1
|
Guillaumin S, Rossoni A, Zeugolis D. State-of the-art and future perspective in co-culture systems for tendon engineering. BIOMATERIALS AND BIOSYSTEMS 2025; 17:100110. [PMID: 40130022 PMCID: PMC11932666 DOI: 10.1016/j.bbiosy.2025.100110] [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: 08/08/2024] [Revised: 11/05/2024] [Accepted: 03/04/2025] [Indexed: 03/26/2025] Open
Abstract
Tendon is a connective tissue that links bone to muscle, allowing for maintenance of skeleton posture, joint movement, energy storage and transmission of muscle force to bone. Tendon is a hypocellular and hypovascular tissue of poor self-regeneration capacity. Current surgical treatments are of limited success, frequently resulting in reinjury. Upcoming cell therapies are primarily based on tenocytes, a cell population of limited self-renewal capacity in vitro or mesenchymal stromal cells, a cell population prone to ectopic bone formation in vivo. Over the years mono- or multi- factorial cell culture technologies have failed to effectively maintain tenocyte phenotype in culture during expansion or to prime mesenchymal stromal cells towards tenogenic lineage prior to implantation. Upon these limitations the concept of co-culture was conceived. Here, we comprehensively review and discuss tenogenic differentiation of mesenchymal stromal cells through direct or indirect culture with tenocytes in an attempt to generate a tenocyte or a tendon-like cell population for regenerative medicine purposes.
Collapse
Affiliation(s)
- Salomé Guillaumin
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Dimitrios Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
2
|
de Sousa Moreira A, Lopes B, Sousa AC, Coelho A, Sousa P, Araújo A, Delgado E, Alvites R, Maurício AC. Stem Cell-Based Therapies for Glaucoma Treatment: A Review Bridging the Gap in Veterinary Patients. Int J Mol Sci 2024; 26:232. [PMID: 39796087 PMCID: PMC11719664 DOI: 10.3390/ijms26010232] [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: 11/12/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Retinal diseases are characterized by progressive damage to retinal cells, leading to irreversible vision loss. Among these, glaucoma stands out as a multifactorial neurodegenerative disease involving elevated intraocular pressure, retinal ganglion cell apoptosis, and optic nerve damage, ultimately resulting in blindness in both humans and dogs. Stem cell-based therapies have emerged as a promising therapeutic option for such conditions due to their regenerative and neuroprotective potential. These therapies, particularly those based on mesenchymal stem cells, offer the potential to repair and protect retinal tissues through the bioactive molecules (growth factors, cytokines, chemokines) secreted, their secretome. However, research in this field, especially on the use of umbilical cord mesenchymal stem cells' secretome, remains sparse. Most clinical trials focus on human glaucomatous patients, leaving a significant gap in veterinary patients' application, especially in dogs, with additional research being needed to determine its usefulness in canine glaucoma treatment. Future studies should aim to evaluate these therapies across both human and veterinary contexts, broadening treatment possibilities for glaucoma.
Collapse
Affiliation(s)
- Alícia de Sousa Moreira
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - André Coelho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Ana Araújo
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| | - Esmeralda Delgado
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
- Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
- Instituto Universitário de Ciências da Saúde (CESPU), Avenida Central de Gandra n° 1317, 4585-116 Paredes, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto (UP), Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (A.d.S.M.); (B.L.); (A.C.S.); (A.C.); (P.S.); (R.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculdade de Medicina Veterinária (FMV), Universidade de Lisboa (UL), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal;
| |
Collapse
|
3
|
Kim YS, Lupatov AY, Burunova VV, Bagmet NN, Chardarov NK, Malov SL, Kholodenko RV, Shatverian GA, Manukyan GV, Yarygin KN, Kholodenko IV. Human Liver MSCs Retain Their Basic Cellular Properties in Chronically Inflamed Liver Tissue. Int J Mol Sci 2024; 25:13374. [PMID: 39769138 PMCID: PMC11676302 DOI: 10.3390/ijms252413374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Every 25th death worldwide is associated with liver pathology. The development of novel approaches to liver diseases therapy and protocols for maintaining the vital functions of patients on the liver transplant waiting list are urgently needed. Resident mesenchymal stem cells (MSCs) play a significant role in supporting liver tissue integrity and improve the liver condition after infusion. However, it remains unclear whether MSCs isolated from chronically inflamed livers are similar in their basic cellular properties to MSCs obtained from healthy livers. We applied a large array of tests to compare resident MSCs isolated from apparently normal liver tissue and from chronically inflamed livers of patients with fibrosis, cirrhosis, and viral hepatitis. Chronic inflammatory environment did not alter the major cellular characteristics of MSCs, including the expression of MSC markers, stem cell markers, adhesion molecules, and the hallmarks of senescence, as well as cell proliferation, migration, and secretome. Only the expression of some immune checkpoints and toll-like receptors was different. Evidently, MSCs with unchanged cellular properties are present in human liver even at late stages of inflammatory diseases. These cells can be isolated and used as starting material in the development of cell therapies of liver diseases.
Collapse
Affiliation(s)
- Yan S. Kim
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Alexey Yu. Lupatov
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Veronika V. Burunova
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Nikolay N. Bagmet
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Nikita K. Chardarov
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Svyatoslav L. Malov
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Roman V. Kholodenko
- Laboratory of Molecular Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Garnik A. Shatverian
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Garik V. Manukyan
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
- Department of General Pathology and Pathophysiology, Russian Medical Academy of Continuous Professional Education, 125284 Moscow, Russia
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| |
Collapse
|
4
|
Aghazadeh S, Peng Q, Dardmeh F, Hjortdal JØ, Zachar V, Alipour H. Immunophenotypical Characterization of Limbal Mesenchymal Stromal Cell Subsets during In Vitro Expansion. Int J Mol Sci 2024; 25:8684. [PMID: 39201371 PMCID: PMC11354999 DOI: 10.3390/ijms25168684] [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: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Limbal mesenchymal stromal cells (LMSCs) reside in the limbal niche, supporting corneal integrity and facilitating regeneration. While mesenchymal stem/stromal cells (MSCs) are used in regenerative therapies, there is limited knowledge about LMSC subpopulations and their characteristics. This study characterized human LMSC subpopulations through the flow cytometric assessment of fifteen cell surface markers, including MSC, wound healing, immune regulation, ASC, endothelial, and differentiation markers. Primary LMSCs were established from remnant human corneal transplant specimens and passaged eight times to observe changes during subculture. The results showed the consistent expression of typical MSC markers and distinct subpopulations with the passage-dependent expression of wound healing, immune regulation, and differentiation markers. High CD166 and CD248 expressions indicated a crucial role in ocular surface repair. CD29 expression suggested an immunoregulatory role. Comparable pigment-epithelial-derived factor (PEDF) expression supported anti-inflammatory and anti-angiogenic roles. Sustained CD201 expression indicated maintained differentiation capability, while VEGFR2 expression suggested potential endothelial differentiation. LMSCs showed higher VEGF expression than fibroblasts and endothelial cells, suggesting a potential contribution to ocular surface regeneration through the modulation of angiogenesis and inflammation. These findings highlight the heterogeneity and multipotent potential of LMSC subpopulations during in vitro expansion, informing the development of standardized protocols for regenerative therapies and improving treatments for ocular surface disorders.
Collapse
Affiliation(s)
- Sara Aghazadeh
- Regenerative Medicine, Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.A.); (Q.P.); (F.D.); (V.Z.)
| | - Qiuyue Peng
- Regenerative Medicine, Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.A.); (Q.P.); (F.D.); (V.Z.)
| | - Fereshteh Dardmeh
- Regenerative Medicine, Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.A.); (Q.P.); (F.D.); (V.Z.)
| | | | - Vladimir Zachar
- Regenerative Medicine, Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.A.); (Q.P.); (F.D.); (V.Z.)
| | - Hiva Alipour
- Regenerative Medicine, Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.A.); (Q.P.); (F.D.); (V.Z.)
| |
Collapse
|
5
|
Milián L, Molina P, Oliver-Ferrándiz M, Fernández-Sellers C, Monzó A, Sánchez-Sánchez R, Braza-Boils A, Mata M, Zorio E. Cadaveric Adipose-Derived Stem Cells for Regenerative Medicine and Research. Int J Mol Sci 2023; 24:15696. [PMID: 37958680 PMCID: PMC10647636 DOI: 10.3390/ijms242115696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Advances in regenerative medicine have enabled the search for new solutions to current health problems in so far unexplored fields. Thus, we focused on cadaveric subcutaneous fat as a promising source of adipose-derived stem cells (ADSCs) that have potential to differentiate into different cell lines. With this aim, we isolated and characterized ADSCs from cadaveric samples with a postmortem interval ranging from 30 to 55 h and evaluated their ability to differentiate into chondrocytes or osteocytes. A commercial ADSC line was used as reference. Morphological and protein expression analyses were used to confirm the final stage of differentiation. Eight out of fourteen samples from patients were suitable to complete the whole protocol. Cadaveric ADSCs exhibited features of stem cells based upon several markers: CD29 (84.49 ± 14.07%), CD105 (94.38 ± 2.09%), and CD44 (99.77 ± 0.32%). The multiparametric assessment of differentiation confirmed the generation of stable lines of chondrocytes and osteocytes. In conclusion, we provide evidence supporting the feasibility of obtaining viable postmortem human subcutaneous fat ADSCs with potential application in tissue engineering and research fields.
Collapse
Affiliation(s)
- Lara Milián
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (L.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Pilar Molina
- Department of Pathology, Instituto de Medicina Legal y Ciencias Forenses, 46010 Valencia, Spain; (P.M.); (C.F.-S.); (A.M.)
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - María Oliver-Ferrándiz
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (L.M.)
| | - Carlos Fernández-Sellers
- Department of Pathology, Instituto de Medicina Legal y Ciencias Forenses, 46010 Valencia, Spain; (P.M.); (C.F.-S.); (A.M.)
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - Ana Monzó
- Department of Pathology, Instituto de Medicina Legal y Ciencias Forenses, 46010 Valencia, Spain; (P.M.); (C.F.-S.); (A.M.)
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - Rafael Sánchez-Sánchez
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
| | - Aitana Braza-Boils
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
- CIBERCV, Center for Biomedical Network Research on Cardiovascular Diseases, 28015 Madrid, Spain
| | - Manuel Mata
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (L.M.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Esther Zorio
- CAFAMUSME Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (R.S.-S.); (A.B.-B.); (E.Z.)
- CIBERCV, Center for Biomedical Network Research on Cardiovascular Diseases, 28015 Madrid, Spain
- Inherited Cardiac Diseases Unit, Cardiology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Department of Medicine, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain
| |
Collapse
|
6
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
7
|
Fonseca LN, Bolívar-Moná S, Agudelo T, Beltrán LD, Camargo D, Correa N, Del Castillo MA, Fernández de Castro S, Fula V, García G, Guarnizo N, Lugo V, Martínez LM, Melgar V, Peña MC, Pérez WA, Rodríguez N, Pinzón A, Albarracín SL, Olaya M, Gutiérrez-Gómez ML. Cell surface markers for mesenchymal stem cells related to the skeletal system: A scoping review. Heliyon 2023; 9:e13464. [PMID: 36865479 PMCID: PMC9970931 DOI: 10.1016/j.heliyon.2023.e13464] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) have been described as bone marrow stromal cells, which can form cartilage, bone or hematopoietic supportive stroma. In 2006, the International Society for Cell Therapy (ISCT) established a set of minimal characteristics to define MSCs. According to their criteria, these cells must express CD73, CD90 and CD105 surface markers; however, it is now known they do not represent true stemness epitopes. The objective of the present work was to determine the surface markers for human MSCs associated with skeletal tissue reported in the literature (1994-2021). To this end, we performed a scoping review for hMSCs in axial and appendicular skeleton. Our findings determined the most widely used markers were CD105 (82.9%), CD90 (75.0%) and CD73 (52.0%) for studies performed in vitro as proposed by the ISCT, followed by CD44 (42.1%), CD166 (30.9%), CD29 (27.6%), STRO-1 (17.7%), CD146 (15.1%) and CD271 (7.9%) in bone marrow and cartilage. On the other hand, only 4% of the articles evaluated in situ cell surface markers. Even though most studies use the ISCT criteria, most publications in adult tissues don't evaluate the characteristics that establish a stem cell (self-renewal and differentiation), which will be necessary to distinguish between a stem cell and progenitor populations. Collectively, MSCs require further understanding of their characteristics if they are intended for clinical use.
Collapse
Affiliation(s)
- Luisa Nathalia Fonseca
- Master Student in Biological Sciences - School of Science, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Santiago Bolívar-Moná
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Tatiana Agudelo
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Liz Daniela Beltrán
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Daniel Camargo
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Nestor Correa
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - María Alexandra Del Castillo
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | | | - Valeria Fula
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Gabriela García
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Natalia Guarnizo
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Valentina Lugo
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Liz Mariana Martínez
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Verónica Melgar
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - María Clara Peña
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Wilfran Arbey Pérez
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Nicolás Rodríguez
- Medical Student - Stem Cell Research Group – School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Andrés Pinzón
- Department of Orthopedics and Traumatology - School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Sonia Luz Albarracín
- Department of Nutrition and Biochemistry -School of Science, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - Mercedes Olaya
- Department of Pathology - School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| | - María Lucía Gutiérrez-Gómez
- Department of Morphology - School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
- Institute of Human Genetics - School of Medicine, Pontificia Universidad Javeriana. Bogotá, Colombia
| |
Collapse
|
8
|
Matthews EZ, Lanham S, White K, Kyriazi ME, Alexaki K, El-Sagheer AH, Brown T, Kanaras AG, J West J, MacArthur BD, Stumpf PS, Oreffo ROC. Single-cell RNA-sequence analysis of human bone marrow reveals new targets for isolation of skeletal stem cells using spherical nucleic acids. J Tissue Eng 2023; 14:20417314231169375. [PMID: 37216034 PMCID: PMC10192814 DOI: 10.1177/20417314231169375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/24/2023] [Indexed: 05/24/2023] Open
Abstract
There is a wealth of data indicating human bone marrow contains skeletal stem cells (SSC) with the capacity for osteogenic, chondrogenic and adipogenic differentiation. However, current methods to isolate SSCs are restricted by the lack of a defined marker, limiting understanding of SSC fate, immunophenotype, function and clinical application. The current study applied single-cell RNA-sequencing to profile human adult bone marrow populations from 11 donors and identified novel targets for SSC enrichment. Spherical nucleic acids were used to detect these mRNA targets in SSCs. This methodology was able to rapidly isolate potential SSCs found at a frequency of <1 in 1,000,000 in human bone marrow, with the capacity for tri-lineage differentiation in vitro and ectopic bone formation in vivo. The current studies detail the development of a platform to advance SSC enrichment from human bone marrow, offering an invaluable resource for further SSC characterisation, with significant therapeutic impact therein.
Collapse
Affiliation(s)
- Elloise Z Matthews
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
| | - Stuart Lanham
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Cancer Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
| | - Kate White
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
| | - Maria-Eleni Kyriazi
- College of Engineering and Technology,
American University of the Middle East, Kuwait
| | - Konstantina Alexaki
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Afaf H El-Sagheer
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford, Oxford, UK
- Chemistry Branch, Department of Science
and Mathematics, Faculty of Petroleum and Mining Engineering, Suez University, Suez,
Egypt
| | - Tom Brown
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford, Oxford, UK
| | - Antonios G Kanaras
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
| | - Jonathan J West
- Cancer Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Ben D MacArthur
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
- Mathematical Sciences, University of
Southampton, Southampton, UK
| | - Patrick S Stumpf
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Joint Research Center for Computational
Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Richard OC Oreffo
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
- College of Biomedical Engineering,
China Medical University, Taichung, Taiwan
| |
Collapse
|
9
|
Zhang J, Zhang M, Lin R, Du Y, Wang L, Yao Q, Zannettino A, Zhang H. Chondrogenic preconditioning of mesenchymal stem/stromal cells within a magnetic scaffold for osteochondral repair. Biofabrication 2022; 14. [PMID: 35226893 DOI: 10.1088/1758-5090/ac5935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022]
Abstract
Stem cell therapy using mesenchymal stromal/stem cells (MSCs) represents a novel approach to treating severe diseases, including osteoarthritis (OA). However, the therapeutic benefit of MSCs is highly dependent on their differentiation state, which can be regulated by many factors. Herein, three-dimensional (3D) magnetic scaffolds were successfully fabricated by incorporating magnetic nanoparticles (MNPs) into electrospun gelatin nanofibers. When positioned near a rotating magnet (f= 0.5 Hz), the magnetic scaffolds with the embedded MSCs were driven upward/downward in the culture container to induce mechanical stimulation to MSCs due to spatial confinement and fluid flow. The extracellular matrix-mimicking scaffold and the alternating magnetic field significantly enhanced chondrogenesis instead of osteogenesis. Furthermore, the fibre topography could be tuned with different compositions of the coating layer on MNPs, and the topography had a significant impact on MSC differentiation. Selective up-regulation of chondrogenesis-related genes (COL2A1andACAN) was found for the magnetic scaffolds with citric acid-coated MNPs (CAG). In contrast, osteogenesis-related genes (RUNX2andSPARC) were selectively and significantly up-regulated for the magnetic scaffolds with polyvinylpyrrolidone-coated MNPs (PVPG). Prior to implantation in vivo, chondrogenic preconditioning of MSCs within the CAG scaffolds under a dynamic magnetic field resulted in superior osteochondral repair. Hence, the magnetic scaffolds together with an in-house rotating magnet device could be a novel platform to initiate multiple stimuli on stem cell differentiation for effective repair of osteochondral defects.
Collapse
Affiliation(s)
- Jiabin Zhang
- Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510275, CHINA
| | - Ming Zhang
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing , Jiangsu Province, China, Nangjing, Jiangsu, 210009, CHINA
| | - Rongcai Lin
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing, 210006, CHINA
| | - Yuguang Du
- Institute of Process Engineering Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China, Beijing, Beijing, 100190, CHINA
| | - Liming Wang
- Department of Orthopaedic Surgery Nanjing First Hospital, Nanjing Medical University, Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing, Jiangsu Province, 210006, CHINA
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China, Nanjing, 210006, CHINA
| | - Andrew Zannettino
- Adelaide Medical School Research, The University of Adelaide, Adelaide, Australia, Adelaide, South Australia, 5005, AUSTRALIA
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, CA 91711, USA, 535 Watson Drive, Claremont, CA, USA, Claremont, California, 91711, UNITED STATES
| |
Collapse
|
10
|
Alvites R, Branquinho M, Sousa AC, Lopes B, Sousa P, Maurício AC. Mesenchymal Stem/Stromal Cells and Their Paracrine Activity-Immunomodulation Mechanisms and How to Influence the Therapeutic Potential. Pharmaceutics 2022; 14:381. [PMID: 35214113 PMCID: PMC8875256 DOI: 10.3390/pharmaceutics14020381] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023] Open
Abstract
With high clinical interest to be applied in regenerative medicine, Mesenchymal Stem/Stromal Cells have been widely studied due to their multipotency, wide distribution, and relative ease of isolation and expansion in vitro. Their remarkable biological characteristics and high immunomodulatory influence have opened doors to the application of MSCs in many clinical settings. The therapeutic influence of these cells and the interaction with the immune system seems to occur both directly and through a paracrine route, with the production and secretion of soluble factors and extracellular vesicles. The complex mechanisms through which this influence takes place is not fully understood, but several functional manipulation techniques, such as cell engineering, priming, and preconditioning, have been developed. In this review, the knowledge about the immunoregulatory and immunomodulatory capacity of MSCs and their secretion products is revisited, with a special focus on the phenomena of migration and homing, direct cell action and paracrine activity. The techniques for homing improvement, cell modulation and conditioning prior to the application of paracrine factors were also explored. Finally, multiple assays where different approaches were applied with varying success were used as examples to justify their exploration.
Collapse
Affiliation(s)
- Rui Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (R.A.); (M.B.); (A.C.S.); (B.L.); (P.S.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Mariana Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (R.A.); (M.B.); (A.C.S.); (B.L.); (P.S.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Ana C. Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (R.A.); (M.B.); (A.C.S.); (B.L.); (P.S.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (R.A.); (M.B.); (A.C.S.); (B.L.); (P.S.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (R.A.); (M.B.); (A.C.S.); (B.L.); (P.S.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal; (R.A.); (M.B.); (A.C.S.); (B.L.); (P.S.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| |
Collapse
|
11
|
Karlsen TA, Sundaram AYM, Brinchmann JE. Single-Cell RNA Sequencing of In Vitro Expanded Chondrocytes: MSC-Like Cells With No Evidence of Distinct Subsets. Cartilage 2021; 13:774S-784S. [PMID: 31072202 PMCID: PMC8804791 DOI: 10.1177/1947603519847746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To investigate the heterogeneity of in vitro expanded chondrocytes used for autologous chondrocyte implantation. METHODS Human articular chondrocytes were expanded in vitro for 14 days, sorted into 86 single cells using fluorescence-activated cell sorting and subjected to single-cell RNA sequencing. Principal component, Cross R2 hierarchical clustering, and differential gene expression analyses were used for data evaluation. Flow cytometry and single-cell RT-qPCR (reverse transcriptase quantitative polymerase chain reaction) was used to validate the results of the RNA sequencing data Polyclonal chondrocyte populations from the same donor were differentiated in vitro toward the osteogenic and adipogenic lineages. RESULTS There was considerable variation in gene expression between individual cells, but we found no evidence for separate cell subpopulations based on principal component, hierarchical clustering, and differential gene expression analysis. Most of the cells expressed all the markers defining mesenchymal stem cells, and as polyclonal chondrocyte populations from the same donor were shown to differentiate into osteocytes and adipocytes in vitro, these cells formally qualify as mesenchymal stem cells. CONCLUSIONS In vitro expanded chondrocytes consist of one single population of cells with heterogeneity in gene expression between the cells. Dedifferentiated chondrocytes qualify as mesenchymal stem cells as they fulfill all the criteria suggested by the International Society for Cellular Therapy.
Collapse
Affiliation(s)
- Tommy A. Karlsen
- Norwegian Center for Stem Cell
Research, Department of Immunology, Oslo University Hospital Rikshospitalet,
Oslo, Norway,Tommy A. Karlsen, Department of
Immunology, Oslo University Hospital Rikshospitalet, PO Box 4950
Nydalen, Oslo 0424, Norway.
| | - Arvind Y. M. Sundaram
- Norwegian Sequencing Centre,
Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jan E. Brinchmann
- Norwegian Center for Stem Cell
Research, Department of Immunology, Oslo University Hospital Rikshospitalet,
Oslo, Norway,Department of Molecular Medicine,
Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
12
|
Xavier M, Kyriazi ME, Lanham S, Alexaki K, Matthews E, El-Sagheer AH, Brown T, Kanaras AG, Oreffo ROC. Enrichment of Skeletal Stem Cells from Human Bone Marrow Using Spherical Nucleic Acids. ACS NANO 2021; 15:6909-6916. [PMID: 33751885 DOI: 10.1021/acsnano.0c10683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Human bone marrow (BM)-derived stromal cells contain a population of skeletal stem cells (SSCs), with the capacity to differentiate along the osteogenic, adipogenic, and chondrogenic lineages, enabling their application to clinical therapies. However, current methods to isolate and enrich SSCs from human tissues remain, at best, challenging in the absence of a specific SSC marker. Unfortunately, none of the current proposed markers alone can isolate a homogeneous cell population with the ability to form bone, cartilage, and adipose tissue in humans. Here, we have designed DNA-gold nanoparticles able to identify and sort SSCs displaying specific mRNA signatures. The current approach demonstrates the significant enrichment attained in the isolation of SSCs, with potential therein to enhance our understanding of bone cell biology and translational applications.
Collapse
Affiliation(s)
- Miguel Xavier
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Maria-Eleni Kyriazi
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Stuart Lanham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Konstantina Alexaki
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Elloise Matthews
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Afaf H El-Sagheer
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
- Chemistry Branch, Department of Science and Mathematics, Faculty of Petroleum and Mining Engineering, Suez University, Suez 43721, Egypt
| | - Tom Brown
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Antonios G Kanaras
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
13
|
Donsante S, Palmisano B, Serafini M, Robey PG, Corsi A, Riminucci M. From Stem Cells to Bone-Forming Cells. Int J Mol Sci 2021; 22:ijms22083989. [PMID: 33924333 PMCID: PMC8070464 DOI: 10.3390/ijms22083989] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 12/22/2022] Open
Abstract
Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.
Collapse
Affiliation(s)
- Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA;
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Correspondence:
| |
Collapse
|
14
|
Katoh S, Yoshioka H, Iwasaki M, Senthilkumar R, Rajmohan M, Karthick R, Preethy S, Abraham SJ. A three-dimensional in vitro culture environment of a novel polymer scaffold, yielding chondroprogenitors and mesenchymal stem cells in human chondrocytes derived from osteoarthritis-affected cartilage tissue. J Orthop 2021; 23:138-141. [PMID: 33510554 DOI: 10.1016/j.jor.2021.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/10/2021] [Indexed: 01/30/2023] Open
Abstract
Objective We evaluated the expression of stem/progenitor biomarkers in osteoarthritic tissue derived chondrocytes cultured using a three-dimensional (3D) thermo-reversible gelation polymer (TGP). Methods The chondrocytes from discarded biopsy tissues obtained from human elderly patients with osteoarthritis were cultured using the 3D-TGP up to six weeks. Results The chondrocytes grew in a tissue-like manner, without de-differentiation into fibroblasts, and the cells thus tissue-engineered were proven positive for CD49e, OCT4, CD-105 and STRO-1 by immunohistochemistry. Conclusion This study establishes the efficacy of this 3D-TGP platform for clinically useable in-vitro tissue-engineered cartilage for improvising the clinical outcome of cell therapy for cartilage repair.
Collapse
Affiliation(s)
- Shojiro Katoh
- Edogawa Evolutionary Lab of Science, Edogawa Hospital Campus, 2-24-18, Higashi Koiwa, Edogawa-Ku, Tokyo, 133-0052, Japan.,Department of Orthopaedic Surgery, Edogawa Hospital, 2-24-18, Higashi Koiwa, Edogawa-Ku, Tokyo, 133-0052, Japan
| | - Hiroshi Yoshioka
- Mebiol Inc, 1-25-8, Nakahara, Hiratsuka, 254-0075, Kanagawa, Japan
| | - Masaru Iwasaki
- Centre for Advancing Clinical Research (CACR), University of Yamanashi -Faculty of Medicine, 1110, Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Rajappa Senthilkumar
- The Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), PB 1262, Chennai, 600034, Tamil Nadu, India
| | - Mathaiyan Rajmohan
- The Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), PB 1262, Chennai, 600034, Tamil Nadu, India
| | - Ramalingam Karthick
- The Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), PB 1262, Chennai, 600034, Tamil Nadu, India
| | - Senthilkumar Preethy
- The Fujio-Eiji Academic Terrain (FEAT), Nichi-In Centre for Regenerative Medicine (NCRM), PB 1262, Chennai, 600034, Tamil Nadu, India
| | - Samuel Jk Abraham
- Centre for Advancing Clinical Research (CACR), University of Yamanashi -Faculty of Medicine, 1110, Shimokato, Chuo, Yamanashi, 409-3898, Japan.,The Mary-Yoshio Translational Hexagon (MYTH), Nichi-In Centre for Regenerative Medicine (NCRM), PB 1262, Chennai, 600034, Tamil Nadu, India.,JBM Inc, 3-1-14, Higashi Koiwa, Edogawa-Ku, Tokyo, 133-0052, Japan.,GN Corporation Co. Ltd, 3-8, Wakamatsu, Kofu, Yamanashi, 400-0866, Japan
| |
Collapse
|
15
|
Stumpf PS, Du X, Imanishi H, Kunisaki Y, Semba Y, Noble T, Smith RCG, Rose-Zerili M, West JJ, Oreffo ROC, Farrahi K, Niranjan M, Akashi K, Arai F, MacArthur BD. Transfer learning efficiently maps bone marrow cell types from mouse to human using single-cell RNA sequencing. Commun Biol 2020; 3:736. [PMID: 33277618 PMCID: PMC7718277 DOI: 10.1038/s42003-020-01463-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 10/30/2020] [Indexed: 12/22/2022] Open
Abstract
Biomedical research often involves conducting experiments on model organisms in the anticipation that the biology learnt will transfer to humans. Previous comparative studies of mouse and human tissues were limited by the use of bulk-cell material. Here we show that transfer learning-the branch of machine learning that concerns passing information from one domain to another-can be used to efficiently map bone marrow biology between species, using data obtained from single-cell RNA sequencing. We first trained a multiclass logistic regression model to recognize different cell types in mouse bone marrow achieving equivalent performance to more complex artificial neural networks. Furthermore, it was able to identify individual human bone marrow cells with 83% overall accuracy. However, some human cell types were not easily identified, indicating important differences in biology. When re-training the mouse classifier using data from human, less than 10 human cells of a given type were needed to accurately learn its representation. In some cases, human cell identities could be inferred directly from the mouse classifier via zero-shot learning. These results show how simple machine learning models can be used to reconstruct complex biology from limited data, with broad implications for biomedical research.
Collapse
Affiliation(s)
- Patrick S Stumpf
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK.
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen, 52074, Germany.
| | - Xin Du
- Electronics and Computer Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Haruka Imanishi
- Kyushu University, Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yuya Kunisaki
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Yuichiro Semba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Timothy Noble
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| | - Rosanna C G Smith
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Matthew Rose-Zerili
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Jonathan J West
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Katayoun Farrahi
- Electronics and Computer Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Mahesan Niranjan
- Electronics and Computer Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, 812-8582, Japan
| | - Fumio Arai
- Kyushu University, Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Ben D MacArthur
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK.
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- Mathematical Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
- The Alan Turing Institute, London, NW1 2DB, UK.
| |
Collapse
|
16
|
Choudhary P, Gupta A, Singh S. Therapeutic Advancement in Neuronal Transdifferentiation of Mesenchymal Stromal Cells for Neurological Disorders. J Mol Neurosci 2020; 71:889-901. [PMID: 33047251 DOI: 10.1007/s12031-020-01714-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders have become the leading cause of chronic pain and death. Treatments available are not sufficient to help the patients as they only alleviate the symptoms and not the cause. In this regard, stem cells therapy has emerged as an upcoming option for the replacement of dead and damaged neurons. Stem cells, in general, are characterized as cells exhibiting potency properties, i.e., on being subjected to specific conditions they transform into cells of another lineage. Of all the types, mesenchymal stem cells (MSCs) are known for their pluripotent nature without the obstacle of ethical concern surrounding the procurement of other cell types. Although fibroblasts are quite similar to MSCs morphologically, certain markers like CD73, CD 90 are specific to MSCs, making both the cell types distinguishable from each other. This is implemented while procuring MSCs from a plethora of sources like umbilical cord blood, adipose tissue, bone marrow, etc. Among these, bone marrow MSCs are the most widely used type for neural regeneration. Neural regeneration is achieved via transdifferentiation. Several studies have either transplanted the stem cells into rodent models or have carried out transdifferentiation in vitro. The process involves a combination of growth factors, pre-treatment factors, and neuronal differentiation inducing mediums. The results obtained are characterized by neuron-like morphology, expression of markers, along with electrophysical activity in some. Recent attempts involve exploring biomaterials that may mimic the native ECM and therefore can be directly introduced at the site of interest. The review gives a brief description of MSCs, their sources and markers, and the different attempts that have been made towards achieving the goal of differentiating MSCs into neurons.
Collapse
Affiliation(s)
- Princy Choudhary
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Ayushi Gupta
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India
| | - Sangeeta Singh
- Applied Science Department, Indian Institute of Information Technology, Allahabad, UP, India.
| |
Collapse
|
17
|
Zhang W, Jia K, Jia P, Xiang Y, Lu X, Liu W, Yi M. Marine medaka heat shock protein 90ab1 is a receptor for red-spotted grouper nervous necrosis virus and promotes virus internalization through clathrin-mediated endocytosis. PLoS Pathog 2020; 16:e1008668. [PMID: 32639977 PMCID: PMC7371229 DOI: 10.1371/journal.ppat.1008668] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/20/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022] Open
Abstract
Nervous necrosis virus (NNV) can infect many species of fish and causes serious acute or persistent infection. However, its pathogenic mechanism is still far from clear. Specific cellular surface receptors are crucial determinants of the species tropism of a virus and its pathogenesis. Here, the heat shock protein 90ab1 of marine model fish species marine medaka (MmHSP90ab1) was identified as a novel receptor of red-spotted grouper NNV (RGNNV). MmHSP90ab1 interacted directly with RGNNV capsid protein (CP). Specifically, MmHSP90ab1 bound to the linker region (LR) of CP through its NM domain. Inhibition of MmHSP90ab1 by HSP90-specific inhibitors or MmHSP90ab1 siRNA caused significant inhibition of viral binding and entry, whereas its overexpression led to the opposite effect. The binding of RGNNV to cultured marine medaka hMMES1 cells was inhibited by blocking cell surface-localized MmHSP90ab1 with anti-HSP90β antibodies or pretreating virus with recombinant MmHSP90ab1 or MmHSP90ab1-NM protein, indicating MmHSP90ab1 was an attachment receptor for RGNNV. Furthermore, we found that MmHSP90ab1 formed a complex with CP and marine medaka heat shock cognate 70, a known NNV receptor. Exogenous expression of MmHSP90ab1 independently facilitated the internalization of RGNNV into RGNNV impenetrable cells (HEK293T), which was blocked by chlorpromazine, an inhibitor of clathrin-dependent endocytosis. Further study revealed that MmHSP90ab1 interacted with the marine medaka clathrin heavy chain. Collectively, these data suggest that MmHSP90ab1 is a functional part of the RGNNV receptor complex and involved in the internalization of RGNNV via the clathrin endocytosis pathway.
Collapse
Affiliation(s)
- Wanwan Zhang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Kuntong Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
- * E-mail: (KJ); (MY)
| | - Peng Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Yangxi Xiang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Xiaobing Lu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Wei Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China
- * E-mail: (KJ); (MY)
| |
Collapse
|
18
|
Melatonin and Mesenchymal Stem Cells as a Key for Functional Integrity for Liver Cancer Treatment. Int J Mol Sci 2020; 21:ijms21124521. [PMID: 32630505 PMCID: PMC7350224 DOI: 10.3390/ijms21124521] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common hepatobiliary malignancy with limited therapeutic options. On the other hand, melatonin is an indoleamine that modulates a variety of potential therapeutic effects. In addition to its important role in the regulation of sleep–wake rhythms, several previous studies linked the biologic effects of melatonin to various substantial endocrine, neural, immune and antioxidant functions, among others. Furthermore, the effects of melatonin could be influenced through receptor dependent and receptor independent manner. Among the other numerous physiological and therapeutic effects of melatonin, controlling the survival and differentiation of mesenchymal stem cells (MSCs) has been recently discussed. Given its controversial interaction, several previous reports revealed the therapeutic potential of MSCs in controlling the hepatocellular carcinoma (HCC). Taken together, the intention of the present review is to highlight the effects of melatonin and mesenchymal stem cells as a key for functional integrity for liver cancer treatment. We hope to provide solid piece of information that may be helpful in designing novel drug targets to control HCC.
Collapse
|
19
|
Bartold M, Gronthos S, Haynes D, Ivanovski S. Mesenchymal stem cells and biologic factors leading to bone formation. J Clin Periodontol 2019; 46 Suppl 21:12-32. [PMID: 30624807 DOI: 10.1111/jcpe.13053] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/23/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Physiological bone formation and bone regeneration occurring during bone repair can be considered distinct but similar processes. Mesenchymal stem cells (MSC) and associated biologic factors are crucial to both bone formation and bone regeneration. AIM To perform a narrative review of the current literature regarding the role of MSC and biologic factors in bone formation with the aim of discussing the clinical relevance of in vitro and in vivo animal studies. METHODS The literature was searched for studies on MSC and biologic factors associated with the formation of bone in the mandible and maxilla. The search specifically targeted studies on key aspects of how stem cells and biologic factors are important in bone formation and how this might be relevant to bone regeneration. The results are summarized in a narrative review format. RESULTS Different types of MSC and many biologic factors are associated with bone formation in the maxilla and mandible. CONCLUSION Bone formation and regeneration involve very complex and highly regulated cellular and molecular processes. By studying these processes, new clinical opportunities will arise for therapeutic bone regenerative treatments.
Collapse
Affiliation(s)
- Mark Bartold
- School of Dentistry, University of Adelaide, Adelaide, SA, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - David Haynes
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Saso Ivanovski
- School of Dentistry, University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
20
|
Kanczler J, Tare RS, Stumpf P, Noble TJ, Black C, Oreffo ROC. Isolation, Differentiation, and Characterization of Human Bone Marrow Stem Cells In Vitro and In Vivo. Methods Mol Biol 2019; 1914:53-70. [PMID: 30729460 DOI: 10.1007/978-1-4939-8997-3_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this chapter, we describe techniques for the isolation and characterisation of skeletal stem cells from human bone marrow. The methods for enrichment of STRO-1+ and STRO-4+ cells using magnetic activated cell sorting are described and we also detail techniques for establishing and characterizing osteogenic, adipogenic, and chondrogenic cultures from these cells. Finally, we present methods for studying the ability of these cells to produce bone in vivo using diffusion chambers which have been implanted subcutaneously into mice.
Collapse
Affiliation(s)
- Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton, UK
| | - Rahul S Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton, UK
| | - Patrick Stumpf
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton, UK
| | - Timothy J Noble
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton, UK
| | - Cameron Black
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton, UK.
| |
Collapse
|
21
|
Abstract
Teeth are exposed to hundreds of oral bacteria and also challenged by the mastication forces; because teeth are situated in oral cavity, the entrance of the digestive tract, and penetrates through the oral epithelium. The periodontal ligament is a noncalcified tissue that possesses abundant blood vessels, which exist between tooth root and alveolar bone. The ligament is thought to play an important role in absorbing the impact of mastication, in the maintenance of periodontal homeostasis, and in periodontal wound healing. We succeeded in isolating mesenchymal stem cells (MSCs), so-called periodontal stem cells (PDLSCs), with self-renewability and multipotency from the periodontal ligament. We also demonstrated that PDLSCs share some cell surface markers with pericytes and that PDLSCs distribute themselves to stay with the endothelial cell networks and that PDLSCs maintain the endothelial cell networks when added to endothelial cell network formation systems. Pericytes are located in the proximity of microvascular endothelial cells and thought to stabilize and supply nutrients to blood vessels. Recently, it was also reported that pericytes possess multipotency and can be the source of tissue stem cells and/or progenitor cells. This review explores the distinctive features of the periodontal ligament tissue and PDLSCs as well as the puzzling similarities between PDLSCs and pericytes.
Collapse
|
22
|
Fitzsimmons REB, Mazurek MS, Soos A, Simmons CA. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem Cells Int 2018; 2018:8031718. [PMID: 30210552 PMCID: PMC6120267 DOI: 10.1155/2018/8031718] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/08/2023] Open
Abstract
As a result of over five decades of investigation, mesenchymal stromal/stem cells (MSCs) have emerged as a versatile and frequently utilized cell source in the fields of regenerative medicine and tissue engineering. In this review, we summarize the history of MSC research from the initial discovery of their multipotency to the more recent recognition of their perivascular identity in vivo and their extraordinary capacity for immunomodulation and angiogenic signaling. As well, we discuss long-standing questions regarding their developmental origins and their capacity for differentiation toward a range of cell lineages. We also highlight important considerations and potential risks involved with their isolation, ex vivo expansion, and clinical use. Overall, this review aims to serve as an overview of the breadth of research that has demonstrated the utility of MSCs in a wide range of clinical contexts and continues to unravel the mechanisms by which these cells exert their therapeutic effects.
Collapse
Affiliation(s)
- Ross E. B. Fitzsimmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Matthew S. Mazurek
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada T2N 4Z6
| | - Agnes Soos
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Craig A. Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada M5S 3G8
| |
Collapse
|
23
|
Xavier M, de Andrés MC, Spencer D, Oreffo ROC, Morgan H. Size and dielectric properties of skeletal stem cells change critically after enrichment and expansion from human bone marrow: consequences for microfluidic cell sorting. J R Soc Interface 2018; 14:rsif.2017.0233. [PMID: 28835540 PMCID: PMC5582119 DOI: 10.1098/rsif.2017.0233] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/27/2017] [Indexed: 12/14/2022] Open
Abstract
The capacity of bone and cartilage to regenerate can be attributed to skeletal stem cells (SSCs) that reside within the bone marrow (BM). Given SSCs are rare and lack specific surface markers, antibody-based sorting has failed to deliver the cell purity required for clinical translation. Microfluidics offers new methods of isolating cells based on biophysical features including, but not limited to, size, electrical properties and stiffness. Here we report the characterization of the dielectric properties of unexpanded SSCs using single-cell microfluidic impedance cytometry (MIC). Unexpanded SSCs had a mean size of 9.0 µm; larger than the majority of BM cells. During expansion, often used to purify and increase the number of SSCs, cell size and membrane capacitance increased significantly, highlighting the importance of characterizing unaltered SSCs. In addition, MIC was used to track the osteogenic differentiation of SSCs and showed an increased membrane capacitance with differentiation. The electrical properties of primary SSCs were indistinct from other BM cells precluding its use as an isolation method. However, the current studies indicate that cell size in combination with another biophysical parameter, such as stiffness, could be used to design label-free devices for sorting SSCs with significant clinical impact.
Collapse
Affiliation(s)
- Miguel Xavier
- Faculty of Physical Sciences and Engineering, and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.,Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Southampton General Hospital, Tremona Road, SO16 6YD Southampton, UK
| | - María C de Andrés
- Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Southampton General Hospital, Tremona Road, SO16 6YD Southampton, UK
| | - Daniel Spencer
- Faculty of Physical Sciences and Engineering, and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Southampton General Hospital, Tremona Road, SO16 6YD Southampton, UK
| | - Hywel Morgan
- Faculty of Physical Sciences and Engineering, and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
24
|
Zhang J, Yun S, Bi J, Dai S, Du Y, Zannettino ACW, Zhang H. Enhanced multi-lineage differentiation of human mesenchymal stem/stromal cells within poly(N-isopropylacrylamide-acrylic acid) microgel-formed three-dimensional constructs. J Mater Chem B 2018; 6:1799-1814. [PMID: 32254252 DOI: 10.1039/c8tb00376a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human mesenchymal stem/stromal cells (hMSCs) are a potential cell source of stem cell therapy for many serious diseases and hMSC spheroids have emerged to replace single cell suspensions for cell therapy. Three-dimensional (3D) scaffolds or hydrogels which can mimic properties of the extracellular matrix (ECM) have been widely explored for their application in tissue regeneration. However, there are considerably less studies on inducing differentiation of hMSC spheroids using 3D scaffolds or hydrogels. This study is the first to explore multi-lineage differentiation of a stem cell line and primary stem cells within poly(N-isopropylacrylamide) (p(NIPAAm))-based thermosensitive microgel-formed constructs. We first demonstrated that poly(N-isopropylacrylamide-co-acrylic acid) (p(NIPAAm-AA)) was not toxic to hMSCs and the microgel-formed constructs facilitated formation of uniform stem cell spheroids. Due to functional enhancement of cell spheroids, hMSCs within the 3D microgel-formed constructs were induced for multi-lineage differentiation as evidenced by significant up-regulation of messenger RNA (mRNA) expression of chondrogenic and osteogenic genes even in the absence of induction media on day 9. When induction media were in situ supplied on day 9, mRNA expression of chondrogenic, osteogenic and adipogenic genes within the microgel-formed constructs were significantly higher than that in the pellet and 2D cultures, respectively, on day 37. In addition, histological and immunofluorescent images also confirmed successful multi-lineage differentiation of hMSCs within the 3D microgel-formed constructs. Hence, the thermosensitive p(NIPAAm-AA) microgel can be potentially used in an in vitro model for cell differentiation or in vivo transplantation of pre-differentiated human mesenchymal stromal cells into patients for specific lineage differentiation.
Collapse
Affiliation(s)
- Jiabin Zhang
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | | | | | | | | | | | | |
Collapse
|
25
|
Tribe HC, McEwan J, Taylor H, Oreffo ROC, Tare RS. Mesenchymal Stem Cells: Potential Role in the Treatment of Osteochondral Lesions of the Ankle. Biotechnol J 2017; 12:1700070. [PMID: 29068173 PMCID: PMC5765412 DOI: 10.1002/biot.201700070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/13/2017] [Indexed: 12/11/2022]
Abstract
Given articular cartilage has a limited repair potential, untreated osteochondral lesions of the ankle can lead to debilitating symptoms and joint deterioration necessitating joint replacement. While a wide range of reparative and restorative surgical techniques have been developed to treat osteochondral lesions of the ankle, there is no consensus in the literature regarding which is the ideal treatment. Tissue engineering strategies, encompassing stem cells, somatic cells, biomaterials, and stimulatory signals (biological and mechanical), have a potentially valuable role in the treatment of osteochondral lesions. Mesenchymal stem cells (MSCs) are an attractive resource for regenerative medicine approaches, given their ability to self-renew and differentiate into multiple stromal cell types, including chondrocytes. Although MSCs have demonstrated significant promise in in vitro and in vivo preclinical studies, their success in treating osteochondral lesions of the ankle is inconsistent, necessitating further clinical trials to validate their application. This review highlights the role of MSCs in cartilage regeneration and how the application of biomaterials and stimulatory signals can enhance chondrogenesis. The current treatments for osteochondral lesions of the ankle using regenerative medicine strategies are reviewed to provide a clinical context. The challenges for cartilage regeneration, along with potential solutions and safety concerns are also discussed.
Collapse
Affiliation(s)
- Howard C. Tribe
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Josephine McEwan
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Heath Taylor
- Foot and Ankle Orthopaedic DepartmentRoyal Bournemouth HospitalBournemouthBH7 7DWUK
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
| | - Rahul S. Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and RegenerationFaculty of MedicineUniversity of SouthamptonSouthamptonSO16 6YDUK
- Bioengineering Science, Mechanical Engineering DepartmentFaculty of Engineering and the EnvironmentUniversity of SouthamptonSouthamptonSO17 1BJUK
| |
Collapse
|
26
|
Samsonraj RM, Raghunath M, Nurcombe V, Hui JH, van Wijnen AJ, Cool SM. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem Cells Transl Med 2017; 6:2173-2185. [PMID: 29076267 PMCID: PMC5702523 DOI: 10.1002/sctm.17-0129] [Citation(s) in RCA: 511] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC) hold great potential for regenerative medicine because of their ability for self-renewal and differentiation into tissue-specific cells such as osteoblasts, chondrocytes, and adipocytes. MSCs orchestrate tissue development, maintenance and repair, and are useful for musculoskeletal regenerative therapies to treat age-related orthopedic degenerative diseases and other clinical conditions. Importantly, MSCs produce secretory factors that play critical roles in tissue repair that support both engraftment and trophic functions (autocrine and paracrine). The development of uniform protocols for both preparation and characterization of MSCs, including standardized functional assays for evaluation of their biological potential, are critical factors contributing to their clinical utility. Quality control and release criteria for MSCs should include cell surface markers, differentiation potential, and other essential cell parameters. For example, cell surface marker profiles (surfactome), bone-forming capacities in ectopic and orthotopic models, as well as cell size and granularity, telomere length, senescence status, trophic factor secretion (secretome), and immunomodulation, should be thoroughly assessed to predict MSC utility for regenerative medicine. We propose that these and other functionalities of MSCs should be characterized prior to use in clinical applications as part of comprehensive and uniform guidelines and release criteria for their clinical-grade production to achieve predictably favorable treatment outcomes for stem cell therapy. Stem Cells Translational Medicine 2017;6:2173-2185.
Collapse
Affiliation(s)
- Rebekah M. Samsonraj
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore
- Department of Orthopaedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Michael Raghunath
- Department of Biomedical EngineeringNational University of SingaporeSingapore
- Center for Cell Biology and Tissue Engineering, Competence Center for Tissue Engineering and Substance Testing (TEDD)Institute for Chemistry and Biotechnology, ZHAW School of Life Sciences and Facility Management, Zurich University of Applied SciencesSwitzerland
| | - Victor Nurcombe
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
| | - James H. Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | | | - Simon M. Cool
- Glycotherapeutics GroupInstitute of Medical Biology, Agency for Science, Technology and Research (A*STAR)Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| |
Collapse
|
27
|
Bearden RN, Huggins SS, Cummings KJ, Smith R, Gregory CA, Saunders WB. In-vitro characterization of canine multipotent stromal cells isolated from synovium, bone marrow, and adipose tissue: a donor-matched comparative study. Stem Cell Res Ther 2017; 8:218. [PMID: 28974260 PMCID: PMC5627404 DOI: 10.1186/s13287-017-0639-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Background The dog represents an excellent large animal model for translational cell-based studies. Importantly, the properties of canine multipotent stromal cells (cMSCs) and the ideal tissue source for specific translational studies have yet to be established. The aim of this study was to characterize cMSCs derived from synovium, bone marrow, and adipose tissue using a donor-matched study design and a comprehensive series of in-vitro characterization, differentiation, and immunomodulation assays. Methods Canine MSCs were isolated from five dogs with cranial cruciate ligament rupture. All 15 cMSC preparations were evaluated using colony forming unit (CFU) assays, flow cytometry analysis, RT-PCR for pluripotency-associated genes, proliferation assays, trilineage differentiation assays, and immunomodulation assays. Data were reported as mean ± standard deviation and compared using repeated-measures analysis of variance and Tukey post-hoc test. Significance was established at p < 0.05. Results All tissue samples produced plastic adherent, spindle-shaped preparations of cMSCs. Cells were negative for CD34, CD45, and STRO-1 and positive for CD9, CD44, and CD90, whereas the degree to which cells were positive for CD105 was variable depending on tissue of origin. Cells were positive for the pluripotency-associated genes NANOG, OCT4, and SOX2. Accounting for donor and tissue sources, there were significant differences in CFU potential, rate of proliferation, trilineage differentiation, and immunomodulatory response. Synovium and marrow cMSCs exhibited superior early osteogenic activity, but when assessing late-stage osteogenesis no significant differences were detected. Interestingly, bone morphogenic protein-2 (BMP-2) supplementation was necessary for early-stage and late-stage osteogenic differentiation, a finding consistent with other canine studies. Additionally, synovium and adipose cMSCs proliferated more rapidly, displayed higher CFU potential, and formed larger aggregates in chondrogenic assays, although proteoglycan and collagen type II staining were subjectively decreased in adipose pellets as compared to synovial and marrow pellets. Lastly, cMSCs derived from all three tissue sources modulated murine macrophage TNF-α and IL-6 levels in a lipopolysaccharide-stimulated coculture assay. Conclusions While cMSCs from synovium, marrow, and adipose tissue share a number of similarities, important differences in proliferation and trilineage differentiation exist and should be considered when selecting cMSCs for translational studies. These results and associated methods will prove useful for future translational studies involving the canine model. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0639-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert N Bearden
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shannon S Huggins
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Kevin J Cummings
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Roger Smith
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Institute for Regenerative Medicine, College of Medicine, Texas A&M University, College Station, TX, USA
| | - William B Saunders
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
28
|
Akhter S, Chakraborty S, Moutinho D, Álvarez-Coiradas E, Rosa I, Viñuela J, Domínguez E, García A, Requena JR. The human VGF-derived bioactive peptide TLQP-21 binds heat shock 71 kDa protein 8 (HSPA8)on the surface of SH-SY5Y cells. PLoS One 2017; 12:e0185176. [PMID: 28934328 PMCID: PMC5608341 DOI: 10.1371/journal.pone.0185176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/07/2017] [Indexed: 01/16/2023] Open
Abstract
VGF (non-acronymic)is a secreted chromogranin/secretogranin that gives rise to a number of bioactive peptides by a complex proteolysis mechanism. VGF-derived peptides exert an extensive array of biological effects in energy metabolism, mood regulation, pain, gastric secretion function, reproduction and, perhaps, cancer. It is therefore surprising that very little is known about receptors and binding partners of VGF-derived peptides and their downstream molecular mechanisms of action. Here, using affinity chromatography and mass spectrometry-based protein identification, we have identified the heat shock cognate 71 kDa protein A8 (HSPA8)as a binding partner of human TLQP-21 on the surface of human neuroblastomaSH-SY5Y cells. Binding of TLQP-21 to membrane associated HSPA8 in live SH-SY5Y cells was further supported by cross-linking to live cells. Interaction between HSPA8 and TLQP-21 was confirmed in vitro by label-free Dynamic Mass Redistribution (DMR) studies. Furthermore, molecular modeling studies show that TLQP-21 can be docked into the HSPA8 peptide binding pocket. Identification of HSPA8 as a cell surface binding partner of TLQP-21 opens new avenues to explore the molecular mechanisms of its physiological actions, and of pharmacological modulation thereof.
Collapse
Affiliation(s)
- Shamim Akhter
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | | | - Daniela Moutinho
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Elia Álvarez-Coiradas
- BioFarma Research Group, CIMUS, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Isaac Rosa
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Juan Viñuela
- Immunology Laboratory, Santiago University Hospital, Santiago de Compostela, Spain
| | - Eduardo Domínguez
- BioFarma Research Group, CIMUS, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Angel García
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| | - Jesús R. Requena
- CIMUS Biomedical Research Institute, University of Santiago de Compostela-IDIS, Santiago de Compostela, Spain
| |
Collapse
|