (I.H.); (M.B.)
- Correspondence:
| | - Stefania Croce
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (L.C.)
| | - Simone Mauramati
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.M.); (I.H.); (M.B.)
| | - Marta Marmonti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (M.M.); (R.D.); (I.G.); (B.C.)
| | - Lorenzo Cobianchi
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (L.C.)
| | - Irene Herman
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.M.); (I.H.); (M.B.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (M.M.); (R.D.); (I.G.); (B.C.)
| | | | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (M.M.); (R.D.); (I.G.); (B.C.)
| | - Marco Benazzo
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.M.); (I.H.); (M.B.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (M.M.); (R.D.); (I.G.); (B.C.)
| |
Collapse
|
22
|
Kazemi T, Mohammadpour AA, Matin MM, Mahdavi-Shahri N, Dehghani H, Kazemi Riabi SH. Decellularized bovine aorta as a promising 3D elastin scaffold for vascular tissue engineering applications. Regen Med 2021; 16:1037-1050. [PMID: 34852636 DOI: 10.2217/rme-2021-0062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To evaluate the suitability of using aorta elastin scaffold, in combination with human adipose-derived mesenchymal stem cells (hAd-MSCs), as an approach for cardiovascular tissue engineering. Materials & Methods: Human adipose-derived MSCs were seeded on elastin samples of decellularized bovine aorta. The samples were cultured in vitro to investigate the inductive effects of this scaffold on the cells. The results were evaluated using histological, and immunohistochemical methods, as well as MTT assay, DNA content, reverse transcription-PCR and scanning electron microscopy. Results: Histological staining and DNA content confirmed the efficacy of decellularization procedure (82% DNA removal). MTT assay showed the construct's ability to support cell viability and proliferation. Cell differentiation was confirmed by reverse transcription-PCR and positive immunohistochemistry for alfa smooth muscle actin and von Willebrand. Conclusion: The prepared aortic elastin samples act as a potential scaffold, in combination with MSCs, for applications in cardiovascular tissue engineering. Further experiments in animal models are required to confirm this.
Collapse
Affiliation(s)
- Tahmineh Kazemi
- Department of Basic Sciences, Faculty of Veterinary Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad A Mohammadpour
- Department of Basic Sciences, Faculty of Veterinary Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics & Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell & Regenerative Medicine Research Group; Iranian Academic Center for Education, Culture & Research (ACECR) Khorasan Razavi Branch, Mashhad, Iran
| | - Nasser Mahdavi-Shahri
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics & Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam Dehghani
- Department of Basic Sciences, Faculty of Veterinary Science, Ferdowsi University of Mashhad, Mashhad, Iran; Embryonic & Stem Cell Biology & Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed H Kazemi Riabi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
23
|
Sigmarsdottir TB, McGarrity S, Yurkovich JT, Rolfsson Ó, Sigurjónsson ÓE. Analyzing Metabolic States of Adipogenic and Osteogenic Differentiation in Human Mesenchymal Stem Cells via Genome Scale Metabolic Model Reconstruction. Front Cell Dev Biol 2021; 9:642681. [PMID: 34150750 PMCID: PMC8212021 DOI: 10.3389/fcell.2021.642681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/29/2021] [Indexed: 01/14/2023] Open
Abstract
Since their initial discovery in 1976, mesenchymal stem cells (MSCs) have been gathering interest as a possible tool to further the development and enhancement of various therapeutics within regenerative medicine. However, our current understanding of both metabolic function and existing differences within the varying cell lineages (e.g., cells in either osteogenesis or adipogenesis) is severely lacking making it more difficult to fully realize the therapeutic potential of MSCs. Here, we reconstruct the MSC metabolic network to understand the activity of various metabolic pathways and compare their usage under different conditions and use these models to perform experimental design. We present three new genome-scale metabolic models (GEMs) each representing a different MSC lineage (proliferation, osteogenesis, and adipogenesis) that are biologically feasible and have distinctive cell lineage characteristics that can be used to explore metabolic function and increase our understanding of these phenotypes. We present the most distinctive differences between these lineages when it comes to enriched metabolic subsystems and propose a possible osteogenic enhancer. Taken together, we hope these mechanistic models will aid in the understanding and therapeutic potential of MSCs.
Collapse
Affiliation(s)
| | - Sarah McGarrity
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland.,Center for Systems Biology, University of Iceland, Reykjavík, Iceland
| | - James T Yurkovich
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavík, Iceland
| | - Ólafur Eysteinn Sigurjónsson
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland.,The Blood Bank, Landspitali - The National University Hospital of Iceland, Reykjavík, Iceland
| |
Collapse
|
24
|
Bari E, Scocozza F, Perteghella S, Sorlini M, Auricchio F, Torre ML, Conti M. 3D Bioprinted Scaffolds Containing Mesenchymal Stem/Stromal Lyosecretome: Next Generation Controlled Release Device for Bone Regenerative Medicine. Pharmaceutics 2021; 13:515. [PMID: 33918073 PMCID: PMC8070453 DOI: 10.3390/pharmaceutics13040515] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 12/29/2022] Open
Abstract
Three-dimensional printing of poly(ε-caprolactone) (PCL) is a consolidated scaffold manufacturing technique for bone regenerative medicine. Simultaneously, the mesenchymal stem/stromal cell (MSC) secretome is osteoinductive, promoting scaffold colonization by cells, proliferation, and differentiation. The present paper combines 3D-printed PCL scaffolds with lyosecretome, a freeze-dried formulation of MSC secretome, containing proteins and extracellular vesicles (EVs). We designed a lyosecretome 3D-printed scaffold by two loading strategies: (i) MSC secretome adsorption on 3D-printed scaffold and (ii) coprinting of PCL with an alginate-based hydrogel containing MSC secretome (at two alginate concentrations, i.e., 6% or 10% w/v). A fast release of proteins and EVs (a burst of 75% after 30 min) was observed from scaffolds obtained by absorption loading, while coprinting of PCL and hydrogel, encapsulating lyosecretome, allowed a homogeneous loading of protein and EVs and a controlled slow release. For both loading modes, protein and EV release was governed by diffusion as revealed by the kinetic release study. The secretome's diffusion is influenced by alginate, its concentration, or its cross-linking modes with protamine due to the higher steric hindrance of the polymer chains. Moreover, it is possible to further slow down protein and EV release by changing the scaffold shape from parallelepiped to cylindrical. In conclusion, it is possible to control the release kinetics of proteins and EVs by changing the composition of the alginate hydrogel, the scaffold's shape, and hydrogel cross-linking. Such scaffold prototypes for bone regenerative medicine are now available for further testing of safety and efficacy.
Collapse
Affiliation(s)
- Elia Bari
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.B.); (S.P.)
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, 27100 Pavia, Italy; (F.S.); (F.A.); (M.C.)
- P4P S.r.l., 27100 Pavia, Italy
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.B.); (S.P.)
- PharmaExceed S.r.l., 27100 Pavia, Italy;
| | - Marzio Sorlini
- PharmaExceed S.r.l., 27100 Pavia, Italy;
- SUPSI—Department of Innovative Technologies, Lugano University Centre, 6962 Viganello, Switzerland
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, 27100 Pavia, Italy; (F.S.); (F.A.); (M.C.)
- P4P S.r.l., 27100 Pavia, Italy
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (E.B.); (S.P.)
- PharmaExceed S.r.l., 27100 Pavia, Italy;
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, 27100 Pavia, Italy; (F.S.); (F.A.); (M.C.)
- P4P S.r.l., 27100 Pavia, Italy
| |
Collapse
|
25
|
Cai X, Daniels O, Cucchiarini M, Madry H. Ectopic models recapitulating morphological and functional features of articular cartilage. Ann Anat 2021; 237:151721. [PMID: 33753232 DOI: 10.1016/j.aanat.2021.151721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Articular cartilage is an extremely specialized connective tissue which covers all diarthrodial joints. Implantation of chondrogenic cells without or with additional biomaterial scaffolds in ectopic locationsin vivo generates substitutes of cartilage with structural and functional characteristics that are used in fundamental investigations while also serving as a basis for translational studies. METHODS Literature search in Pubmed. RESULTS AND DISCUSSION This narrative review summarizes the most relevant ectopic models, among which subcutaneous, intramuscular, and kidney capsule transplantation and elaborates on implanted cells and biomaterial scaffolds and on their use to recapitulate morphological and functional features of articular cartilage. Although the absence of a physiological joint environment and biomechanical stimuli is the major limiting factor, ectopic models are an established component for articular cartilage research aiming to generate a bridge between in vitro data and the clinically more relevant translational orthotopic in vivo models when their limitations are considered.
Collapse
Affiliation(s)
- Xiaoyu Cai
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - Oliver Daniels
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| |
Collapse
|
26
|
Chilmonczyk MA, Doron G, Kottke PA, Culberson AL, Leguineche K, Guldberg RE, Horwitz EM, Fedorov AG. Localized Sampling Enables Monitoring of Cell State via Inline Electrospray Ionization Mass Spectrometry. Biotechnol J 2021; 16:e2000277. [PMID: 32975016 PMCID: PMC7940552 DOI: 10.1002/biot.202000277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/09/2020] [Indexed: 12/21/2022]
Abstract
Nascent advanced therapies, including regenerative medicine and cell and gene therapies, rely on the production of cells in bioreactors that are highly heterogeneous in both space and time. Unfortunately, advanced therapies have failed to reach a wide patient population due to unreliable manufacturing processes that result in batch variability and cost prohibitive production. This can be attributed largely to a void in existing process analytical technologies (PATs) capable of characterizing the secreted critical quality attribute (CQA) biomolecules that correlate with the final product quality. The Dynamic Sampling Platform (DSP) is a PAT for cell bioreactor monitoring that can be coupled to a suite of sensor techniques to provide real-time feedback on spatial and temporal CQA content in situ. In this study, DSP is coupled with electrospray ionization mass spectrometry and direct-from-culture sampling to obtain measures of CQA content in bulk media and the cell microenvironment throughout the entire cell culture process (≈3 weeks). Post hoc analysis of this real-time data reveals that sampling from the microenvironment enables cell state monitoring (e.g., confluence, differentiation). These results demonstrate that an effective PAT should incorporate both spatial and temporal resolution to serve as an effective input for feedback control in biomanufacturing.
Collapse
Affiliation(s)
- Mason A. Chilmonczyk
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Parker H. Petit Institute for Bioengineering & Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Gilad Doron
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Parker H. Petit Institute for Bioengineering & Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Peter A. Kottke
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Parker H. Petit Institute for Bioengineering & Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Austin L. Culberson
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Parker H. Petit Institute for Bioengineering & Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| | - Kelly Leguineche
- The Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR
| | - Robert E. Guldberg
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Parker H. Petit Institute for Bioengineering & Biosciences, Georgia Institute of Technology, Atlanta, Georgia
- The Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR
| | | | - Andrei G. Fedorov
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia
- NSF Engineering Research Center (ERC) for Cell Manufacturing Technologies (CMaT), Parker H. Petit Institute for Bioengineering & Biosciences, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
27
|
Jing D, Li C, Yao K, Xie X, Wang P, Zhao H, Feng JQ, Zhao Z, Wu Y, Wang J. The vital role of Gli1 + mesenchymal stem cells in tissue development and homeostasis. J Cell Physiol 2021; 236:6077-6089. [PMID: 33533019 DOI: 10.1002/jcp.30310] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 02/05/2023]
Abstract
The hedgehog (Hh) signaling pathway plays an essential role in both tissue development and homeostasis. Glioma-associated oncogene homolog 1 (Gli1) is one of the vital transcriptional factors as well as the direct target gene in the Hh signaling pathway. The cells expressing the Gli1 gene (Gli1+ cells) have been identified as mesenchymal stem cells (MSCs) that are responsible for various tissue developments, homeostasis, and injury repair. This review outlines some recent discoveries on the crucial roles of Gli1+ MSCs in the development and homeostasis of varieties of hard and soft tissues.
Collapse
Affiliation(s)
- Dian Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chaoyuan Li
- Department of Oral Implantology, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hu Zhao
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Lewandowski RB, Stępińska M, Gietka A, Dobrzyńska M, Łapiński MP, Trafny EA. The red-light emitting diode irradiation increases proliferation of human bone marrow mesenchymal stem cells preserving their immunophenotype. Int J Radiat Biol 2021; 97:553-563. [PMID: 33471577 DOI: 10.1080/09553002.2021.1876947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 11/20/2020] [Accepted: 01/05/2021] [Indexed: 01/02/2023]
Abstract
PURPOSE For effective clinical application of human bone marrow mesenchymal stem cells (hBM-MSCs), the enhancement of their proliferation in vitro together with maintaining the expression of their crucial surface antigens and differentiation potential is necessary. The present study aimed to investigate the effect of light-emitting diode (LED) irradiation on hBM-MSCs proliferation after two, five, or nine days post-irradiation. MATERIALS AND METHODS The hBM-MSCs were exposed to the LED light at 630 nm, 4 J/cm2, and power densities of 7, 17, or 30 mW/cm2. To assess the cell proliferation rate in the sham-irradiated and irradiated samples the cells metabolic activity and DNA content were determined. The number of apoptotic and necrotic cells in the samples was also evaluated. The expression of the crucial surface antigens of the hBM-MSCs up to nine days after irradiation at 4 J/cm2 and 17 mW/cm2 was monitored with flow cytometry. Additionally, the potential of hBM-MSCs for induced differentiation was measured. RESULTS When the metabolic activity was assayed, the significant increase in the cell proliferation rate by 31 and 50% after the irradiation with 4 J/cm2 and 17 mW/cm2, respectively, was observed at day five and nine when compared to the sham-irradiated cells (p < .05). Similarly, DNA content within the irradiated hBM-MSCs increased by 31 and 41% at day five and nine after the irradiation with 4 J/cm2 and 17 mW/cm2 in comparison to the sham-irradiated cells. LED irradiation did not change the expression of the crucial surface antigens of the hBM-MSCs up to nine days after irradiation at 4 J/cm2 and 17 mW/cm2. At the same experimental conditions, the hBM-MSCs maintain in vitro their capability for multipotential differentiation into osteoblasts, adipocytes, and chondrocytes. CONCLUSION Therefore, LED irradiation at a wavelength of 630 nm, energy density 4 J/cm2, and power density 17 mW/cm2 can effectively increase the number of viable hBM-MSCs in vitro.
Collapse
Affiliation(s)
- Rafał B Lewandowski
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Małgorzata Stępińska
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Andrzej Gietka
- Optoelectronic Technologies Division, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Monika Dobrzyńska
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Mariusz P Łapiński
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| | - Elżbieta A Trafny
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Warsaw, Poland
| |
Collapse
|
29
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
30
|
Mota SM, Rogers RE, Haskell AW, McNeill EP, Kaunas R, Gregory CA, Giger ML, Maitland KC. Automated mesenchymal stem cell segmentation and machine learning-based phenotype classification using morphometric and textural analysis. J Med Imaging (Bellingham) 2021; 8:014503. [PMID: 33542945 PMCID: PMC7849042 DOI: 10.1117/1.jmi.8.1.014503] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/11/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose: Mesenchymal stem cells (MSCs) have demonstrated clinically relevant therapeutic effects for treatment of trauma and chronic diseases. The proliferative potential, immunomodulatory characteristics, and multipotentiality of MSCs in monolayer culture is reflected by their morphological phenotype. Standard techniques to evaluate culture viability are subjective, destructive, or time-consuming. We present an image analysis approach to objectively determine morphological phenotype of MSCs for prediction of culture efficacy. Approach: The algorithm was trained using phase-contrast micrographs acquired during the early and mid-logarithmic stages of MSC expansion. Cell regions are localized using edge detection, thresholding, and morphological operations, followed by cell marker identification using H-minima transform within each region to differentiate individual cells from cell clusters. Clusters are segmented using marker-controlled watershed to obtain single cells. Morphometric and textural features are extracted to classify cells based on phenotype using machine learning. Results: Algorithm performance was validated using an independent test dataset of 186 MSCs in 36 culture images. Results show 88% sensitivity and 86% precision for overall cell detection and a mean Sorensen-Dice coefficient of 0.849 ± 0.106 for segmentation per image. The algorithm exhibited an area under the curve of 0.816 (CI 95 = 0.769 to 0.886) and 0.787 (CI 95 = 0.716 to 0.851) for classifying MSCs according to their phenotype at early and mid-logarithmic expansion, respectively. Conclusions: The proposed method shows potential to segment and classify low and moderately dense MSCs based on phenotype with high accuracy and robustness. It enables quantifiable and consistent morphology-based quality assessment for various culture protocols to facilitate cytotherapy development.
Collapse
Affiliation(s)
- Sakina M. Mota
- Texas A&M University, Department of Biomedical Engineering, College Station, Texas, United States
| | - Robert E. Rogers
- Texas A&M Health Science Center, College of Medicine, Bryan, Texas, United States
| | - Andrew W. Haskell
- Texas A&M Health Science Center, College of Medicine, Bryan, Texas, United States
| | - Eoin P. McNeill
- Texas A&M Health Science Center, College of Medicine, Bryan, Texas, United States
| | - Roland Kaunas
- Texas A&M University, Department of Biomedical Engineering, College Station, Texas, United States
- Texas A&M Health Science Center, College of Medicine, Bryan, Texas, United States
| | - Carl A. Gregory
- Texas A&M Health Science Center, College of Medicine, Bryan, Texas, United States
| | - Maryellen L. Giger
- University of Chicago, Department of Radiology, Committee on Medical Physics, Chicago, Illinois, United States
| | - Kristen C. Maitland
- Texas A&M University, Department of Biomedical Engineering, College Station, Texas, United States
| |
Collapse
|
31
|
Dubey SK, Alexander A, Sivaram M, Agrawal M, Singhvi G, Sharma S, Dayaramani R. Uncovering the Diversification of Tissue Engineering on the Emergent Areas of Stem Cells, Nanotechnology and Biomaterials. Curr Stem Cell Res Ther 2020; 15:187-201. [PMID: 31957615 DOI: 10.2174/1574888x15666200103124821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/23/2022]
Abstract
Damaged or disabled tissue is life-threatening due to the lack of proper treatment. Many conventional transplantation methods like autograft, iso-graft and allograft are in existence for ages, but they are not sufficient to treat all types of tissue or organ damages. Stem cells, with their unique capabilities like self-renewal and differentiate into various cell types, can be a potential strategy for tissue regeneration. However, the challenges like reproducibility, uncontrolled propagation and differentiation, isolation of specific kinds of cell and tumorigenic nature made these stem cells away from clinical application. Today, various types of stem cells like embryonic, fetal or gestational tissue, mesenchymal and induced-pluripotent stem cells are under investigation for their clinical application. Tissue engineering helps in configuring the stem cells to develop into a desired viable tissue, to use them clinically as a substitute for the conventional method. The use of stem cell-derived Extracellular Vesicles (EVs) is being studied to replace the stem cells, which decreases the immunological complications associated with the direct administration of stem cells. Tissue engineering also investigates various biomaterials to use clinically, either to replace the bones or as a scaffold to support the growth of stemcells/ tissue. Depending upon the need, there are various biomaterials like bio-ceramics, natural and synthetic biodegradable polymers to support replacement or regeneration of tissue. Like the other fields of science, tissue engineering is also incorporating the nanotechnology to develop nano-scaffolds to provide and support the growth of stem cells with an environment mimicking the Extracellular matrix (ECM) of the desired tissue. Tissue engineering is also used in the modulation of the immune system by using patient-specific Mesenchymal Stem Cells (MSCs) and by modifying the physical features of scaffolds that may provoke the immune system. This review describes the use of various stem cells, biomaterials and the impact of nanotechnology in regenerative medicine.
Collapse
Affiliation(s)
- Sunil K Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan 333031, India
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER GUWAHATI), Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Govt. of India, NH 37, NITS Mirza, Kamrup-781125, Guwahati (Assam), India
| | - Munnangi Sivaram
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan 333031, India
| | - Mukta Agrawal
- Rungta College of Pharmaceutical Sciences and Research, Kohka- Kurud Road, Bhilai, Chhattisgarh 490024, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan 333031, India
| | - Swapnil Sharma
- Department of Pharmacy, Banastahli Vidyapith, Tonk, Rajasthan 304022, India
| | | |
Collapse
|
32
|
Frassica MT, Grunlan MA. Perspectives on Synthetic Materials to Guide Tissue Regeneration for Osteochondral Defect Repair. ACS Biomater Sci Eng 2020; 6:4324-4336. [PMID: 33455185 DOI: 10.1021/acsbiomaterials.0c00753] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regenerative engineering holds the potential to treat clinically pervasive osteochondral defects (OCDs). In a synthetic materials-guided approach, the scaffold's chemical and physical properties alone instruct cellular behavior in order to effect regeneration, referred to herein as "instructive" properties. While this alleviates the costs and off-target risks associated with exogenous growth factors, the scaffold must be potently instructive to achieve tissue growth. Moreover, toward achieving functionality, such a scaffold should also recapitulate the spatial complexity of the osteochondral tissues. Thus, in addition to the regeneration of the articular cartilage and underlying cancellous bone, the complex osteochondral interface, composed of calcified cartilage and subchondral bone, should also be restored. In this Perspective, we highlight recent synthetic-based, instructive osteochondral scaffolds that have leveraged new material chemistries as well as innovative fabrication strategies. In particular, scaffolds with spatially complex chemical and morphological features have been prepared with electrospinning, solvent-casting-particulate-leaching, freeze-drying, and additive manufacturing. While few synthetic scaffolds have advanced to clinical studies to treat OCDs, these recent efforts point to the promising use of the chemical and physical properties of synthetic materials for regeneration of osteochondral tissues.
Collapse
Affiliation(s)
- Michael T Frassica
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-2120, United States
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-2120, United States.,Department of Materials Science & Engineering, Texas A&M University, College Station, Texas 77843-3003, United States.,Department of Chemistry, Texas A&M University, College Station, Texas 77843-3255, United States
| |
Collapse
|
33
|
Quantum dots-βcyclodextrin-histidine labeled human adipose stem cells-laden chitosan hydrogel for bone tissue engineering. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 27:102217. [DOI: 10.1016/j.nano.2020.102217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022]
|
34
|
Mallis P, Alevrogianni V, Sarri P, Velentzas AD, Stavropoulos-Giokas C, Michalopoulos E. Effect of Cord Blood Platelet Gel on wound healing capacity of human Mesenchymal Stromal Cells. Transfus Apher Sci 2020; 59:102734. [PMID: 32005441 DOI: 10.1016/j.transci.2020.102734] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/02/2020] [Accepted: 01/17/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Wound healing is a dynamic process, involving the recruitment of growth factors, cytokines, chemokines and cellular populations. Recently, the Cord Blood Platelet Gel (CBPG) has been applied successfully in wound closure and tissue regeneration. Moreover, its proper combination with stem cell populations such as Mesenchymal Stromal Cells (MSCs) may positively improve the wound healing process. Based on the above data, this study aimed to the evaluation of wound healing capacity of MSCs combined with CBPG under in vitro conditions. METHODS Initially, CBPG was developed from Cord Blood Units (CBUs). The determination of wound healing ability of MSCs was performed using the scratch wound assay. In addition, the morphological features, immunophenotypical characteristics and differentiation capacity of MSCs were evaluated. RESULTS Scratch wound assay results showed, that CBPG could positively stimulate the MSCs migration. Moreover, MSCs cultured in presence of CBPG were characterized by elongated shape and improved stemness properties as it was indicated by flow cytometric analysis and differentiation process. CONCLUSION These results clearly showed the beneficial effect of CBPG in combination with MSCs in wound healing. The proper combination of CBPG with stem cells strategy may enhance the healing process in patients with skin erosions.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Greece.
| | - Vivian Alevrogianni
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Greece
| | - Phaedra Sarri
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Greece
| | - Athanassios D Velentzas
- Department of Biology, Section of Cell Biology and Biophysics, School of Science, National and Kapodistrian University of Athens, Greece
| | | | | |
Collapse
|
35
|
Gopinath VK, Soumya S, Jayakumar MN. Osteogenic and odontogenic differentiation potential of dental pulp stem cells isolated from inflamed dental pulp tissues (I-DPSCs) by two different methods. Acta Odontol Scand 2020; 78:281-289. [PMID: 31855089 DOI: 10.1080/00016357.2019.1702716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective: The objective of the present study is to isolate stem cells from inflamed dental pulp tissues (I-DPSCs) and study the characteristic such as surface markers, osteo/odontogenic differentiation potential between the outgrowth (OG) and enzymatic digestion (COL) methods.Materials and methods: I-DPSCs harvested by both methods were analysed for Mesenchymal Stem Cell marker expression by flow cytometry. The metabolic activity of the isolated cells was assessed by MTT assay. The Alkaline Phosphatase (ALP) and Alizarin red staining was done to analyse the osteogenic potential of isolated cells. The osteo/odontogenic differentiation was done by checking the expression of Dentine Matrix Protein 1 (DMP1), Dentine Sialophosphoprotein (DSPP), ALP and Bone Gamma-Carboxyglutamate Protein (BGLAP) by Real time PCR.Results: The isolated cells were positive for MSC markers such as CD-90, CD-105 and CD-73 and negative for CD-14, CD-45 and STRO-1. MTT assay indicated that the I-DPSCs from OG method showed higher metabolic activity than cells from COL. However, the osteo/odontogenic differentiation was in favour of cells isolated by COL method.Conclusion: Although the cell metabolic rate was more in OG, the osteo/odontogenic differentiation was higher in COL, suggesting that the isolation method and culture conditions do affect the differentiation capacity of isolated cells.
Collapse
Affiliation(s)
- Vellore Kannan Gopinath
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, UAE
| | - S. Soumya
- The Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | | |
Collapse
|
36
|
New Insights on Mechanical Stimulation of Mesenchymal Stem Cells for Cartilage Regeneration. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10082927] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Successful tissue regeneration therapies require further understanding of the environment in which the cells are destined to be set. The aim is to structure approaches that aspire to a holistic view of biological systems and to scientific reliability. Mesenchymal stem cells represent a valuable resource for cartilage tissue engineering, due to their chondrogenic differentiation capacity. Promoting chondrogenesis, not only by growth factors but also by exogenous enhancers such as biomechanics, represents a technical enhancement. Tribological evaluation of the articular joint has demonstrated how mechanical stimuli play a pivotal role in cartilage repair and participate in the homeostasis of this tissue. Loading stresses, physiologically experienced by chondrocytes, can upregulate the production of proteins like glycosaminoglycan or collagen, fundamental for articular wellness, as well as promote and preserve cell viability. Therefore, there is a rising interest in the development of bioreactor devices that impose compression, shear stress, and hydrostatic pressure on stem cells. This strategy aims to mimic chondrogenesis and overcome complications like hypertrophic phenotyping and inappropriate mechanical features. This review will analyze the dynamics inside the joint, the natural stimuli experienced by the chondrocytes, and how the biomechanical stimuli can be applied to a stem cell culture in order to induce chondrogenesis.
Collapse
|
37
|
Sigmarsdóttir Þ, McGarrity S, Rolfsson Ó, Yurkovich JT, Sigurjónsson ÓE. Current Status and Future Prospects of Genome-Scale Metabolic Modeling to Optimize the Use of Mesenchymal Stem Cells in Regenerative Medicine. Front Bioeng Biotechnol 2020; 8:239. [PMID: 32296688 PMCID: PMC7136564 DOI: 10.3389/fbioe.2020.00239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells are a promising source for externally grown tissue replacements and patient-specific immunomodulatory treatments. This promise has not yet been fulfilled in part due to production scaling issues and the need to maintain the correct phenotype after re-implantation. One aspect of extracorporeal growth that may be manipulated to optimize cell growth and differentiation is metabolism. The metabolism of MSCs changes during and in response to differentiation and immunomodulatory changes. MSC metabolism may be linked to functional differences but how this occurs and influences MSC function remains unclear. Understanding how MSC metabolism relates to cell function is however important as metabolite availability and environmental circumstances in the body may affect the success of implantation. Genome-scale constraint based metabolic modeling can be used as a tool to fill gaps in knowledge of MSC metabolism, acting as a framework to integrate and understand various data types (e.g., genomic, transcriptomic and metabolomic). These approaches have long been used to optimize the growth and productivity of bacterial production systems and are being increasingly used to provide insights into human health research. Production of tissue for implantation using MSCs requires both optimized production of cell mass and the understanding of the patient and phenotype specific metabolic situation. This review considers the current knowledge of MSC metabolism and how it may be optimized along with the current and future uses of genome scale constraint based metabolic modeling to further this aim.
Collapse
Affiliation(s)
- Þóra Sigmarsdóttir
- The Blood Bank, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| | - Sarah McGarrity
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Óttar Rolfsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Ólafur E. Sigurjónsson
- The Blood Bank, Landspitali – The National University Hospital of Iceland, Reykjavik, Iceland
- School of Science and Engineering, Reykjavik University, Reykjavik, Iceland
| |
Collapse
|
38
|
Ding Y, Johnson R, Sharma S, Ding X, Bryant SJ, Tan W. Tethering transforming growth factor β1 to soft hydrogels guides vascular smooth muscle commitment from human mesenchymal stem cells. Acta Biomater 2020; 105:68-77. [PMID: 31982589 DOI: 10.1016/j.actbio.2020.01.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) hold great promise for vascular smooth muscle regeneration. However, most studies have mainly relied on extended supplementation of sophisticated biochemical regimen to drive MSC differentiation towards vascular smooth muscle cells (vSMCs). Herein we demonstrate a concomitant method that exploits the advantages of biomimetic matrix stiffness and tethered transforming growth factor β1 (TGF-β1) to guide vSMC commitment from human MSCs. Our designed poly(ethylene glycol) hydrogels, presenting a biomimetic stiffness and tethered TGF-β1, provide an instructive environment to potently upregulate smooth muscle marker expression in vitro and in vivo. Importantly, it significantly enhances the functional contractility of vSMCs derived from MSCs within 3 days. Interestingly, compared to non-tethered one, tethered TGF-β1 enhanced the potency of vSMC commitment on hydrogels. We provide compelling evidence that combining stiffness and tethered TGF-β1 on poly(ethylene glycol) hydrogels can be a promising approach to drastically enhance maturation and function of vSMCs from stem cell differentiation in vitro and in vivo. STATEMENT OF SIGNIFICANCE: A fast, reliable and safe regeneration of vascular smooth muscle cells (vSMCs) from stem cell differentiation is promising for vascular tissue engineering and regenerative medicine applications, but remains challenging. Herein, a photo-click hydrogel platform is devised to recapitulate the stiffness of vascular tissue and appropriate presentation of transforming growth factor β1 (TGF-β1) to guide vSMC commitment from mesenchymal stem cells (MSCs). We demonstrate that such concomitant method drastically enhanced regeneration of mature, functional vSMCs from MSCs in vitro and in vivo within only a 3-days span. This work is not only of fundamental scientific importance, revealing how physiochemical factors and the manner of their presentation direct stem cell differentiation, but also attacks the long-standing difficulty in regenerating highly functional vSMCs within a short period.
Collapse
|
39
|
Lindsay SL, McCanney GA, Willison AG, Barnett SC. Multi-target approaches to CNS repair: olfactory mucosa-derived cells and heparan sulfates. Nat Rev Neurol 2020; 16:229-240. [PMID: 32099190 DOI: 10.1038/s41582-020-0311-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George A McCanney
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alice G Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
40
|
Ayoub S, Berbéri A, Fayyad-Kazan M. An update on human periapical cyst-mesenchymal stem cells and their potential applications in regenerative medicine. Mol Biol Rep 2020; 47:2381-2389. [PMID: 32026284 DOI: 10.1007/s11033-020-05298-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/31/2020] [Indexed: 12/16/2022]
Abstract
The broad clinical applications of Mesenchymal Stem Cells (MSCs) in the regenerative medicine field is attributed to their ability to self-renew and differentiate into multiple cellular lineages. Nowadays, MSCs can be derived from a variety of adult and fetal tissues including bone marrow, adipose tissue, umbilical cord and placenta. The difficulties associated with the isolation of MSCs from certain tissues such as bone marrow promoted the search for alternative tissues which are easily accessible. Oral derived MSCs include dental pulp stem cells (DPSCs), dental follicle progenitor cells (DFPC), and periodontal ligament stem cells (PDLSC). Being abundant and easily accessible, oral derived MSCs represent an interesting alternative MSC type to be employed in regenerative medicine. Human periapical cyst-mesenchymal stem cells (hPCy-MSCs) correspond to a newly discovered and characterized MSC subtype. Interestingly, hPCy-MSCs are collected from periapical cysts, which are a biological waste, without any influence on the other healthy tissues in oral cavity. hPCy-MSCs exhibit cell surface marker profile similar to that of other oral derived MSCs, show high proliferative potency, and possess the potential to differentiate into different cell types such as osteoblasts, adipocytes and neurons-like cells. hPCy-MSCs, therefore, represent a novel promising MSCs type to be applied in regenerative medicine domain. In this review, we will compare the different types of dental derived MSCs, we will highlight the isolation technique, the characteristics, and the therapeutic potential of hPCy-MSCs.
Collapse
Affiliation(s)
- Sara Ayoub
- Department of Prosthodontics, Faculty of Dental Medicine, Lebanese University, Beirut, Lebanon
| | - Antoine Berbéri
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Lebanese University, Beirut, Lebanon
| | - Mohammad Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon. .,Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
41
|
Ni L, Yu J, Gui X, Lu Z, Wang X, Guo H, Zhou Y. Overexpression of RPN2 promotes osteogenic differentiation of hBMSCs through the JAK/STAT3 pathway. FEBS Open Bio 2019; 10:158-167. [PMID: 31743606 PMCID: PMC6943221 DOI: 10.1002/2211-5463.12766] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 10/28/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is characterized by decreased bone mass and degenerating bone structure, which cause severe bone fragility and increase the risk for fractures. Human bone mesenchymal stem cells (hBMSCs) differentiate into osteoblasts through osteogenesis, and disturbances in the balance between bone generation and degeneration underlie the pathogenesis of senile osteoporosis. The highly conserved glycoprotein Ribophorin II (RPN2) is involved in multiple biological reactions, but the role of RPN2 in the osteogenic differentiation of hBMSCs and their molecular etiology is incompletely understood. Here, we show that RPN2 expression is up‐regulated in hBMSCs during osteogenic differentiation. In vitro assays revealed that silencing of RPN2 inhibited hBMSC differentiation into osteoblasts. Moreover, RPN2 overexpression enhanced the expression of linked genes and resulted in high alkaline phosphatase activity. Our results suggest that RPN2 targets Janus kinase 1 (JAK1), and RPN2 overexpression was observed to induce JAK1 ubiquitination. Depletion of JAK1 facilitated osteogenic differentiation of RPN2‐silenced hBMSCs. Moreover, western blot analysis revealed that RPN2 silencing suppressed the stimulation and nuclear translocation of the downstream signal transducer and activator of transcription 3 sensor; this could be reversed via RPN2 overexpression. This research sheds light on an innovative molecular mechanism that is associated with hBMSC differentiation into osteoblasts and may facilitate bone anabolism through RPN2.
Collapse
Affiliation(s)
- Ling Ni
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| | - Jianhua Yu
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| | - Xueqiong Gui
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| | - Zhonghua Lu
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| | - Xiwen Wang
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| | - Hongyan Guo
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| | - Ying Zhou
- Department of Geriatrics, Yangpu District Shidong Hospital, Shanghai, China
| |
Collapse
|
42
|
Xiao F, Zhou Y, Liu Y, Xie M, Guo G. Inhibitory Effect of Sirtuin6 (SIRT6) on Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. Med Sci Monit 2019; 25:8412-8421. [PMID: 31701920 PMCID: PMC6858786 DOI: 10.12659/msm.917118] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The imbalance between bone resorption and formation is the basic mechanism underlying osteoporosis in the elderly. Osteogenesis is the differentiation of human mesenchymal stem cells (hMSCs) into osteoblasts. Sirtuin6 (SIRT6) regulates various biological functions, including differentiation. Transient receptor potential cation channel subfamily V member 1 (TRPV1) is a non-selective cation channel that can be activated by physical and chemical stimulation. However, experimental data supporting the role of SIRT6 in osteogenic differentiation (OD) of hMSCs are lacking. MATERIAL AND METHODS Differentiation of hMSCs was induced. The expressions of SIRT6, TRPV1, and CGRP were detected by Q-PCR, Western blot, and ELISA, respectively. SIRT6 was overexpressed in hMSCs by transfection. ALP activity and Alizarin Red staining were utilized to detect the effect of SIRT6 on hMSC OD. Then, capsaicin and capsazepine, the TRPV1 agonist and antagonist, respectively, were administrated to assess the role of TRPV1. RESULTS SIRT6 expression was downregulated during hMSC differentiation. SIRT6 overexpression was accompanied by reduced expression of specific genes and alkaline phosphatase (ALP) activity in osteoblasts. Furthermore, TRPV1 channel was also reduced by SIRT6 overexpression via ubiquitinating TRPV1. Capsaicin was utilized in SIRT6-overexpressed cells. Capsaicin therapy counteracted the effect of SIRT6 overexpression on OD, and markedly decreased OD. CONCLUSIONS The SIRT6-TRPV1-CGRP signal axis is the key to regulating OD in hMSCs, which could be a potential therapeutic target for osteoporosis and bone loss-related diseases.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Yun Zhou
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Yongfu Liu
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Mian Xie
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Guancheng Guo
- Department of Emergency Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
43
|
Lee DJ, Kwon J, Current L, Yoon K, Zalal R, Hu X, Xue P, Ko CC. Osteogenic potential of mesenchymal stem cells from rat mandible to regenerate critical sized calvarial defect. J Tissue Eng 2019; 10:2041731419830427. [PMID: 30886687 PMCID: PMC6415471 DOI: 10.1177/2041731419830427] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/19/2019] [Indexed: 12/19/2022] Open
Abstract
Although bone marrow–derived mesenchymal stem cells (MSCs) have been extensively explored in bone tissue engineering, only few studies using mesenchymal stem cells from mandible (M-MSCs) have been reported. However, mesenchymal stem cells from mandible have the potential to be as effective as femur-derived mesenchymal stem cells (F-MSCs) in regenerating bone, especially in the orofacial regions, which share embryonic origin, proximity, and accessibility. M-MSCs were isolated and characterized using mesenchymal stem cell–specific markers, colony forming assay, and multi-potential differentiation. In vitro osteogenic potential, including proliferation, osteogenic gene expression, alkaline phosphatase activity, and mineralization, was examined and compared. Furthermore, in vivo bone formations of F-MSCs and M-MSCs in rat critical sized defect were evaluated using microCT and histology. M-MSCs from rat could be successfully isolated and expanded while preserving their MSC’s characteristics. M-MSCs demonstrated a comparable proliferation and mineralization potentials and in vivo bone formation as F-MSCs. M-MSCs is a promising cell source candidate for craniofacial bone tissue engineering.
Collapse
Affiliation(s)
- Dong Joon Lee
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jane Kwon
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luke Current
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kun Yoon
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rahim Zalal
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiangxiang Hu
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peng Xue
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ching-Chang Ko
- Oral and Craniofacial Health Sciences Research and North Carolina Oral Health Institute, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Orthodontics, School of Dentistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
44
|
Cai Q, Zheng P, Ma F, Zhang H, Li Z, Fu Q, Han C, Sun Y. MicroRNA-224 enhances the osteoblastic differentiation of hMSCs via Rac1. Cell Biochem Funct 2019; 37:62-71. [PMID: 30773655 DOI: 10.1002/cbf.3373] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/07/2018] [Accepted: 12/10/2018] [Indexed: 12/30/2022]
Abstract
Osteogenesis is the differentiation of mesenchymal stem cells (MSCs) into osteoblasts. MicroRNAs (miRNAs) are short noncoding RNAs that target specific genes to mediate translational activities. In this study, we investigated how miR-224 regulates the osteoblastic differentiation of human MSCs (hMSCs) as well as the underlying mechanism. The results revealed the upregulation of miR-224 during hMSC differentiation. In vitro experiments showed that the downregulation of miR-224 suppressed the differentiation of hMSCs into osteoblasts. However, upregulation of miR-224 was concomitant with increased expression of relevant genes and augmented activity of alkaline phosphatase. Furthermore, the results indicated that Rac1 acted as the bona fide target of miR-224 and that Rac1 depletion promoted osteogenic differentiation in miR-224-silenced hMSCs. In addition, we found that both JAK/STAT3 and Wnt/β-catenin pathways were repressed by Rac1 depletion using quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting, and immunofluorescence. Our data indicate a novel molecular mechanism in relation to hMSCs differentiation into osteoblasts, which may facilitate bone anabolism via miR-224. SIGNIFICANCE OF THE STUDY: In this study, we mainly explored the effects of miR-224 on hMSCs differentiation into osteoblasts. We find that induced miR-224 expression in hMSCs is considered closely associated with specific osteogenesis-related genes, alkaline phosphatase activity, and matrix mineralization, indicating that miR-224 may serve as a promising biomarker for osteogenic differentiation. Our data indicate a novel molecular mechanism in relation to hMSCs differentiation into osteoblasts, which may facilitate bone anabolism via miR-224.
Collapse
Affiliation(s)
- Qing Cai
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China.,Jinlin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Peng Zheng
- Department of Endodontics, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Fuzhe Ma
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Huiyan Zhang
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Zuntai Li
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Qiyue Fu
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Chunyu Han
- Department of Dental Implantology, School and Hospital of Stomotology, Jinlin University, Changchun, China
| | - Yingying Sun
- Department of Stomatology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Dynamic in vitro models for tumor tissue engineering. Cancer Lett 2019; 449:178-185. [PMID: 30763717 DOI: 10.1016/j.canlet.2019.01.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 01/04/2023]
Abstract
Cancer research uses in vitro studies for controllable analysis of tumor behavior and preclinical testing of therapeutics. Shortcomings of basic cell culture systems in recreating in vivo interactions have driven the development of more efficient and biomimetic in vitro environments for cancer research. Assimilation of certain developments in tissue engineering will accelerate and improve the design of these environments. With the continual improvement of the tumor engineering field, the next step is towards macroscopic systems such as scaffold-supported, flow-perfused macroscale tumor bioreactors. Surface modifications of synthetic scaffolds allow for targeted cell adhesion and improved ECM development. Flow perfusion has emerged as means to expose cancerous tissues to critical biomechanical forces for tumor progression while simultaneously improving nutrient and waste transport. Macroscale perfusable systems allow for non-destructive real-time monitoring using biosensors capable of improving understanding of in vitro tumor development at reduced cost and waste. The combination of macroscale perfusable systems, surface-modified synthetic scaffolds, and non-destructive real-time monitoring will provide advanced platforms for in vitro modeling of tumor development, with broad applications in basic tumor research and preclinical drug development.
Collapse
|
46
|
Deliormanlı AM, Türk M, Atmaca H. Response of mouse bone marrow mesenchymal stem cells to graphene-containing grid-like bioactive glass scaffolds produced by robocasting. J Biomater Appl 2018; 33:488-500. [PMID: 30249149 DOI: 10.1177/0885328218799610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In the study, three-dimensional, grid-like silicate-based bioactive glass scaffolds were manufactured using a robotic deposition technique. Inks were prepared by mixing 13-93 bioactive glass particles in Pluronic® F-127 solution. After deposition, scaffolds were dried at room temperature and sintered at 690°C for 1 h. The surface of the sintered scaffolds was coated with graphene nanopowder (1, 3, 5, 10 wt%) containing poly(ε-caprolactone) solution. The in vitro mineralization ability of the prepared composite scaffolds was investigated in simulated body fluid. The surface of the simulated body fluid-treated scaffolds was analyzed using scanning electron microscopy to investigate the hydroxyapatite formation. Mechanical properties were tested under compression. Results revealed that graphene coating has no detrimental effect on the hydroxyapatite forming ability of the prepared glass scaffolds. On the other hand, it decreased the compression strength of the scaffolds at high graphene concentrations. The prepared grid-like bioactive glass-based composite scaffolds did not show toxic response to bone marrow mesenchymal stem cells. It was shown that stem cells seeded onto the scaffolds attached and proliferated well on the surface. Cells seeded on the scaffolds surface also demonstrated osteogenic differentiation under in vitro conditions in the absence of transforming growth factors.
Collapse
Affiliation(s)
- Aylin M Deliormanlı
- 1 Manisa Celal Bayar University, Faculty of Engineering, Department of Metallurgical and Materials Engineering, Yunusemre, Manisa, Turkey
| | - Mert Türk
- 1 Manisa Celal Bayar University, Faculty of Engineering, Department of Metallurgical and Materials Engineering, Yunusemre, Manisa, Turkey
| | - Harika Atmaca
- 2 Manisa Celal Bayar University, Faculty of Science and Literature, Department of Biology, Yunusemre, Manisa, Turkey
| |
Collapse
|
47
|
Goetzke R, Sechi A, De Laporte L, Neuss S, Wagner W. Why the impact of mechanical stimuli on stem cells remains a challenge. Cell Mol Life Sci 2018; 75:3297-3312. [PMID: 29728714 PMCID: PMC11105618 DOI: 10.1007/s00018-018-2830-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 02/08/2023]
Abstract
Mechanical stimulation affects growth and differentiation of stem cells. This may be used to guide lineage-specific cell fate decisions and therefore opens fascinating opportunities for stem cell biology and regenerative medicine. Several studies demonstrated functional and molecular effects of mechanical stimulation but on first sight these results often appear to be inconsistent. Comparison of such studies is hampered by a multitude of relevant parameters that act in concert. There are notorious differences between species, cell types, and culture conditions. Furthermore, the utilized culture substrates have complex features, such as surface chemistry, elasticity, and topography. Cell culture substrates can vary from simple, flat materials to complex 3D scaffolds. Last but not least, mechanical forces can be applied with different frequency, amplitude, and strength. It is therefore a prerequisite to take all these parameters into consideration when ascribing their specific functional relevance-and to only modulate one parameter at the time if the relevance of this parameter is addressed. Such research questions can only be investigated by interdisciplinary cooperation. In this review, we focus particularly on mesenchymal stem cells and pluripotent stem cells to discuss relevant parameters that contribute to the kaleidoscope of mechanical stimulation of stem cells.
Collapse
Affiliation(s)
- Roman Goetzke
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
- Institute for Biomedical Engineering - Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Antonio Sechi
- Institute for Biomedical Engineering - Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Laura De Laporte
- DWI - Leibniz-Institute for Interactive Materials, 52074, Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, Biointerface Group, RWTH Aachen University Medical School, 52074, Aachen, Germany.
- Institute of Pathology, RWTH Aachen University Medical School, Aachen, Germany.
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany.
- Institute for Biomedical Engineering - Cell Biology, RWTH Aachen University Medical School, Aachen, Germany.
- Helmholtz Institute for Biomedical Engineering, Biointerface Group, RWTH Aachen University Medical School, 52074, Aachen, Germany.
| |
Collapse
|
48
|
Shalumon KT, Kuo CY, Wong CB, Chien YM, Chen HA, Chen JP. Gelatin/Nanohyroxyapatite Cryogel Embedded Poly(lactic- co-glycolic Acid)/Nanohydroxyapatite Microsphere Hybrid Scaffolds for Simultaneous Bone Regeneration and Load-Bearing. Polymers (Basel) 2018; 10:620. [PMID: 30966654 PMCID: PMC6403993 DOI: 10.3390/polym10060620] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 11/18/2022] Open
Abstract
It is desirable to combine load-bearing and bone regeneration capabilities in a single bone tissue engineering scaffold. For this purpose, we developed a high strength hybrid scaffold using a sintered poly(lactic-co-glycolic acid) (PLGA)/nanohydroxyapatite (nHAP) microsphere cavity fitted with gelatin/nHAP cryogel disks in the center. Osteo-conductive/osteo-inductive nHAP was incorporated in 250⁻500 μm PLGA microspheres at 40% (w/w) as the base matrix for the high strength cavity-shaped microsphere scaffold, while 20% (w/w) nHAP was incorporated into gelatin cryogels as an embedded core for bone regeneration purposes. The physico-chemical properties of the microsphere, cryogel, and hybrid scaffolds were characterized in detail. The ultimate stress and Young's modulus of the hybrid scaffold showed 25- and 21-fold increases from the cryogel scaffold. In vitro studies using rabbit bone marrow-derived stem cells (rBMSCs) in cryogel and hybrid scaffolds through DNA content, alkaline phosphatase activity, and mineral deposition by SEM/EDS, showed the prominence of both scaffolds in cell proliferation and osteogenic differentiation of rBMSCs in a normal medium. Calcium contents analysis, immunofluorescent staining of collagen I (COL I), and osteocalcin (OCN) and relative mRNA expression of COL I, OCN and osteopontin (OPN) confirmed in vitro differentiation of rBMSCs in the hybrid scaffold toward the bone lineage. From compression testing, the cell/hybrid scaffold construct showed a 1.93 times increase of Young's modulus from day 14 to day 28, due to mineral deposition. The relative mRNA expression of osteogenic marker genes COL I, OCN, and OPN showed 5.5, 18.7, and 7.2 folds increase from day 14 to day 28, respectively, confirming bone regeneration. From animal studies, the rBMSCs-seeded hybrid constructs could repair mid-diaphyseal tibia defects in rabbits, as evaluated by micro-computed tomography (μ-CT) and histological analyses. The hybrid scaffold will be useful for bone regeneration in load-bearing areas.
Collapse
Affiliation(s)
- K T Shalumon
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
| | - Chak-Bor Wong
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
| | - Yen-Miao Chien
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
| | - Huai-An Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Kwei-San, Taoyuan 33305, Taiwan.
- Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
49
|
Wang YL, Hong A, Yen TH, Hong HH. Isolation of Mesenchymal Stem Cells from Human Alveolar Periosteum and Effects of Vitamin D on Osteogenic Activity of Periosteum-derived Cells. J Vis Exp 2018:57166. [PMID: 29782010 PMCID: PMC6101109 DOI: 10.3791/57166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are present in a variety of tissues and can be differentiated into numerous cell types, including osteoblasts. Among the dental sources of MSCs, the periosteum is an easily accessible tissue, which has been identified to contain MSCs in the cambium layer. However, this source has not yet been widely studied. Vitamin D3 and 1,25-(OH)2D3 have been demonstrated to stimulate in vitro differentiation of MSCs into osteoblasts. In addition, vitamin C facilitates collagen formation and bone cell growth. However, no study has yet investigated the effects of Vitamin D3 and Vitamin C on MSCs. Here, we present a method of isolating MSCs from human alveolar periosteum and examine the hypothesis that 1,25-(OH)2D3 may exert an osteoinductive effect on these cells. We also investigate the presence of MSCs in the human alveolar periosteum and assess stem cell adhesion and proliferation. To assess the ability of vitamin C (as a control) and various concentrations of 1,25-(OH)2D3 (10-10, 10-9, 10-8, and 10-7 M) to alter key mRNA biomarkers in isolated MSCs mRNA expression of alkaline phosphatase (ALP), bone sialoprotein (BSP), core binding factor alpha-1 (CBFA1), collagen-1, and osteocalcin (OCN) are measured using real-time polymerase chain reaction (RT-PCR).
Collapse
Affiliation(s)
- Yen-Li Wang
- Chang Gung University; Department of Periodontics, Chang Gung Memorial Hospital
| | - Adrienne Hong
- California Northstate University College of Medicine
| | - Tzung-Hai Yen
- Chang Gung University; Department of Nephrology, Clinical Poison Center, Chang Gung Memorial Hospital; Kidney Research Center, Chang Gung Memorial Hospital; Center for Tissue Engineering, Chang Gung Memorial Hospital
| | - Hsiang-Hsi Hong
- Chang Gung University; Department of Periodontics, Chang Gung Memorial Hospital; College of Oral Medicine, Taipei Medical University;
| |
Collapse
|
50
|
Becherucci V, Piccini L, Casamassima S, Bisin S, Gori V, Gentile F, Ceccantini R, De Rienzo E, Bindi B, Pavan P, Cunial V, Allegro E, Ermini S, Brugnolo F, Astori G, Bambi F. Human platelet lysate in mesenchymal stromal cell expansion according to a GMP grade protocol: a cell factory experience. Stem Cell Res Ther 2018; 9:124. [PMID: 29720245 PMCID: PMC5930506 DOI: 10.1186/s13287-018-0863-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/23/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The use of platelet lysate (PL) for the ex-vivo expansion of mesenchymal stromal/stem cells (MSCs) was initially proposed by Doucet et al. in 2005, as an alternative to animal serum. Moreover, regulatory authorities discourage the use of fetal bovine serum (FBS) or other animal derivatives, to avoid risk of zoonoses and xenogeneic immune reactions. Even if many studies investigated PL composition, there still are some open issues related to its use in ex-vivo MSC expansion, especially according to good manufacturing practice (GMP) grade protocols. METHODS As an authorized cell factory, we report our experience using standardized PL produced by Azienda Ospedaliero Universitaria Meyer Transfusion Service for MSC expansion according to a GMP grade clinical protocol. As suggested by other authors, we performed an in-vitro test on MSCs versus MSCs cultured with FBS that still represents the best way to test PL batches. We compared 12 MSC batches cultured with DMEM 5% PL with similar batches cultured with DMEM 10% FBS, focusing on the MSC proliferation rate, MSC surface marker expression, MSC immunomodulatory and differentiation potential, and finally MSC relative telomere length. RESULTS Results confirmed the literature data as PL increases cell proliferation without affecting the MSC immunophenotype, immunomodulatory potential, differentiation potential and relative telomere length. CONCLUSIONS PL can be considered a safe alternative to FBS for ex-vivo expansion of MSC according to a GMP grade protocol. Our experience confirms the literature data: a large number of MSCs for clinical applications can be obtained by expansion with PL, without affecting the MSC main features. Our experience underlines the benefits of a close collaboration between the PL producers (transfusion service) and the end users (cell factory) in a synergy of skills and experiences that can lead to standardized PL production.
Collapse
Affiliation(s)
| | - Luisa Piccini
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Serena Casamassima
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Silvia Bisin
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Valentina Gori
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Francesca Gentile
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Riccardo Ceccantini
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Elena De Rienzo
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Barbara Bindi
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Paola Pavan
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Vanessa Cunial
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Elisa Allegro
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Stefano Ermini
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Francesca Brugnolo
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Cellular Therapy and Hematology, San Bortolo Hospital, Vicenza, Italy
| | - Franco Bambi
- Cell Factory Meyer, "A. Meyer" University Children's Hospital, Florence, Italy
| |
Collapse
|