1
|
Vogt A, Faher A, Kucharczak J, Birch M, McCaskie A, Khan W. The Effects of Gender on Mesenchymal Stromal Cell (MSC) Proliferation and Differentiation In Vitro: A Systematic Review. Int J Mol Sci 2024; 25:13585. [PMID: 39769346 PMCID: PMC11677156 DOI: 10.3390/ijms252413585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Mesenchymal stromal cells (MSCs) have the potential for novel treatments of several musculoskeletal conditions due to their ability to differentiate into several cell lineages including chondrocytes, adipocytes and osteocytes. Researchers are exploring whether this could be utilized for novel therapies for joint afflictions. The role of gender in the ability of MSCs to differentiate and proliferate into different cells has not been clearly defined. This systematic review aims to report the current literature on studies, characterized by high quality and in-depth analysis even though quantitatively limited, that have looked at the role of gender in the differentiation and proliferation of MSCs. Sixteen studies were identified during the literature search, reporting 533 patients, of which 202 were male and 331 were female. MSC proliferation, phenotypic analysis and differentiation are reported and contrasted in terms of donor gender. Heterogeneity in methodologies across studies likely contributes to the inconclusive findings presented here, with no discernible statistical disparity observed between genders in differentiation traits. Nevertheless, the proliferation results indicate a notable gender-related impact. Future investigations should aim to ascertain the potential influence of gender on MSC proliferation capacities more conclusively, emphasizing the necessity of standardized protocols for MSC analyses to enhance accuracy and comparability across studies.
Collapse
Affiliation(s)
- Antonia Vogt
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| | - Anissa Faher
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 2SP, UK
| | - Joanna Kucharczak
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 2SP, UK
| | - Mark Birch
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| | - Andrew McCaskie
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; (A.V.)
| |
Collapse
|
2
|
Pal D, Das P, Roy S, Mukherjee P, Halder S, Ghosh D, Nandi SK. Recent trends of stem cell therapies in the management of orthopedic surgical challenges. Int J Surg 2024; 110:6330-6344. [PMID: 38716973 PMCID: PMC11487011 DOI: 10.1097/js9.0000000000001524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/14/2024] [Indexed: 10/20/2024]
Abstract
Emerged health-related problems especially with increasing population and with the wider occurrence of these issues have always put the utmost concern and led medicine to outgrow its usual mode of treatment, to achieve better outcomes. Orthopedic interventions are one of the most concerning hitches, requiring advancement in several issues, that show complications with conventional approaches. Advanced studies have been undertaken to address the issue, among which stem cell therapy emerged as a better area of growth. The capacity of the stem cells to renovate themselves and adapt into different cell types made it possible to implement its use as a regenerative slant. Harvesting the stem cells, particularly mesenchymal stem cells (MSCs) is easier and can be further grown in vitro . In this review, we have discussed orthopedic-related issues including bone defects and fractures, nonunions, ligament and tendon injuries, degenerative changes, and associated conditions, which require further approaches to execute better outcomes, and the advanced strategies that can be tagged along with various ways of application of MSCs. It aims to objectify the idea of stem cells, with a major focus on the application of MSCs from different sources in various orthopedic interventions. It also discusses the limitations, and future scopes for further approaches in the field of regenerative medicine. The involvement of MSCs may transition the procedures in orthopedic interventions from predominantly surgical substitution and reconstruction to bio-regeneration and prevention. Nevertheless, additional improvements and evaluations are required to explore the effectiveness and safety of mesenchymal stem cell treatment in orthopedic regenerative medicine.
Collapse
Affiliation(s)
| | - Pratik Das
- Department of Veterinary Surgery and Radiology
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal
| | | | | | | |
Collapse
|
3
|
Cho Y, Lee H, Jeong W, Jung KB, Lee SY, Park S, Yeun J, Kwon O, Son JG, Lee TG, Son MY, Im SG. Long-Term Culture of Human Pluripotent Stem Cells in Xeno-Free Condition Using Functional Polymer Films. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403952. [PMID: 39015054 DOI: 10.1002/adma.202403952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/13/2024] [Indexed: 07/18/2024]
Abstract
Human pluripotent stem cells (hPSCs), encompassing human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold immense potential in regenerative medicine, offering new opportunities for personalized cell therapies. However, their clinical translation is hindered by the inevitable reliance on xenogeneic components in culture environments. This study addresses this challenge by engineering a fully synthetic, xeno-free culture substrate, whose surface composition is tailored systematically for xeno-free culture of hPSCs. A functional polymer surface, pGC2 (poly(glycidyl methacrylate-grafting-guanidine-co-carboxylic acrylate)), offers excellent cell-adhesive properties as well as non-cytotoxicity, enabling robust hESCs and hiPSCs growth while presenting cost-competitiveness and scalability over Matrigel. This investigation includes comprehensive evaluations of pGC2 across diverse experimental conditions, demonstrating its wide adaptability with various pluripotent stem cell lines, culture media, and substrates. Crucially, pGC2 supports long-term hESCs and hiPSCs expansion, up to ten passages without compromising their stemness and pluripotency. Notably, this study is the first to confirm an identical proteomic profile after ten passages of xeno-free cultivation of hiPSCs on a polymeric substrate compared to Matrigel. The innovative substrate bridges the gap between laboratory research and clinical translation, offering a new promising avenue for advancing stem cell-based therapies.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hana Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Wonji Jeong
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kwang Bo Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Sun Young Lee
- Korea Research Institute of Standards and Science (KRISS), Daejeon, 34141, Republic of Korea
| | - Seonghyeon Park
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jemin Yeun
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Ohman Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Jin Gyeong Son
- Korea Research Institute of Standards and Science (KRISS), Daejeon, 34141, Republic of Korea
| | - Tae Geol Lee
- Korea Research Institute of Standards and Science (KRISS), Daejeon, 34141, Republic of Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sung Gap Im
- Functional Thin Film Laboratory, Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
4
|
Toholova J, Hornak S, Kuricova M. Non-surgical pain management for hip joint disease in veterinary medicine. VET MED-CZECH 2024; 69:261-272. [PMID: 39296629 PMCID: PMC11406501 DOI: 10.17221/19/2024-vetmed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/24/2024] [Indexed: 09/21/2024] Open
Abstract
The most common orthopaedic developmental disease in dogs is hip dysplasia. This condition results in coxofemoral laxity due to incongruity and lack of stabilisation of the joint by the soft tissues. Currently, there is no therapeutic plan to correct hip dysplasia without surgical intervention at a very early age. The goal of the non-surgical treatment is to relieve pain and stiffness and to increase the muscle strength, usually through hydrotherapy and the beneficial physical properties of water. Recently, there has been growing interest in regenerative medicine, which involves the use of mesenchymal stem cells (MSCs) and their products to alleviate the characteristic clinical symptoms of osteoarthritis (OA). In vivo studies with canine MSCs have shown that an intra-articular injection of MSCs into cartilage lesions leads to the excellent regeneration of the hyaline cartilage. Regenerative medicine has undergone rapid development in recent years thanks to new therapies based on the application and combination of innovative biomaterials. One of the first known regenerative methods to be used in clinical practice was platelet-rich plasma (PRP). This review summarises the use and potential of MSCs and PRP, including their in vitro properties, their therapeutic effects in the treatment of cartilage lesions in preclinical in vivo studies, their clinical efficacy in the treatment of naturally occurring OA in dogs, and the current limitations of the studies.
Collapse
Affiliation(s)
- Jana Toholova
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Košice, Slovak Republic
| | - Slavomir Hornak
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Košice, Slovak Republic
| | - Maria Kuricova
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Košice, Slovak Republic
| |
Collapse
|
5
|
Ziverec A, Bax D, Cameron R, Best S, Pasdeloup M, Courtial EJ, Mallein-Gerin F, Malcor JD. The diazirine-mediated photo-crosslinking of collagen improves biomaterial mechanical properties and cellular interactions. Acta Biomater 2024; 180:230-243. [PMID: 38574880 DOI: 10.1016/j.actbio.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Abstract
In tissue engineering, crosslinking with carbodiimides such as EDC is omnipresent to improve the mechanical properties of biomaterials. However, in collagen biomaterials, EDC reacts with glutamate or aspartate residues, inactivating the binding sites for cellular receptors and rendering collagen inert to many cell types. In this work, we have developed a crosslinking method that ameliorates the rigidity, stability, and degradation rate of collagen biomaterials, whilst retaining key interactions between cells and the native collagen sequence. Our approach relies on the UV-triggered reaction of diazirine groups grafted on lysines, leaving critical amino acid residues intact. Notably, GxxGER recognition motifs for collagen-binding integrins, ablated by EDC crosslinking, were left unreacted, enabling cell attachment, spreading, and colonization on films and porous scaffolds. In addition, our procedure conserves the architecture of biomaterials, improves their resistance to collagenase and cellular contraction, and yields material stiffness akin to that obtained with EDC. Importantly, diazirine-crosslinked collagen can host mesenchymal stem cells, highlighting its strong potential as a substrate for tissue repair. We have therefore established a new crosslinking strategy to modulate the mechanical features of collagen porous scaffolds without altering its biological properties, thereby offering an advantageous alternative to carbodiimide treatment. STATEMENT OF SIGNIFICANCE: This article describes an approach to improve the mechanical properties of collagen porous scaffolds, without impacting collagen's natural interactions with cells. This is significant because collagen crosslinking is overwhelmingly performed using carbodiimides, which results in a critical loss of cellular affinity. By contrast, our method leaves key cellular binding sites in the collagen sequence intact, enabling cell-biomaterial interactions. It relies on the fast, UV-triggered reaction of diazirine with collagen, and does not produce toxic by-products. It also supports the culture of mesenchymal stem cells, a pivotal cell type in a wide range of tissue repair applications. Overall, our approach offers an attractive option for the crosslinking of collagen, a prominent material in the growing field of tissue engineering.
Collapse
Affiliation(s)
- Audrey Ziverec
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Daniel Bax
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Rd, Cambridge CB3 0FS, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Rd, Cambridge CB3 0FS, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Rd, Cambridge CB3 0FS, United Kingdom
| | - Marielle Pasdeloup
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Edwin-Joffrey Courtial
- 3dFAB, Univ Lyon, Université Lyon1, CNRS, INSA, CPE-Lyon, ICBMS, UMR 5246, 43, Bd du 11 novembre 1918, 69622 Villeurbanne, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France.
| |
Collapse
|
6
|
Jankowski M, Farzaneh M, Ghaedrahmati F, Shirvaliloo M, Moalemnia A, Kulus M, Ziemak H, Chwarzyński M, Dzięgiel P, Zabel M, Piotrowska-Kempisty H, Bukowska D, Antosik P, Mozdziak P, Kempisty B. Unveiling Mesenchymal Stem Cells' Regenerative Potential in Clinical Applications: Insights in miRNA and lncRNA Implications. Cells 2023; 12:2559. [PMID: 37947637 PMCID: PMC10649218 DOI: 10.3390/cells12212559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023] Open
Abstract
It is now widely recognized that mesenchymal stem cells (MSCs) possess the capacity to differentiate into a wide array of cell types. Numerous studies have identified the role of lncRNA in the regulation of MSC differentiation. It is important to elucidate the role and interplay of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in the regulation of signalling pathways that govern MSC function. Furthermore, miRNAs and lncRNAs are important clinical for innovative strategies aimed at addressing a wide spectrum of existing and emerging disease. Hence it is important to consider their impact on MSC function and differentiation. Examining the data available in public databases, we have collected the literature containing the latest discoveries pertaining to human stem cells and their potential in both fundamental research and clinical applications. Furthermore, we have compiled completed clinical studies that revolve around the application of MSCs, shedding light on the opportunities presented by harnessing the regulatory potential of miRNAs and lncRNAs. This exploration of the therapeutic possibilities offered by miRNAs and lncRNAs within MSCs unveils exciting prospects for the development of precision therapies and personalized treatment approaches. Ultimately, these advancements promise to augment the efficacy of regenerative strategies and produce positive outcomes for patients. As research in this field continues to evolve, it is imperative to explore and exploit the vast potential of miRNAs and lncRNAs as therapeutic agents. The findings provide a solid basis for ongoing investigations, fuelling the quest to fully unlock the regenerative potential of MSCs.
Collapse
Affiliation(s)
- Maurycy Jankowski
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, 60-812 Poznan, Poland;
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Milad Shirvaliloo
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Future Science Group, Unitec House, 2 Albert Place, London N3 1QB, UK
| | - Arash Moalemnia
- Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Hanna Ziemak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Mikołaj Chwarzyński
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, 50-038 Wroclaw, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27607, USA
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27613, USA
- Division of Anatomy, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 602 00 Brno, Czech Republic
| |
Collapse
|
7
|
Vargas-Rodríguez P, Cuenca-Martagón A, Castillo-González J, Serrano-Martínez I, Luque RM, Delgado M, González-Rey E. Novel Therapeutic Opportunities for Neurodegenerative Diseases with Mesenchymal Stem Cells: The Focus on Modulating the Blood-Brain Barrier. Int J Mol Sci 2023; 24:14117. [PMID: 37762420 PMCID: PMC10531435 DOI: 10.3390/ijms241814117] [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: 08/02/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Neurodegenerative disorders encompass a broad spectrum of profoundly disabling situations that impact millions of individuals globally. While their underlying causes and pathophysiology display considerable diversity and remain incompletely understood, a mounting body of evidence indicates that the disruption of blood-brain barrier (BBB) permeability, resulting in brain damage and neuroinflammation, is a common feature among them. Consequently, targeting the BBB has emerged as an innovative therapeutic strategy for addressing neurological disorders. Within this review, we not only explore the neuroprotective, neurotrophic, and immunomodulatory benefits of mesenchymal stem cells (MSCs) in combating neurodegeneration but also delve into their recent role in modulating the BBB. We will investigate the cellular and molecular mechanisms through which MSC treatment impacts primary age-related neurological conditions like Alzheimer's disease, Parkinson's disease, and stroke, as well as immune-mediated diseases such as multiple sclerosis. Our focus will center on how MSCs participate in the modulation of cell transporters, matrix remodeling, stabilization of cell-junction components, and restoration of BBB network integrity in these pathological contexts.
Collapse
Affiliation(s)
- Pablo Vargas-Rodríguez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.V.-R.); (J.C.-G.); (I.S.-M.); (M.D.)
| | - Alejandro Cuenca-Martagón
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; (A.C.-M.); (R.M.L.)
| | - Julia Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.V.-R.); (J.C.-G.); (I.S.-M.); (M.D.)
| | - Ignacio Serrano-Martínez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.V.-R.); (J.C.-G.); (I.S.-M.); (M.D.)
| | - Raúl M. Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain; (A.C.-M.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, 14004 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Mario Delgado
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.V.-R.); (J.C.-G.); (I.S.-M.); (M.D.)
| | - Elena González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, 18016 Granada, Spain; (P.V.-R.); (J.C.-G.); (I.S.-M.); (M.D.)
| |
Collapse
|
8
|
Baouche M, Ochota M, Locatelli Y, Mermillod P, Niżański W. Mesenchymal Stem Cells: Generalities and Clinical Significance in Feline and Canine Medicine. Animals (Basel) 2023; 13:1903. [PMID: 37370414 DOI: 10.3390/ani13121903] [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: 03/31/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells: they can proliferate like undifferentiated cells and have the ability to differentiate into different types of cells. A considerable amount of research focuses on the potential therapeutic benefits of MSCs, such as cell therapy or tissue regeneration, and MSCs are considered powerful tools in veterinary regenerative medicine. They are the leading type of adult stem cells in clinical trials owing to their immunosuppressive, immunomodulatory, and anti-inflammatory properties, as well as their low teratogenic risk compared with pluripotent stem cells. The present review details the current understanding of the fundamental biology of MSCs. We focus on MSCs' properties and their characteristics with the goal of providing an overview of therapeutic innovations based on MSCs in canines and felines.
Collapse
Affiliation(s)
- Meriem Baouche
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| | - Małgorzata Ochota
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| | - Yann Locatelli
- Physiology of Reproduction and Behaviors (PRC), UMR085, INRAE, CNRS, University of Tours, 37380 Nouzilly, France
- Museum National d'Histoire Naturelle, Réserve Zoologique de la Haute Touche, 36290 Obterre, France
| | - Pascal Mermillod
- Physiology of Reproduction and Behaviors (PRC), UMR085, INRAE, CNRS, University of Tours, 37380 Nouzilly, France
| | - Wojciech Niżański
- Department of Reproduction and Clinic of Farm Animals, Wrocław University of Environmental and Life Sciences, 50-366 Wrocław, Poland
| |
Collapse
|
9
|
Alarcón-Apablaza J, Prieto R, Rojas M, Fuentes R. Potential of Oral Cavity Stem Cells for Bone Regeneration: A Scoping Review. Cells 2023; 12:1392. [PMID: 37408226 DOI: 10.3390/cells12101392] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 07/07/2023] Open
Abstract
Bone loss is a common problem that ranges from small defects to large defects after trauma, surgery, or congenital malformations. The oral cavity is a rich source of mesenchymal stromal cells (MSCs). Researchers have documented their isolation and studied their osteogenic potential. Therefore, the objective of this review was to analyze and compare the potential of MSCs from the oral cavity for use in bone regeneration. METHODS A scoping review was carried out following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) guidelines. The databases reviewed were PubMed, SCOPUS, Scientific Electronic Library Online (SciELO), and Web of Science. Studies using stem cells from the oral cavity to promote bone regeneration were included. RESULTS A total of 726 studies were found, of which 27 were selected. The MSCs used to repair bone defects were (I) dental pulp stem cells of permanent teeth, (II) stem cells derived from inflamed dental pulp, (III) stem cells from exfoliated deciduous teeth, (IV) periodontal ligament stem cells, (V) cultured autogenous periosteal cells, (VI) buccal fat pad-derived cells, and (VII) autologous bone-derived mesenchymal stem cells. Stem cells associate with scaffolds to facilitate insertion into the bone defect and to enhance bone regeneration. The biological risk and morbidity of the MSC-grafted site were minimal. Successful bone formation after MSC grafting has been shown for small defects with stem cells from the periodontal ligament and dental pulp as well as larger defects with stem cells from the periosteum, bone, and buccal fat pad. CONCLUSIONS Stem cells of maxillofacial origin are a promising alternative to treat small and large craniofacial bone defects; however, an additional scaffold complement is required for stem cell delivery.
Collapse
Affiliation(s)
- Josefa Alarcón-Apablaza
- Research Centre in Dental Sciences (CICO-UFRO), Dental School, Universidad de La Frontera, Temuco 4780000, Chile
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Ruth Prieto
- Department of Pediatrics and Pediatric Surgery, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile
| | - Mariana Rojas
- Comparative Embryology Laboratory, Program of Anatomy and Developmental Biology, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8320000, Chile
| | - Ramón Fuentes
- Research Centre in Dental Sciences (CICO-UFRO), Dental School, Universidad de La Frontera, Temuco 4780000, Chile
- Department of Integral Adults Dentistry, Dental School, Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
10
|
Iwobi N, Sparks NR. Endocrine Disruptor-Induced Bone Damage Due to Hormone Dysregulation: A Review. Int J Mol Sci 2023; 24:ijms24098263. [PMID: 37175969 PMCID: PMC10179611 DOI: 10.3390/ijms24098263] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Hormones are indispensable for bone development, growth, and maintenance. While many of the genes associated with osteogenesis are well established, it is the recent findings in endocrinology that are advancing the fields of bone biology and toxicology. Endocrine-disrupting chemicals (EDCs) are defined as chemicals that interfere with the function of the endocrine system. Here, we report recent discoveries describing key hormone pathways involved in osteogenesis and the EDCs that alter these pathways. EDCs can lead to bone morphological changes via altering hormone receptors, signaling pathways, and gene expression. The objective of this review is to highlight the recent discoveries of the harmful effects of environmental toxicants on bone formation and the pathways impacted. Understanding the mechanisms of how EDCs interfere with bone formation contributes to providing a comprehensive toxicological profile of a chemical.
Collapse
Affiliation(s)
- Nneamaka Iwobi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California, Irvine, CA 92697, USA
| | - Nicole R Sparks
- Department of Occupational and Environmental Health, University of California, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Toosi S, Naderi-Meshkin H, Moradi A, Daliri M, Moghimi V, Majd HM, Sahebkar AH, Heirani-Tabasi A, Behravan J. Scaphoid Bone Nonunions: Clinical and Functional Outcomes of Collagen/PGA Scaffolds and Cell-Based Therapy. ACS Biomater Sci Eng 2023; 9:1928-1939. [PMID: 36939654 DOI: 10.1021/acsbiomaterials.2c00677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
In this study, the procedure for treating the nonunion complication of scaphoid fractures using collagen/poly glycolic acid (CPGA) scaffolds with bone marrow mesenchymal stem cell (BM-MSC) therapy was adopted and compared with the commonly employed autologous bone tissue graft. With conducting a two-armed clinical trial, 10 patients with scaphoid nonunions were enrolled in this investigation. Patients were randomly assigned to two groups treated with (1) CPGA + cell therapy and (2) autologous iliac crest bone graft standard therapy. Treatment outcomes were evaluated three months after surgery, measuring the grip and pinch strengths and wrist range of motion, with two questionnaires: Patient-Rated Wrist Evaluation (PRWE) and Quick form of Disabilities of the Arm, Shoulder, and Hand (QDASH). We have also assessed the union rate using clinical and radiologic healing criteria one and three months post-operatively. Restorative effects of CPGA + cell therapy were similar to those of the autologous bone graft standard therapy, except for the grip strength (P = 0.048) and QDASH score (P = 0.044) changes, which were higher in the CPGA + cell therapy group. Three months following the surgery, radiographic images and computed tomography (CT) scans also demonstrated that the scaphoid union rate in the test group was comparable to that of scaphoids treated with the standard autograft method. Our findings demonstrate that the CPGA + cell therapy is a potential alternative for bone grafting in the treatment of bone nonunions.
Collapse
Affiliation(s)
- Shirin Toosi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Science, Mahhad 9177899191, Iran
| | - Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad 91775-1376, Iran
| | - Ali Moradi
- Orthopedics Research Center, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
| | - Mahla Daliri
- Orthopedics Research Center, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
| | - Vahid Moghimi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad 91775-1376, Iran
| | - Hasan-Mehrad Majd
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 91388-13944, Iran
| | - Amir Hossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran
| | - Asieh Heirani-Tabasi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center Hospital, Tehran University of Medical Sciences, Tehran 14535, Iran
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177899191, Iran.,School of Pharmacy, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
12
|
Carmon I, Kalmus S, Zobrab A, Alterman M, Emram R, Gussarsky M, Kandel L, Reich E, Casap N, Dvir-Ginzberg M. Repairing a critical cranial defect using WISP1-pretreated chondrocyte scaffolds. J Tissue Eng 2023; 14:20417314231159740. [PMID: 36949842 PMCID: PMC10026108 DOI: 10.1177/20417314231159740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/09/2023] [Indexed: 03/19/2023] Open
Abstract
In cranial flat bone fractures, spontaneous bone repair will occur only when the fracture ends are in close contact. However, in cases wherein bone discontinuity is extensive, surgical interventions are often required. To this end, autologous bone is harvested and surgically integrated into the site of fracture. Here we propose to use cartilage, as an alternative autologous source, to promote cranial fracture repair. The advantage of this approach is the potential reduction in donor site morbidity, likely due to the avascular and aneural nature of cartilage. As a first step we attempted to induce cartilage mineralization in vitro, using micromass primary chondrocyte cultures, incubated with BMP2 and/or WISP1, which were examined histologically following a 3-week culture period. Next, chondrocyte seeded collagen scaffolds were evaluated in vitro for expression profiles and ALP activity. Finally, chondrocyte-seeded collagen scaffolds were implanted in a Lewis rats 8 mm critical calvaria defect model, which was imaged via live CT for 12 weeks until sacrifice. End points were analyzed for microCT, histology, and serum levels of bone related markers. Micromass cultures exhibited an osseous inducing trend following WISP1 administration, which was maintained in chondrocyte seeded scaffolds. Accordingly, in vivo analysis was carried out to assess the impact of WISP1-pretreated chondrocytes (WCS) versus untreated chondrocytes (UCS) in calvaria defect model and compared to untreated control comprised of a defect-associated blood clot (BC) or empty collagen scaffold (CS) implant. Live CT and microCT exhibited higher mineralization volumes in critical defect implanted with UCS, with some structural improvements in WCS. Histological analysis exhibited higher anabolic bone formation in WCS and trabecular bone was detected in WCS and UCS groups. Chondrocytes implanted into critical cranial defect expedite the formation of native-like osseous tissue, especially after WISP1 priming in culture. Ultimately, these data support the use of autologous chondrocytes to repair critical maxillofacial defects.
Collapse
Affiliation(s)
- Idan Carmon
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Shira Kalmus
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Anna Zobrab
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Michael Alterman
- Deptatement. of Maxillofacial Surgery,
Faculty of Dental Medicine, Hadassah-Hebrew University, Jerusalem, Israel
| | - Raphaelle Emram
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - May Gussarsky
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Leonid Kandel
- Orthopedic Research Unit,
Hadassah-Hebrew University, Jerusalem, Israel
| | - Eli Reich
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
| | - Nardi Casap
- Deptatement. of Maxillofacial Surgery,
Faculty of Dental Medicine, Hadassah-Hebrew University, Jerusalem, Israel
| | - Mona Dvir-Ginzberg
- Laboratory of Cartilage Biology,
Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine, Hebrew
University of Jerusalem, Jerusalem, Israel
- Mona Dvir-Ginzberg, Laboratory of Cartilage
Biology, Institute of Bio-Medical and Oral Research, Faculty of Dental Medicine,
Hebrew University of Jerusalem, P. O. Box 12272, Jerusalem 9112102, Israel.
| |
Collapse
|
13
|
Malcor JD, Mallein-Gerin F. Biomaterial functionalization with triple-helical peptides for tissue engineering. Acta Biomater 2022; 148:1-21. [PMID: 35675889 DOI: 10.1016/j.actbio.2022.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022]
Abstract
In the growing field of tissue engineering, providing cells in biomaterials with the adequate biological cues represents an increasingly important challenge. Yet, biomaterials with excellent mechanical properties often are often biologically inert to many cell types. To address this issue, researchers resort to functionalization, i.e. the surface modification of a biomaterial with active molecules or substances. Functionalization notably aims to replicate the native cellular microenvironment provided by the extracellular matrix, and in particular by collagen, its major component. As our understanding of biological processes regulating cell behaviour increases, functionalization with biomolecules binding cell surface receptors constitutes a promising strategy. Amongst these, triple-helical peptides (THPs) that reproduce the architectural and biological properties of collagen are especially attractive. Indeed, THPs containing binding sites from the native collagen sequence have successfully been used to guide cell response by establishing cell-biomaterial interactions. Notably, the GFOGER motif recognising the collagen-binding integrins is extensively employed as a cell adhesive peptide. In biomaterials, THPs efficiently improved cell adhesion, differentiation and function on biomaterials designed for tissue repair (especially for bone, cartilage, tendon and heart), vascular graft fabrication, wound dressing, drug delivery or immunomodulation. This review describes the key characteristics of THPs, their effect on cells when combined to biomaterials and their strong potential as biomimetic tools for regenerative medicine. STATEMENT OF SIGNIFICANCE: This review article describes how triple-helical peptides constitute efficient tools to improve cell-biomaterial interactions in tissue engineering. Triple helical peptides are bioactive molecules that mimic the architectural and biological properties of collagen. They have been successfully used to specifically recognize cell-surface receptors and provide cells seeded on biomaterials with controlled biological cues. Functionalization with triple-helical peptides has enabled researchers to improve cell function for regenerative medicine applications, such as tissue repair. However, despite encouraging results, this approach remains limited and under-exploited, and most functionalization strategies reported in the literature rely on biomolecules that are unable to address collagen-binding receptors. This review will assist researchers in selecting the correct tools to functionalize biomaterials in efforts to guide cellular response.
Collapse
Affiliation(s)
- Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, Cedex 07, Lyon 69367, France.
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, Cedex 07, Lyon 69367, France
| |
Collapse
|
14
|
Staniowski T, Zawadzka-Knefel A, Skośkiewicz-Malinowska K. Therapeutic Potential of Dental Pulp Stem Cells According to Different Transplant Types. Molecules 2021; 26:7423. [PMID: 34946506 PMCID: PMC8707085 DOI: 10.3390/molecules26247423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 12/13/2022] Open
Abstract
Stem cells are unspecialised cells capable of perpetual self-renewal, proliferation and differentiation into more specialised daughter cells. They are present in many tissues and organs, including the stomatognathic system. Recently, the great interest of scientists in obtaining stem cells from human teeth is due to their easy availability and a non-invasive procedure of collecting the material. Three key components are required for tissue regeneration: stem cells, appropriate scaffold material and growth factors. Depending on the source of the new tissue or organ, there are several types of transplants. In this review, the following division into four transplant types is applied due to genetic differences between the donor and the recipient: xenotransplantation, allotransplantation, autotransplantation and isotransplantation (however, due to the lack of research, type was not included). In vivo studies have shown that Dental Pulp Stem Cells (DPSCs)can form a dentin-pulp complex, nerves, adipose, bone, cartilage, skin, blood vessels and myocardium, which gives hope for their use in various biomedical areas, such as immunotherapy and regenerative therapy. This review presents the current in vivo research and advances to provide new biological insights and therapeutic possibilities of using DPSCs.
Collapse
Affiliation(s)
| | - Anna Zawadzka-Knefel
- Department of Conservative Dentistry with Endodontics, Wroclaw Medical University, 50-425 Wrocław, Poland; (T.S.); (K.S.-M.)
| | | |
Collapse
|
15
|
Effect of biomolecules derived from human platelet-rich plasma on the ex vivo expansion of human adipose-derived mesenchymal stem cells for clinical applications. Biologicals 2021; 75:37-48. [PMID: 34785135 DOI: 10.1016/j.biologicals.2021.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal stem cells are a tool in cell therapies but demand a large cell number per treatment, for that, suitable culture media is required which contains fetal bovine serum (FBS). However, for cell-based therapy applications, the use of FBS is problematic. Several alternatives to FBS have been explored, including human derivatives from platelet-rich plasma (hD-PRP). Although various studies have evaluated the impact of hD-PRP on MSC proliferation and differentiation, few of them have assessed their influence on processes, such as metabolism and gene expression. Here, we cultured human adipose-derived MSCs (hAD-MSCs) in media supplemented with either 10% hD-PRP (hD-PRP-SM) or 10% FBS (FBS-SM) in order to characterize them and evaluate the effect of hD-PRP on cell metabolism, gene expression of associated regenerative factors, as well as chromosome stability during cell expansion. We found that hAD-MSCs cultured in hD-PRP-SM have a greater cell elongation but express similar surface markers; in addition, hD-PRP-SM promoted a significant osteogenic differentiation in the absence of differentiation medium and increased the growth rate, maintaining chromosomal stability. In terms of cell metabolic profile, hAD-MSC behavior did not reveal any differences between both culture conditions. Conversely, significant differences in collagen I and angiopoietin 2 expression were observed between both conditions. The present results suggest that hD-PRP may influence hAD-MSC behavior.
Collapse
|
16
|
Vatsa P, Negi R, Ansari UA, Khanna VK, Pant AB. Insights of Extracellular Vesicles of Mesenchymal Stem Cells: a Prospective Cell-Free Regenerative Medicine for Neurodegenerative Disorders. Mol Neurobiol 2021; 59:459-474. [PMID: 34714469 DOI: 10.1007/s12035-021-02603-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent, adult stem cells which are found in numerous tissues like the umbilical cord, Wharton's jelly, bone marrow, and adipose tissue. They possess the capacity of self-renewal by dividing and differentiating into various cellular lineages. Their characteristic therapeutic potential exploited so far has made them a desirable candidate in regenerative medicine. Neurodegenerative diseases (NDs) like Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and ischemic stroke have been treated with MSCs and MSC-derived products. Over the past few decades, we have witnessed significant contributions in discovering the etiology of various NDs and their possible therapeutic solutions. One of the MSC-based therapeutics is extracellular vesicles (EVs), which contain multiple biologically active molecules like nucleic acids and proteins. The contents of EVs are ferried between cells for intercellular communication which then leads to regulation of the homeostasis of recipient cells. EVs serve as a considerable means of cell-free therapies like for tissue repair or regeneration as EVs can maintain therapeutically effective cargo of parent cells and are free of various ethical issues in cell-based therapies. Due to paucity of standard protocols in extraction procedures of EVs and their pharmacological properties and mechanisms, the development of new EV dependent therapies is challenging. With this review, an attempt has been made to annotate these mechanisms, which can help advance the novel therapeutic approaches towards the treat and define a more narrowed down approach for each ND to devise effective MSC-based therapies to cure and avert these diseases.
Collapse
Affiliation(s)
- P Vatsa
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - R Negi
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - U A Ansari
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - V K Khanna
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - A B Pant
- System Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, P.O. Box No. 80, Lucknow, Uttar Pradesh, 226001, India.
- CSIR-Human Resource Development Centre (CSIR-HRDC) Campus, Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
17
|
Wiśniewska J, Sadowska A, Wójtowicz A, Słyszewska M, Szóstek-Mioduchowska A. Perspective on Stem Cell Therapy in Organ Fibrosis: Animal Models and Human Studies. Life (Basel) 2021; 11:life11101068. [PMID: 34685439 PMCID: PMC8538998 DOI: 10.3390/life11101068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) components that result from the disruption of regulatory processes responsible for ECM synthesis, deposition, and remodeling. Fibrosis develops in response to a trigger or injury and can occur in nearly all organs of the body. Thus, fibrosis leads to severe pathological conditions that disrupt organ architecture and cause loss of function. It has been estimated that severe fibrotic disorders are responsible for up to one-third of deaths worldwide. Although intensive research on the development of new strategies for fibrosis treatment has been carried out, therapeutic approaches remain limited. Since stem cells, especially mesenchymal stem cells (MSCs), show remarkable self-renewal, differentiation, and immunomodulatory capacity, they have been intensively tested in preclinical studies and clinical trials as a potential tool to slow down the progression of fibrosis and improve the quality of life of patients with fibrotic disorders. In this review, we summarize in vitro studies, preclinical studies performed on animal models of human fibrotic diseases, and recent clinical trials on the efficacy of allogeneic and autologous stem cell applications in severe types of fibrosis that develop in lungs, liver, heart, kidney, uterus, and skin. Although the results of the studies seem to be encouraging, there are many aspects of cell-based therapy, including the cell source, dose, administration route and frequency, timing of delivery, and long-term safety, that remain open areas for future investigation. We also discuss the contemporary status, challenges, and future perspectives of stem cell transplantation for therapeutic options in fibrotic diseases as well as we present recent patents for stem cell-based therapies in organ fibrosis.
Collapse
|
18
|
Banerjee A, Mukherjee S, Maji BK. Worldwide flavor enhancer monosodium glutamate combined with high lipid diet provokes metabolic alterations and systemic anomalies: An overview. Toxicol Rep 2021; 8:938-961. [PMID: 34026558 PMCID: PMC8120859 DOI: 10.1016/j.toxrep.2021.04.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 12/13/2022] Open
Abstract
Flavor enhancing high lipid diet acts as silent killer. Monosodium glutamate mixed with high lipid diet alters redox-status. Monosodium glutamate mixed with high lipid diet induces systemic anomalies.
In this fast-food era, people depend on ready-made foods and engage in minimal physical activities that ultimately change their food habits. Majorities of such foods have harmful effects on human health due to higher percentages of saturated fatty acids, trans-fatty acids, and hydrogenated fats in the form of high lipid diet (HLD). Moreover, food manufacturers add monosodium glutamate (MSG) to enhance the taste and palatability of the HLD. Both MSG and HLD induce the generation of reactive oxygen species (ROS) and thereby alter the redox-homeostasis to cause systemic damage. However, MSG mixed HLD (MH) consumption leads to dyslipidemia, silently develops non-alcoholic fatty liver disease followed by metabolic alterations and systemic anomalies, even malignancies, via modulating different signaling pathways. This comprehensive review formulates health care strategies to create global awareness about the harmful impact of MH on the human body and recommends the daily consumption of more natural foods rich in antioxidants instead of toxic ingredients to counterbalance the MH-induced systemic anomalies.
Collapse
|
19
|
Pievani A, Donsante S, Tomasoni C, Corsi A, Dazzi F, Biondi A, Riminucci M, Serafini M. Acute myeloid leukemia shapes the bone marrow stromal niche in vivo. Haematologica 2021; 106:865-870. [PMID: 32381570 PMCID: PMC7928008 DOI: 10.3324/haematol.2020.247205] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Indexed: 01/04/2023] Open
Affiliation(s)
- Alice Pievani
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | | | - Chiara Tomasoni
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Francesco Dazzi
- Department of Hemato-Oncology, Rayne Institute, King’s College London, London, UK
| | - Andrea Biondi
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
- Department of Pediatrics, Fondazione MBBM/San Gerardo Hospital, Monza, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
20
|
Jeske R, Lewis S, Tsai AC, Sanders K, Liu C, Yuan X, Li Y. Agitation in a Microcarrier-based Spinner Flask Bioreactor Modulates Homeostasis of Human Mesenchymal Stem Cells. Biochem Eng J 2021; 168. [PMID: 33967591 DOI: 10.1016/j.bej.2021.107947] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are well known in cell therapy due to their secretion of trophic factors, multipotent differentiation potential, and ability for self-renewal. As a result, the number of clinical trials has been steadily increasing over the last decade highlighting the need for in vitro systems capable of producing large quantities of cells to meet growing demands. However, hMSCs are highly sensitive to microenvironment conditions, including shear stress caused by dynamic bioreactor systems, and can lead to alteration of cellular homeostasis. In this study, hMSCs were expanded on microcarriers within a 125 mL spinner flask bioreactor system. Our results demonstrate a three-fold expansion over seven days. Furthermore, our results show that culturing hMSCs in the microcarrier-based suspension bioreactor (compared to static planar culture) results in smaller cell size and higher levels of reactive oxidative species (ROS) and ROS regulator Sirtuin-3, which have implications on the nicotinamide adenine dinucleotide metabolic pathway and metabolic homeostasis. In addition, hMSCs in the bioreactor showed the increased Prostaglandin E2 secretion as well as reduced the Indoleamine-pyrrole 2,3-dioxygenase secretion upon stimulus with interferon gamma. The results of this study provide understanding of potential hMSC physiology alterations impacted by bioreactor microenvironment during scalable production of hMSCs for biomanufacturing and clinical trials.
Collapse
Affiliation(s)
- Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Shaquille Lewis
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Kevin Sanders
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Chang Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
21
|
Fathollahi A, Hashemi SM, Haji Molla Hoseini M, Tavakoli S, Farahani E, Yeganeh F. Intranasal administration of small extracellular vesicles derived from mesenchymal stem cells ameliorated the experimental autoimmune encephalomyelitis. Int Immunopharmacol 2021; 90:107207. [PMID: 33290966 DOI: 10.1016/j.intimp.2020.107207] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/26/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a mouse model for the human multiple sclerosis, which is characterized by inflammation in the central nervous system (CNS), de-myelination of axonal neurons, and loss of motor coordination. The aim of the current study was to evaluate the effect of intranasal administration of mesenchymal stem cells (MSCs) and small extracellular vesicle (SEV) derived from the MSC (MSC-SEV) on disease activity and antigen-specific responses in the EAE mouse model. MSCs (5 × 105) were administered intranasally to EAE mice (n = 5) on the 15th and 24th days after immunization. In addition, the intranasal administration of MSC-SEV (10 μg) was used to treat EAE mice (n = 5) on a daily basis from the 15th to the 27th day after induction of the disease. The outcomes of therapies were evaluated using studying clinical symptoms and histological analysis of CNS lesions. Moreover, T cell proliferation, the frequency of regulatory T cells, the expression of transcription factors of T-helper subsets, and the levels of their corresponded cytokines were evaluated in splenocytes culture that was stimulated with specific-antigen. The results of treatment of EAE mice with MSC- SEV and MSC showed a significant decrease in the clinical scores, and it was found that treatment with MSC-SEV was more effective in alleviating clinical scores than MSC. In addition, the decrease in clinical symptoms was associated with an increase in immunomodulatory responses, including an increase in the frequency of Foxp3+ CD25+ regulatory T cells. Moreover, the level of TGF-β was increased by both treatments; however, interleukin-10 was increased only by MSC treatment. Ultimately, it was achieved that the intranasal administration of MSC-SEV to EAE mice was more effective than the administration of MSC to reduce clinical scores and histological lesions of the CNS tissue.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/surgery
- Extracellular Vesicles/immunology
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/transplantation
- Female
- Gene Expression Regulation
- Inflammation Mediators/metabolism
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/immunology
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Mice
Collapse
Affiliation(s)
- Anwar Fathollahi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Tavakoli
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Elnaz Farahani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Farshid Yeganeh
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
A Human Osteochondral Tissue Model Mimicking Cytokine-Induced Key Features of Arthritis In Vitro. Int J Mol Sci 2020; 22:ijms22010128. [PMID: 33374446 PMCID: PMC7794893 DOI: 10.3390/ijms22010128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/15/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
Adequate tissue engineered models are required to further understand the (patho)physiological mechanism involved in the destructive processes of cartilage and subchondral bone during rheumatoid arthritis (RA). Therefore, we developed a human in vitro 3D osteochondral tissue model (OTM), mimicking cytokine-induced cellular and matrix-related changes leading to cartilage degradation and bone destruction in order to ultimately provide a preclinical drug screening tool. To this end, the OTM was engineered by co-cultivation of mesenchymal stromal cell (MSC)-derived bone and cartilage components in a 3D environment. It was comprehensively characterized on cell, protein, and mRNA level. Stimulating the OTM with pro-inflammatory cytokines, relevant in RA (tumor necrosis factor α, interleukin-6, macrophage migration inhibitory factor), caused cell- and matrix-related changes, resulting in a significantly induced gene expression of lactate dehydrogenase A, interleukin-8 and tumor necrosis factor α in both, cartilage and bone, while the matrix metalloproteases 1 and 3 were only induced in cartilage. Finally, application of target-specific drugs prevented the induction of inflammation and matrix-degradation. Thus, we here provide evidence that our human in vitro 3D OTM mimics cytokine-induced cell- and matrix-related changes—key features of RA—and may serve as a preclinical tool for the evaluation of both new targets and potential drugs in a more translational setup.
Collapse
|
23
|
Park JW, Fu S, Huang B, Xu RH. Alternative splicing in mesenchymal stem cell differentiation. Stem Cells 2020; 38:1229-1240. [PMID: 32627865 PMCID: PMC7586970 DOI: 10.1002/stem.3248] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/03/2020] [Accepted: 06/11/2020] [Indexed: 01/07/2023]
Abstract
The differentiation and maturation of mesenchymal stem cells (MSCs) to mesodermal and other lineages are known to be controlled by various extrinsic and intrinsic signals. The dysregulation of the MSC differentiation balance has been linked to several pathophysiological conditions, including obesity and osteoporosis. Previous research of the molecular mechanisms governing MSC differentiation has mostly focused on transcriptional regulation. However, recent findings are revealing the underrated role of alternative splicing (AS) in MSC differentiation and functions. In this review, we discuss recent progress in elucidating the regulatory roles of AS in MSC differentiation. We catalogue and highlight the key AS events that modulate MSC differentiation to major osteocytes, chondrocytes, and adipocytes, and discuss the regulatory mechanisms by which AS is regulated.
Collapse
Affiliation(s)
- Jung Woo Park
- Center for Reproduction, Development, and Aging and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Siyi Fu
- Center for Reproduction, Development, and Aging and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Borong Huang
- Center for Reproduction, Development, and Aging and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Ren-He Xu
- Center for Reproduction, Development, and Aging and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| |
Collapse
|
24
|
Mocini F, Monteleone AS, Piazza P, Cardona V, Vismara V, Messinese P, Campana V, Sircana G, Maccauro G, Saccomanno MF. The role of adipose derived stem cells in the treatment of rotator cuff tears: from basic science to clinical application. Orthop Rev (Pavia) 2020; 12:8682. [PMID: 32913610 PMCID: PMC7459379 DOI: 10.4081/or.2020.8682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, regenerative medicine has become increasingly popular throughout the scientific community. The poor healing capacity at the tendon-bone interface makes the rotator cuff an appealing target for biologic agents. Adipose derived stem cells are mesenchymal cells with the capacity for self-renewal and multipotential differentiation. They have been recently proposed, both in isolation and as adjuvants to existing surgical therapies, for the treatment of rotator cuff tears. Several studies have been carried out in this research field, starting from the biological characteristics of adipose derived stem cells, their preparation and culture, up to the application in the experimental field on animal models and on humans. The purpose of this study was to provide a state of the art about the current basic science and clinical literature for the effectiveness of adipose derived stem cells in the treatment of rotator cuff tears.
Collapse
Affiliation(s)
- Fabrizio Mocini
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | | | - Piero Piazza
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | - Valentina Cardona
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | - Valeria Vismara
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | - Piermarco Messinese
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | - Vincenzo Campana
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | - Giuseppe Sircana
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | - Giulio Maccauro
- Orthopaedic Institute, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | | |
Collapse
|
25
|
Motais B, Charvátová S, Hrdinka M, Šimíček M, Jelínek T, Ševčíková T, Kořístek Z, Hájek R, Bagó JR. A Bird's-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers. Cancers (Basel) 2020; 12:E1333. [PMID: 32456165 PMCID: PMC7281611 DOI: 10.3390/cancers12051333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/25/2022] Open
Abstract
: Hematological malignancies comprise over a hundred different types of cancers and account for around 6.5% of all cancers. Despite the significant improvements in diagnosis and treatment, many of those cancers remain incurable. In recent years, cancer cell-based therapy has become a promising approach to treat those incurable hematological malignancies with striking results in different clinical trials. The most investigated, and the one that has advanced the most, is the cell-based therapy with T lymphocytes modified with chimeric antigen receptors. Those promising initial results prepared the ground to explore other cell-based therapies to treat patients with blood cancer. In this review, we want to provide an overview of the different types of cell-based therapies in blood cancer, describing them according to the cell source.
Collapse
Affiliation(s)
- Benjamin Motais
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Sandra Charvátová
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Matouš Hrdinka
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Michal Šimíček
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tereza Ševčíková
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Zdeněk Kořístek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Roman Hájek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Juli R. Bagó
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| |
Collapse
|
26
|
Sveiven SN, Nordgren TM. Lung-resident mesenchymal stromal cells are tissue-specific regulators of lung homeostasis. Am J Physiol Lung Cell Mol Physiol 2020; 319:L197-L210. [PMID: 32401672 DOI: 10.1152/ajplung.00049.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Until recently, data supporting the tissue-resident status of mesenchymal stromal cells (MSC) has been ambiguous since their discovery in the 1950-60s. These progenitor cells were first discovered as bone marrow-derived adult multipotent cells and believed to migrate to sites of injury, opposing the notion that they are residents of all tissue types. In recent years, however, it has been demonstrated that MSC can be found in all tissues and MSC from different tissues represent distinct populations with differential protein expression unique to each tissue type. Importantly, these cells are efficient mediators of tissue repair, regeneration, and prove to be targets for therapeutics, demonstrated by clinical trials (phase 1-4) for MSC-derived therapies for diseases like graft-versus-host-disease, multiple sclerosis, rheumatoid arthritis, and Crohn's disease. The tissue-resident status of MSC found in the lung is a key feature of their importance in the context of disease and injuries of the respiratory system, since these cells could be instrumental to providing more specific and targeted therapies. Currently, bone marrow-derived MSC have been established in the treatment of disease, including diseases of the lung. However, with lung-resident MSC representing a unique population with a different phenotypic and gene expression pattern than MSC derived from other tissues, their role in remediating lung inflammation and injury could provide enhanced efficacy over bone marrow-derived MSC methods. Through this review, lung-resident MSC will be characterized, using previously published data, by surface markers, gene expression patterns, and compared with bone-marrow MSC to highlight similarities and, importantly, differences in these cell types.
Collapse
Affiliation(s)
- Stefanie Noel Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| | - Tara M Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, California
| |
Collapse
|
27
|
Kruminis-Kaszkiel E, Osowski A, Bejer-Oleńska E, Dziekoński M, Wojtkiewicz J. Differentiation of Human Mesenchymal Stem Cells from Wharton's Jelly Towards Neural Stem Cells Using A Feasible and Repeatable Protocol. Cells 2020; 9:cells9030739. [PMID: 32192154 PMCID: PMC7140706 DOI: 10.3390/cells9030739] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
The transplantation of neural stem cells (NSCs) capable of regenerating to the cells of the central nervous system (CNS) is a promising strategy in the treatment of CNS diseases and injury. As previous studies have highlighted mesenchymal stem cells (MSCs) as a source of NSCs, this study aimed to develop a feasible, efficient, and reproducible method for the neural induction of MSCs isolated from Wharton's jelly (hWJ-MSCs). We induced neural differentiation in a monolayer culture using epidermal growth factor, basic fibroblast growth factor, N2, and B27 supplements. This resulted in a homogenous population of proliferating cells that expressed certain neural markers at both the protein and mRNA levels. Flow cytometry and immunocytochemistry confirmed the expression of neural markers: nestin, sex-determining region Y (SRY) box 1 and 2 (SOX1 and SOX2), microtubule-associated protein 2 (MAP2), and glial fibrillary acidic protein (GFAP). The qRT-PCR analysis revealed significantly enhanced expression of nestin and MAP2 in differentiated cells. This study confirms that it is possible to generate NSCs-like cells from hWJ-MSCs in a 2D culture using a practical method. However, the therapeutic effectiveness of such differentiated cells should be extended to confirm the terminal differentiation ability and electrophysiological properties of neurons derived from them.
Collapse
Affiliation(s)
- Ewa Kruminis-Kaszkiel
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
- Correspondence:
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| | - Ewa Bejer-Oleńska
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| | - Mariusz Dziekoński
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| |
Collapse
|
28
|
Attia N, Mashal M. Mesenchymal Stem Cells: The Past Present and Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1312:107-129. [PMID: 33159306 DOI: 10.1007/5584_2020_595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The biomedical applications of mesenchymal stem cells (MSCs) have gained expanding attention over the past three decades. MSCs are easily obtained from various tissue types (e.g. bone marrow, fat, cord blood, etc.), are capable of self-renewal, and could be induced to differentiate into several cell lineages for countless biomedical applications. In addition, when transplanted, MSCs are not detected by immune surveillance, thus do not lead to graft rejection. Moreover, they can home towards affected tissues and induce their therapeutic effect in a cell-base and/or a cell-free manner. These properties, and many others, have made MSCs appealing therapeutic cell candidates (for cell and/or gene therapy) in myriad clinical conditions. However, similar to any other therapeutic tool, MSCs still have their own limitations and grey areas that entail more research for better understanding and optimization. Herein, we present a brief overview of various pre-clinical/clinical applications of MSCs in regenerative medicine and discuss limitations and future challenges.
Collapse
Affiliation(s)
- Noha Attia
- Department of Basic Sciences, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda. .,The Center of research and evaluation, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda. .,Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria, Egypt. .,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
| | - Mohamed Mashal
- The Center of research and evaluation, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda.,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
29
|
Rodella A, Pozzobon M, Rigon M, Franchin C, Arrigoni G, Simonato M, Ghinelli E, Vedovelli L. Topical application of lyophilized and powdered human amniotic membrane promotes diabetic ulcer healing. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.wndm.2019.100171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Zavala G, Sandoval C, Meza D, Contreras R, Gubelin W, Khoury M. Differentiation of adipose-derived stem cells to functional CD105 neg CD73 low melanocyte precursors guided by defined culture condition. Stem Cell Res Ther 2019; 10:249. [PMID: 31399041 PMCID: PMC6688240 DOI: 10.1186/s13287-019-1364-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/18/2022] Open
Abstract
Background The generation of functional human epidermal melanocytes (HEM) from stem cells provides an unprecedented source for cell-based therapy in vitiligo. Despite the important efforts exerted to obtain melanin-producing cells from stem cells, pre-clinical results still lack the safety and scalability characteristics essential for their translational application. Methods Here, we report a rapid and efficient protocol based on defined culture conditions capable of differentiating adult adipose-derived stem cells (ADSC) to scalable amounts of proliferative melanocyte precursors (PreMel) within 30 days. PreMel were characterized in vitro through qPCR, Western blot, flow cytometry, biochemical assays, and in vivo assays in immunocompromised mice (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ, or NSG). Results After 30 days of differentiation, the stem cell-derived PreMel were defined as CD105neg CD73low according to immunophenotypic changes in comparison with parental stem cell markers. In addition, expression of microphthalmia-associated transcription factor (MITF), active tyrosinase (TYR), and the terminal differentiation-involved premelanosome protein (PMEL) were detected. Furthermore, PreMel had the potential to synthesize melanin and package it into melanosomes both in vitro and in vivo in NSG mice skin. Conclusions This study proposes a rapid and scalable protocol for the generation of proliferative melanocyte precursors (PreMel) from ADSC. These PreMel display the essential functional characteristics of bona fide HEM, opening a new path for an autologous cellular therapy for vitiligo patients. Electronic supplementary material The online version of this article (10.1186/s13287-019-1364-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Zavala
- Consorcio Regenero, La Plaza 2501, Las Condes, Santiago, Chile.,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Carolina Sandoval
- Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Daniel Meza
- Consorcio Regenero, La Plaza 2501, Las Condes, Santiago, Chile.,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Rafael Contreras
- Cells for Cells, La Plaza 2501, Las Condes, Santiago, Chile.,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Walter Gubelin
- Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile
| | - Maroun Khoury
- Consorcio Regenero, La Plaza 2501, Las Condes, Santiago, Chile. .,Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile. .,Cells for Cells, La Plaza 2501, Las Condes, Santiago, Chile. .,Biomedical Research Center (CIB), Faculty of Medicine, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago, Chile.
| |
Collapse
|
31
|
Larson BL, Yu SN, Park H, Estes BT, Moutos FT, Bloomquist CJ, Wu PB, Welter JF, Langer R, Guilak F, Freed LE. Chondrogenic, hypertrophic, and osteochondral differentiation of human mesenchymal stem cells on three-dimensionally woven scaffolds. J Tissue Eng Regen Med 2019; 13:1453-1465. [PMID: 31115161 DOI: 10.1002/term.2899] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022]
Abstract
The development of mechanically functional cartilage and bone tissue constructs of clinically relevant size, as well as their integration with native tissues, remains an important challenge for regenerative medicine. The objective of this study was to assess adult human mesenchymal stem cells (MSCs) in large, three-dimensionally woven poly(ε-caprolactone; PCL) scaffolds in proximity to viable bone, both in a nude rat subcutaneous pouch model and under simulated conditions in vitro. In Study I, various scaffold permutations-PCL alone, PCL-bone, "point-of-care" seeded MSC-PCL-bone, and chondrogenically precultured Ch-MSC-PCL-bone constructs-were implanted in a dorsal, ectopic pouch in a nude rat. After 8 weeks, only cells in the Ch-MSC-PCL constructs exhibited both chondrogenic and osteogenic gene expression profiles. Notably, although both tissue profiles were present, constructs that had been chondrogenically precultured prior to implantation showed a loss of glycosaminoglycan (GAG) as well as the presence of mineralization along with the formation of trabecula-like structures. In Study II of the study, the GAG loss and mineralization observed in Study I in vivo were recapitulated in vitro by the presence of either nearby bone or osteogenic culture medium additives but were prevented by a continued presence of chondrogenic medium additives. These data suggest conditions under which adult human stem cells in combination with polymer scaffolds synthesize functional and phenotypically distinct tissues based on the environmental conditions and highlight the potential influence that paracrine factors from adjacent bone may have on MSC fate, once implanted in vivo for chondral or osteochondral repair.
Collapse
Affiliation(s)
- Benjamin L Larson
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Sarah N Yu
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Hyoungshin Park
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | | | | | | | - Patrick B Wu
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Jean F Welter
- Skeletal Research Center and Case Center for Multimodal Evaluation of Engineered Cartilage, Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Robert Langer
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Farshid Guilak
- Cytex Therapeutics, Inc., Durham, NC.,Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO.,Shriners Hospitals for Children-St. Louis, St. Louis, MO.,Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO
| | - Lisa E Freed
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
32
|
Kozlowska U, Krawczenko A, Futoma K, Jurek T, Rorat M, Patrzalek D, Klimczak A. Similarities and differences between mesenchymal stem/progenitor cells derived from various human tissues. World J Stem Cells 2019; 11:347-374. [PMID: 31293717 PMCID: PMC6600850 DOI: 10.4252/wjsc.v11.i6.347] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/03/2018] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) constitute a promising tool in regenerative medicine and can be isolated from different human tissues. However, their biological properties are still not fully characterized. Whereas MSCs from different tissue exhibit many common characteristics, their biological activity and some markers are different and depend on their tissue of origin. Understanding the factors that underlie MSC biology should constitute important points for consideration for researchers interested in clinical MSC application.
AIM To characterize the biological activity of MSCs during longterm culture isolated from: bone marrow (BM-MSCs), adipose tissue (AT-MSCs), skeletal muscles (SM-MSCs), and skin (SK-MSCs).
METHODS MSCs were isolated from the tissues, cultured for 10 passages, and assessed for: phenotype with immunofluorescence and flow cytometry, multipotency with differentiation capacity for osteo-, chondro-, and adipogenesis, stemness markers with qPCR for mRNA for Sox2 and Oct4, and genetic stability for p53 and c-Myc; 27 bioactive factors were screened using the multiplex ELISA array, and spontaneous fusion involving a co-culture of SM-MSCs with BM-MSCs or AT-MSCs stained with PKH26 (red) or PKH67 (green) was performed.
RESULTS All MSCs showed the basic MSC phenotype; however, their expression decreased during the follow-up period, as confirmed by fluorescence intensity. The examined MSCs express CD146 marker associated with proangiogenic properties; however their expression decreased in AT-MSCs and SM-MSCs, but was maintained in BM-MSCs. In contrast, in SK-MSCs CD146 expression increased in late passages. All MSCs, except BM-MSCs, expressed PW1, a marker associated with differentiation capacity and apoptosis. BM-MSCs and AT-MSCs expressed stemness markers Sox2 and Oct4 in long-term culture. All MSCs showed a stable p53 and c-Myc expression. BM-MSCs and AT-MSCs maintained their differentiation capacity during the follow-up period. In contrast, SK-MSCs and SM-MSCs had a limited ability to differentiate into adipocytes. BM-MSCs and AT-MSCs revealed similarities in phenotype maintenance, capacity for multilineage differentiation, and secretion of bioactive factors. Because AT-MSCs fused with SM-MSCs as effectively as BM-MSCs, AT-MSCs may constitute an alternative source for BM-MSCs.
CONCLUSION Long-term culture affects the biological activity of MSCs obtained from various tissues. The source of MSCs and number of passages are important considerations in regenerative medicine.
Collapse
Affiliation(s)
- Urszula Kozlowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| | - Agnieszka Krawczenko
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| | - Katarzyna Futoma
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| | - Tomasz Jurek
- Department of Forensic Medicine, Wroclaw Medical University, Wroclaw 50-345, Poland
| | - Marta Rorat
- Department of Forensic Medicine, Wroclaw Medical University, Wroclaw 50-345, Poland
| | - Dariusz Patrzalek
- Faculty of Health Science, Department of Physiotherapy, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Aleksandra Klimczak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw 53-114, Poland
| |
Collapse
|
33
|
Guilak F, Sandell LJ, Huard J. Journal of Orthopaedic Research: Special Issue on Stem Cells. J Orthop Res 2019; 37:1209-1211. [PMID: 31050013 DOI: 10.1002/jor.24338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri.,Center of Regenerative Medicine, Washington University, St. Louis, Missouri
| | - Johnny Huard
- Steadman Philippon Research Institute, Vail, Colorado
| |
Collapse
|
34
|
Guilak F, Pferdehirt L, Ross AK, Choi YR, Collins KH, Nims RJ, Katz DB, Klimak M, Tabbaa S, Pham CT. Designer Stem Cells: Genome Engineering and the Next Generation of Cell-Based Therapies. J Orthop Res 2019; 37:1287-1293. [PMID: 30977548 PMCID: PMC6546536 DOI: 10.1002/jor.24304] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/14/2019] [Accepted: 03/15/2019] [Indexed: 02/04/2023]
Abstract
Stem cells provide tremendous promise for the development of new therapeutic approaches for musculoskeletal conditions. In addition to their multipotency, certain types of stem cells exhibit immunomodulatory effects that can mitigate inflammation and enhance tissue repair. However, the translation of stem cell therapies to clinical practice has proven difficult due to challenges in intradonor and interdonor variability, engraftment, variability in recipient microenvironment and patient indications, and limited therapeutic biological activity. In this regard, the success of stem cell-based therapies may benefit from cellular engineering approaches to enhance factors such as purification, homing and cell survival, trophic effects, or immunomodulatory signaling. By combining recent advances in gene editing, synthetic biology, and tissue engineering, the potential exists to create new classes of "designer" cells that have prescribed cell-surface molecules and receptors as well as synthetic gene circuits that provide for autoregulated drug delivery or enhanced tissue repair. Published by Wiley Periodicals, Inc. J Orthop Res 37:1287-1293, 2019.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110,Correspondence: Farshid Guilak, Ph.D. Center of Regenerative Medicine, Washington University, St. Louis, Campus Box 8233, McKinley Research Bldg, Room 3121, St. Louis, MO 63110-1624.
| | - Lara Pferdehirt
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Alison K. Ross
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Robert J. Nims
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110
| | - Dakota B. Katz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | - Molly Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110,Shriners Hospitals for Children – St. Louis, St. Louis, MO 63110,Department of Biomedical Engineering, Washington University, St. Louis, MO 63110
| | | | - Christine T.N. Pham
- Division of Rheumatology, Department of Medicine, Washington University in St. Louis, MO, 63110
| |
Collapse
|
35
|
Sacchetti B. Post-natal "mesenchymal" stem cells: the assayable skeletal potency. J Stem Cells Regen Med 2019; 15:12-15. [PMID: 31239607 PMCID: PMC6586765 DOI: 10.46582/jsrm.1501004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/19/2018] [Indexed: 10/13/2023]
Affiliation(s)
- Benedetto Sacchetti
- Department of Science, University ROMA TRE, Viale Marconi, 446 - 00146 Rome, Italy
| |
Collapse
|
36
|
Kim S, Kim TM. Generation of mesenchymal stem-like cells for producing extracellular vesicles. World J Stem Cells 2019; 11:270-280. [PMID: 31171955 PMCID: PMC6545523 DOI: 10.4252/wjsc.v11.i5.270] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/02/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitor cells with therapeutic potential against autoimmune diseases, inflammation, ischemia, and metabolic disorders. Contrary to the previous conceptions, recent studies have revealed that the tissue repair and immunomodulatory functions of MSCs are largely attributed to their secretome, rather than their potential to differentiate into desired cell types. The composition of MSC secretome encompasses cytokines and growth factors, in addition to the cell-derived structures known as extracellular vesicles (EVs). EVs are membrane-enclosed nanoparticles that are capable of delivering biomolecules, and it is now believed that MSC-derived EVs are the major players that induce biological changes in the target tissues. Based on these EVs’ characteristics, the potential of EVs derived from MSC (MSC-EV) in terms of tissue regeneration and immune modulation has grown during the last decade. However, the use of MSCs for producing sufficient amount of EVs has not been satisfactory due to limitations in the cell growth and large variations among the donor cell types. In this regard, pluripotent stem cells (PSCs)-derived MSC-like cells, which can be robustly induced and expanded in vitro, have emerged as more accessible cell source that can overcome current limitations of using MSCs for EV production. In this review, we have highlighted the methods of generating MSC-like cells from PSCs and their therapeutic outcome in preclinical studies. Finally, we have also discussed future requirements for making this cell-free therapy clinically feasible.
Collapse
Affiliation(s)
- Soo Kim
- Brexogen Research Center, Brexogen Inc., Seoul, Songpa-gu 05718, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Gangwon-do, Pyeongchang 25354, South Korea
| |
Collapse
|
37
|
TRPV4-mediated calcium signaling in mesenchymal stem cells regulates aligned collagen matrix formation and vinculin tension. Proc Natl Acad Sci U S A 2019; 116:1992-1997. [PMID: 30674675 DOI: 10.1073/pnas.1811095116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Microarchitectural cues drive aligned fibrillar collagen deposition in vivo and in biomaterial scaffolds, but the cell-signaling events that underlie this process are not well understood. Utilizing a multicellular patterning model system that allows for observation of intracellular signaling events during collagen matrix assembly, we investigated the role of calcium (Ca2+) signaling in human mesenchymal stem cells (MSCs) during this process. We observed spontaneous Ca2+ oscillations in MSCs during fibrillar collagen assembly, and hypothesized that the transient receptor potential vanilloid 4 (TRPV4) ion channel, a mechanosensitive Ca2+-permeable channel, may regulate this signaling. Inhibition of TRPV4 nearly abolished Ca2+ signaling at initial stages of collagen matrix assembly, while at later times had reduced but significant effects. Importantly, blocking TRPV4 activity dramatically reduced aligned collagen fibril assembly; conversely, activating TRPV4 accelerated aligned collagen formation. TRPV4-dependent Ca2+ oscillations were found to be independent of pattern shape or subpattern cell location, suggesting this signaling mechanism is necessary for aligned collagen formation but not sufficient in the absence of physical (microarchitectural) cues that force multicellular alignment. As cell-generated mechanical forces are known to be critical to the matrix assembly process, we examined the role of TRPV4-mediated Ca2+ signaling in force generated across the load-bearing focal adhesion protein vinculin within MSCs using an FRET-based tension sensor. Inhibiting TRPV4 decreased tensile force across vinculin, whereas TRPV4 activation caused a dynamic unloading and reloading of vinculin. Together, these findings suggest TRPV4 activity regulates forces at cell-matrix adhesions and is critical to aligned collagen matrix assembly by MSCs.
Collapse
|
38
|
Duan Y, Zeng L, Zheng C, Song B, Li F, Kong X, Xu K. Inflammatory Links Between High Fat Diets and Diseases. Front Immunol 2018; 9:2649. [PMID: 30483273 PMCID: PMC6243058 DOI: 10.3389/fimmu.2018.02649] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, chronic overnutrition, such as consumption of a high-fat diet (HFD), has been increasingly viewed as a significant modifiable risk factor for diseases such as diabetes and certain types of cancer. However, the mechanisms by which HFDs exert adverse effects on human health remains poorly understood. Here, this paper will review the recent scientific literature about HFD-induced inflammation and subsequent development of diseases and cancer, with an emphasis on mechanisms involved. Given the expanding global epidemic of excessive HFD intake, understanding the impacts of a HFD on these medical conditions, gaining great insights into possible underlying mechanisms, and developing effective therapeutic strategies are of great importance.
Collapse
Affiliation(s)
- Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Liming Zeng
- Science College of Jiangxi Agricultural University, Nanchang, China
| | - Changbing Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Bo Song
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Xiangfeng Kong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Kang Xu
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| |
Collapse
|
39
|
Jiang A, Chen Y, Shi L, Li F. Differentiation of brown adipose-derived stem cells into cardiomyocyte-like cells is regulated by a combination of low 5-azacytidine concentration and bone morphogenetic protein 4. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:5514-5524. [PMID: 31949639 PMCID: PMC6963047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 09/27/2018] [Indexed: 06/10/2023]
Abstract
Adipose-derived stem cells (ADSCs) could be an ideal candidate for seed cells to regenerate damaged heart tissue. This study examined and compared the cardio-myogenic differentiation efficacy of neonatal rat brown ADSCs (rbADSCs) treated with either 5-azacytidine (5-AZA), bone morphogenetic protein 4 (BMP4), or lower doses of both molecules. Briefly, by investigating the protein expression of cardiac-specific markers (i.e., cardiac troponin-I, α-sarcomeric actinin, sarcoplasmic reticulum Ca2+-ATPase, and connexin 43), our data indicated that rbADSCs could be differentiated into cardiomyocyte-like cells by all three treatments. By quantitatively measuring the number of cells with positive staining for the above markers, we found that the low-dose combined treatment showed higher differentiation efficiency compared to standard dose 5-AZA and BMP4 treatment. Similarly, the expression levels of these proteins as determined by western blotting were higher in the low-dose combination group than in the standard dose 5-AZA and BMP4 groups. Also, the combined strategy maintained the decreased cell viability caused by cytotoxicity of 5-AZA, probably through reducing the ratio of apoptotic rbADSCs. Furthermore, the extracellular regulated protein kinase (ERK) signaling pathways participate in the differentiation process, but the observed effects between the BMP4 and 5-Aza treatments are quite different.
Collapse
Affiliation(s)
- Aixia Jiang
- Department of Cardiology, Shanghai Jiao Tong University School of Medicine-Affiliated Shanghai Children's Medical Centre Shanghai, China
| | - Yiwei Chen
- Department of Cardiology, Shanghai Jiao Tong University School of Medicine-Affiliated Shanghai Children's Medical Centre Shanghai, China
| | - Lin Shi
- Department of Cardiology, Shanghai Jiao Tong University School of Medicine-Affiliated Shanghai Children's Medical Centre Shanghai, China
| | - Fen Li
- Department of Cardiology, Shanghai Jiao Tong University School of Medicine-Affiliated Shanghai Children's Medical Centre Shanghai, China
| |
Collapse
|
40
|
Adkar SS, Wu CL, Willard VP, Dicks A, Ettyreddy A, Steward N, Bhutani N, Gersbach CA, Guilak F. Step-Wise Chondrogenesis of Human Induced Pluripotent Stem Cells and Purification Via a Reporter Allele Generated by CRISPR-Cas9 Genome Editing. Stem Cells 2018; 37:65-76. [PMID: 30378731 DOI: 10.1002/stem.2931] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/26/2018] [Accepted: 09/05/2018] [Indexed: 01/23/2023]
Abstract
The differentiation of human induced pluripotent stem cells (hiPSCs) to prescribed cell fates enables the engineering of patient-specific tissue types, such as hyaline cartilage, for applications in regenerative medicine, disease modeling, and drug screening. In many cases, however, these differentiation approaches are poorly controlled and generate heterogeneous cell populations. Here, we demonstrate cartilaginous matrix production in three unique hiPSC lines using a robust and reproducible differentiation protocol. To purify chondroprogenitors (CPs) produced by this protocol, we engineered a COL2A1-GFP knock-in reporter hiPSC line by CRISPR-Cas9 genome editing. Purified CPs demonstrated an improved chondrogenic capacity compared with unselected populations. The ability to enrich for CPs and generate homogenous matrix without contaminating cell types will be essential for regenerative and disease modeling applications. Stem Cells 2019;37:65-76.
Collapse
Affiliation(s)
- Shaunak S Adkar
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA
| | | | - Amanda Dicks
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Adarsh Ettyreddy
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA
| | - Nidhi Bhutani
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA.,Cytex Therapeutics, Inc., Durham, North Carolina, USA
| |
Collapse
|
41
|
Moffat KL, Goon K, Moutos FT, Estes BT, Oswald SJ, Zhao X, Guilak F. Composite Cellularized Structures Created from an Interpenetrating Polymer Network Hydrogel Reinforced by a 3D Woven Scaffold. Macromol Biosci 2018; 18:e1800140. [PMID: 30040175 PMCID: PMC6687075 DOI: 10.1002/mabi.201800140] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/21/2018] [Indexed: 11/10/2022]
Abstract
Biomaterial scaffolds play multiple roles in cartilage tissue engineering, including controlling architecture of newly formed tissue while facilitating growth of embedded cells and simultaneously providing functional properties to withstand the mechanical environment within the native joint. In particular, hydrogels-with high water content and desirable transport properties-while highly conducive to chondrogenesis, often lack functional mechanical properties. In this regard, interpenetrating polymer network (IPN) hydrogels can provide mechanical toughness greatly exceeding that of individual components; however, many IPN materials are not biocompatible for cell encapsulation. In this study, an agarose and poly(ethylene) glycol IPN hydrogel is seeded with human mesenchymal stem cells (MSCs). Results show high viability of MSCs within the IPN hydrogel, with improved mechanical properties compared to constructs comprised of individual components. These properties are further strengthened by integrating the hydrogel with a 3D woven structure. The resulting fiber-reinforced hydrogels display functional macroscopic mechanical properties mimicking those of native articular cartilage, while providing a local microenvironment that supports cellular viability and function. These findings suggest that a fiber-reinforced IPN hydrogel can support stem cell chondrogenesis while allowing for significantly enhanced, complex mechanical properties at multiple scales as compared to individual hydrogel or fiber components.
Collapse
Affiliation(s)
- Kristen L Moffat
- Center of Regenerative Medicine, Washington University, Campus Box 8233, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children, St. Louis, MO, 63110, USA
| | - Kelsey Goon
- Center of Regenerative Medicine, Washington University, Campus Box 8233, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children, St. Louis, MO, 63110, USA
| | | | | | - Sara J Oswald
- Center of Regenerative Medicine, Washington University, Campus Box 8233, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children, St. Louis, MO, 63110, USA
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Farshid Guilak
- Center of Regenerative Medicine, Washington University, Campus Box 8233, St. Louis, MO, 63110, USA
- Shriners Hospitals for Children, St. Louis, MO, 63110, USA
- Cytex Therapeutics, Inc., Durham, NC, 27704, USA
| |
Collapse
|
42
|
Mehta S, McClarren B, Aijaz A, Chalaby R, Cook-Chennault K, Olabisi RM. The effect of low-magnitude, high-frequency vibration on poly(ethylene glycol)-microencapsulated mesenchymal stem cells. J Tissue Eng 2018; 9:2041731418800101. [PMID: 30245801 PMCID: PMC6146326 DOI: 10.1177/2041731418800101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
Low-magnitude, high-frequency vibration has stimulated osteogenesis in mesenchymal stem cells when these cells were cultured in certain types of three-dimensional environments. However, results of osteogenesis are conflicting with some reports showing no effect of vibration at all. A large number of vibration studies using three-dimensional scaffolds employ scaffolds derived from natural sources. Since these natural sources potentially have inherent biochemical and microarchitectural cues, we explored the effect of low-magnitude, high-frequency vibration at low, medium, and high accelerations when mesenchymal stem cells were encapsulated in poly(ethylene glycol) diacrylate microspheres. Low and medium accelerations enhanced osteogenesis in mesenchymal stem cells while high accelerations inhibited it. These studies demonstrate that the isolated effect of vibration alone induces osteogenesis.
Collapse
Affiliation(s)
- Sneha Mehta
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Brooke McClarren
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Ayesha Aijaz
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Rabab Chalaby
- Department of Materials Science and Engineering, Rutgers University, Piscataway, NJ, USA
| | | | - Ronke M Olabisi
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
43
|
Huynh NPT, Brunger JM, Gloss CC, Moutos FT, Gersbach CA, Guilak F. Genetic Engineering of Mesenchymal Stem Cells for Differential Matrix Deposition on 3D Woven Scaffolds. Tissue Eng Part A 2018; 24:1531-1544. [PMID: 29756533 DOI: 10.1089/ten.tea.2017.0510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Tissue engineering approaches for the repair of osteochondral defects using biomaterial scaffolds and stem cells have remained challenging due to the inherent complexities of inducing cartilage-like matrix and bone-like matrix within the same local environment. Members of the transforming growth factor β (TGFβ) family have been extensively utilized in the engineering of skeletal tissues, but have distinct effects on chondrogenic and osteogenic differentiation of progenitor cells. The goal of this study was to develop a method to direct human bone marrow-derived mesenchymal stem cells (MSCs) to deposit either mineralized matrix or a cartilaginous matrix rich in glycosaminoglycan and type II collagen within the same biochemical environment. This differential induction was performed by culturing cells on engineered three-dimensionally woven poly(ɛ-caprolactone) (PCL) scaffolds in a chondrogenic environment for cartilage-like matrix production while inhibiting TGFβ3 signaling through Mothers against DPP homolog 3 (SMAD3) knockdown, in combination with overexpressing RUNX2, to achieve mineralization. The highest levels of mineral deposition and alkaline phosphatase activity were observed on scaffolds with genetically engineered MSCs and exhibited a synergistic effect in response to SMAD3 knockdown and RUNX2 expression. Meanwhile, unmodified MSCs on PCL scaffolds exhibited accumulation of an extracellular matrix rich in glycosaminoglycan and type II collagen in the same biochemical environment. This ability to derive differential matrix deposition in a single culture condition opens new avenues for developing complex tissue replacements for chondral or osteochondral defects.
Collapse
Affiliation(s)
- Nguyen P T Huynh
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri.,3 Department of Cell Biology, Duke University , Durham, North Carolina
| | | | - Catherine C Gloss
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri
| | | | - Charles A Gersbach
- 6 Department of Biomedical Engineering, Duke University , Durham, North Carolina
| | - Farshid Guilak
- 1 Department of Orthopaedic Surgery, Washington University in Saint Louis , Saint Louis, Missouri.,2 Shriners Hospitals for Children-St. Louis , St. Louis, Missouri.,5 Cytex Therapeutics, Inc. , Durham, North Carolina
| |
Collapse
|
44
|
Milner DJ, Bionaz M, Monaco E, Cameron JA, Wheeler MB. Myogenic potential of mesenchymal stem cells isolated from porcine adipose tissue. Cell Tissue Res 2018; 372:507-522. [PMID: 29318389 DOI: 10.1007/s00441-017-2764-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/02/2017] [Indexed: 12/31/2022]
Abstract
Advances in stem cell biology and materials science have provided a basis for developing tissue engineering methods to repair muscle injury. Among stem cell populations with potential to aid muscle repair, adipose-derived mesenchymal stem cells (ASC) hold great promise. To evaluate the possibility of using porcine ASC for muscle regeneration studies, we co-cultured porcine ASC with murine C2C12 myoblasts. These experiments demonstrated that porcine ASC display significant myogenic potential. Co-culture of ASC expressing green fluorescent protein (GFP) with C2C12 cells resulted in GFP+ myotube formation, indicating fusion of ASC with myoblasts to form myotubes. The presence of porcine lamin A/C positive nuclei in myotubes and RTqPCR analysis of porcine myogenin and desmin expression confirmed that myotube nuclei derived from ASC contribute to muscle gene expression. Co-culturing GFP+ASC with porcine satellite cells demonstrated enhanced myogenic capability of ASC, as the percentage of labeled myotubes increased compared to mouse co-cultures. Enhancing myogenic potential of ASC through soluble factor treatment or expansion of ASC with innate myogenic capacity should allow for their therapeutic use to regenerate muscle tissue lost to disease or injury.
Collapse
Affiliation(s)
- Derek J Milner
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Massimo Bionaz
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Elisa Monaco
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jo Ann Cameron
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Matthew B Wheeler
- Carl R. Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA.
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
45
|
McClarren B, Olabisi R. Strain and Vibration in Mesenchymal Stem Cells. Int J Biomater 2018; 2018:8686794. [PMID: 29545825 PMCID: PMC5818976 DOI: 10.1155/2018/8686794] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells capable of differentiating into any mesenchymal tissue, including bone, cartilage, muscle, and fat. MSC differentiation can be influenced by a variety of stimuli, including environmental and mechanical stimulation, scaffold physical properties, or applied loads. Numerous studies have evaluated the effects of vibration or cyclic tensile strain on MSCs towards developing a mechanically based method of differentiation, but there is no consensus between studies and each investigation uses different culture conditions, which also influence MSC fate. Here we present an overview of the response of MSCs to vibration and cyclic tension, focusing on the effect of various culture conditions and strain or vibration parameters. Our review reveals that scaffold type (e.g., natural versus synthetic; 2D versus 3D) can influence cell response to vibration and strain to the same degree as loading parameters. Hence, in the efforts to use mechanical loading as a reliable method to differentiate cells, scaffold selection is as important as method of loading.
Collapse
Affiliation(s)
- Brooke McClarren
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Ronke Olabisi
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| |
Collapse
|
46
|
Renata S. [Advanced medicinal products medical therapy based on mesenchymal stem cells]. FARMACJA POLSKA 2018; 74:178-183. [PMID: 29674780 PMCID: PMC5903288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
Recent developments in the field of cell therapy and regenerative medicine describe mesenchymal stem cells (MSCs) as potential biological products due to their ability to self-renew and differentiate. MSCs are multipotent cells with immunomodulatory and regenerative properties. Because of their therapeutic potential, they are extensively studied to assess their viability, safety and efficacy. This chapter describes the main characteristics and sources of MSC cells, as well as their properties and current clinical applications. It also presents the latest information on the regulatory aspects that define them as somatic cell therapy medicinal products. MSC-based cell therapy is currently offered as a pharmacological alternative, although we still face many challenges in this area.
Collapse
Affiliation(s)
- Szydlak Renata
- Katedra Biochemii Lekarskiej, Wydział Lekarski, Uniwersytet Jagielloński Collegium Medicum, Kraków
| |
Collapse
|
47
|
Yu F, Shen H, Deng HW. Systemic analysis of osteoblast-specific DNA methylation marks reveals novel epigenetic basis of osteoblast differentiation. Bone Rep 2017; 6:109-119. [PMID: 28409176 PMCID: PMC5384298 DOI: 10.1016/j.bonr.2017.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/20/2017] [Accepted: 04/01/2017] [Indexed: 01/21/2023] Open
Abstract
DNA methylation is an important epigenetic modification that contributes to the lineage commitment and specific functions of different cell types. In this study, we compared ENCODE-generated genome-wide DNA methylation profiles of human osteoblast with 21 other types of human cells in order to identify osteoblast-specific methylation events. For most of the cell strains, data from two isogenic replicates were included, resulting in a total of 51 DNA methylation datasets. We identified 852 significant osteoblast-specific differentially methylated CpGs (DMCs) and 295 significant differentially methylated regions (DMRs). Significant DMCs/DMRs were not enriched in CpG islands (CGIs) and promoters, but more strongly enriched in CGI shores/shelves and in gene body and intergenic regions. The genes associated with significant DMRs were highly enriched in biological processes related to transcriptional regulation and critical for regulating bone metabolism and skeletal development under physiologic and pathologic conditions. By integrating the DMR data with the extensive gene expression and chromatin epigenomics data, we observed complex, context-dependent relationships between DNA methylation, chromatin states, and gene expression, suggesting diverse DNA methylation-mediated regulatory mechanisms. Our results also highlighted a number of novel osteoblast-relevant genes. For example, the integrated evidences from DMR analysis, histone modification and RNA-seq data strongly support that there is a novel isoform of neurexin-2 (NRXN2) gene specifically expressed in osteoblast. NRXN2 was known to function as a cell adhesion molecule in the vertebrate nervous system, but its functional role in bone is completely unknown and thus worth further investigation. In summary, we reported a comprehensive analysis of osteoblast-specific DNA methylation profiles and revealed novel insights into the epigenetic basis of osteoblast differentiation and activity.
Collapse
Affiliation(s)
- Fangtang Yu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hui Shen
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hong-Wen Deng
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
- College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
48
|
From Human Mesenchymal Stem Cells to Insulin-Producing Cells: Comparison between Bone Marrow- and Adipose Tissue-Derived Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3854232. [PMID: 28584815 PMCID: PMC5444016 DOI: 10.1155/2017/3854232] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/18/2017] [Accepted: 04/23/2017] [Indexed: 12/24/2022]
Abstract
The aim of this study is to compare human bone marrow-derived mesenchymal stem cells (BM-MSCs) and adipose tissue-derived mesenchymal stem cells (AT-MSCs), for their differentiation potentials to form insulin-producing cells. BM-MSCs were obtained during elective orthotopic surgery and AT-MSCs from fatty aspirates during elective cosmetics procedures. Following their expansion, cells were characterized by phenotyping, trilineage differentiation ability, and basal gene expression of pluripotency genes and for their metabolic characteristics. Cells were differentiated according to a Trichostatin-A based protocol. The differentiated cells were evaluated by immunocytochemistry staining for insulin and c-peptide. In addition the expression of relevant pancreatic endocrine genes was determined. The release of insulin and c-peptide in response to a glucose challenge was also quantitated. There were some differences in basal gene expression and metabolic characteristics. After differentiation the proportion of the resulting insulin-producing cells (IPCs), was comparable among both cell sources. Again, there were no differences neither in the levels of gene expression nor in the amounts of insulin and c-peptide release as a function of glucose challenge. The properties, availability, and abundance of AT-MSCs render them well-suited for applications in regenerative medicine. Conclusion. BM-MSCs and AT-MSCs are comparable regarding their differential potential to form IPCs. The availability and properties of AT-MSCs render them well-suited for applications in regenerative medicine.
Collapse
|
49
|
Zhang AZ, Ficklscherer A, Gülecyüz MF, Paulus AC, Niethammer TR, Jansson V, Müller PE. Cell Toxicity in Fibroblasts, Tenocytes, and Human Mesenchymal Stem Cells-A Comparison of Necrosis and Apoptosis-Inducing Ability in Ropivacaine, Bupivacaine, and Triamcinolone. Arthroscopy 2017; 33:840-848. [PMID: 28089496 DOI: 10.1016/j.arthro.2016.10.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/19/2016] [Accepted: 10/24/2016] [Indexed: 02/02/2023]
Abstract
PURPOSE To analyze the ability of ropivacaine, bupivacaine, and triamcinolone to induce apoptosis and necrosis in fibroblasts, tenocytes, and human mesenchymal stem cells. METHODS Human dermal fibroblasts, adipose-derived human mesenchymal stem cells (hMSCs), and tenocytes gained from the rotator cuff tendon were seeded with a cell density of 0.5 × 104/cm2. One specimen of ropivacaine, bupivacaine, and triamcinolone was tested separately on the cells with separate concentrations of 0.5%, 0.25%, and 0.125% for each specimen. The negative control received no agent, only a change of medium. The incubation period for each agent was 30 minutes. After a change of medium and 1 hour, 24 hours, and 7 days of incubation, 104 cells were harvested and analyzed via fluorescence-activated cell sorting with double-staining with annexin V and propidium iodide. Statistical analysis to determine significant difference (P < .05) between the groups with SPSS statistics 23 through one-way analysis of variance with a univariate general linear model was performed. RESULTS Bupivacaine showed necrosis-inducing effects on fibroblasts and tenocytes, with the necrotic effect peaking at 0.5% and 0.25%. Ropivacaine and triamcinolone caused no significant necrosis. Compared with fibroblasts and tenocytes, hMSCs did not show significant necrotic or apoptotic effects after exposure to bupivacaine. Overall, no significant differences in apoptosis were detected between different cell lines, varying concentrations, or time measurements. CONCLUSIONS Bupivacaine 0.5% and 0.25% have the most necrosis-inducing effects on fibroblasts and tenocytes. Ropivacaine caused less necrosis than bupivaine. Compared with fibroblasts and tenocytes, hMSCs were not affected by necrosis using any of the tested agents. A significant apoptosis-inducing effect could not be detected for the different cell lines. CLINICAL RELEVANCE Possible cell toxicity raises questions of concern for intra-articular injections using local anesthetics and corticosteroids. The present study demonstrates the necrotic and apoptotic effects of ropivacaine, bupivacaine, and triamcinolone and may give recommendations for intra-articular use of local anesthetics and corticosteroids.
Collapse
Affiliation(s)
- Anja Z Zhang
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich, Munich, Germany.
| | | | - Mehmet F Gülecyüz
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich, Munich, Germany
| | - Alexander C Paulus
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich, Munich, Germany
| | - Thomas R Niethammer
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich, Munich, Germany
| | - Volkmar Jansson
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich, Munich, Germany
| | - Peter E Müller
- Department of Orthopaedic Surgery, Physical Medicine and Rehabilitation, University Hospital of Munich, Munich, Germany
| |
Collapse
|
50
|
Liew LC, Katsuda T, Gailhouste L, Nakagama H, Ochiya T. Mesenchymal stem cell-derived extracellular vesicles: a glimmer of hope in treating Alzheimer’s disease. Int Immunol 2017; 29:11-19. [DOI: 10.1093/intimm/dxx002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 01/13/2017] [Indexed: 12/18/2022] Open
Affiliation(s)
- Lee Chuen Liew
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Department of Pathology, Immunology and Microbiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyou-ku, Tokyo 113-0033, Japan
| | - Takeshi Katsuda
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Luc Gailhouste
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hitoshi Nakagama
- Department of Pathology, Immunology and Microbiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyou-ku, Tokyo 113-0033, Japan
- National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|