1
|
Kurniawan A, Dilogo IH, Pawitan JA, Widodo W, Oesman I, Martinus A. The effects of secretome of umbilical cord mesenchymal stem cells on regeneration of sciatic nerve defects in Sprague dawley rats. PLoS One 2024; 19:e0310467. [PMID: 39739689 DOI: 10.1371/journal.pone.0310467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 09/01/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND AND PURPOSE Current treatments for peripheral nerve defects are suboptimal. Mesenchymal stem cell (MSC) implantation holds promise, with studies indicating their efficacy through the secretome. This study aims to assess the secretome's potency in regenerating peripheral nerve defects. ANIMALS AND METHODS Twenty-eight spraque dawley (SD) rats were divided into four groups, with a 10mm segmental sciatic nerve defect created. Group I received autografts, while Groups II, III, and IV had proximal and distal nerve stumps fixed with a conduit. Group II received MSC implantation, Group III received umbilical cord MSC secretome, and Group IV was treated with silicone conduits. Motoric recovery was assessed using the Sciatic Functional Index (SFI) at 6, 9, and 12 weeks. After 12 weeks, rats were sacrificed for measurements of gastrocnemius muscle weight ratio and sciatic nerve histomorphometry. RESULT The highest ratio of the gastrocnemius muscle were found in groups 1 and 3, namely 0.7 ± 0.06 and 0.7 ± 0.2 (p <0.001). The highest number of myelinated axons was found in group 1 and 3, namely 175.1 ± 15.9 and 168.9 ± 11.7 (p <0.001). The secretome had the similar effectiveness with autograft in promoting regeneration of the sciatic nerve defect. INTERPRETATION The secretome of the umbilical cord MSC can be a novel therapy in replacing autografts for the management of peripheral nerve defects.
Collapse
Affiliation(s)
- Aryadi Kurniawan
- Faculty of Medicine Universitas Indonesia, Department of Orthopaedics & Traumatology, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
| | - Ismail Hadisoebroto Dilogo
- Faculty of Medicine Universitas Indonesia, Department of Orthopaedics & Traumatology, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
| | - Jeanne Adiwinata Pawitan
- Faculty of Medicine Universitas Indonesia, Department of Histology, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
| | - Wahyu Widodo
- Faculty of Medicine Universitas Indonesia, Department of Orthopaedics & Traumatology, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
| | - Ihsan Oesman
- Faculty of Medicine Universitas Indonesia, Department of Orthopaedics & Traumatology, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
| | - Ade Martinus
- Faculty of Medicine Universitas Indonesia, Department of Orthopaedics & Traumatology, Dr Cipto Mangunkusumo National Central Hospital, Jakarta, Indonesia
| |
Collapse
|
2
|
Abuarqoub D, Aslam N, Almajali B, Shajrawi L, Jafar H, Awidi A. Neuro-regenerative potential of dental stem cells: a concise review. Cell Tissue Res 2020; 382:267-279. [PMID: 32725424 DOI: 10.1007/s00441-020-03255-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/06/2020] [Indexed: 10/23/2022]
Abstract
This review will summarize the research information regarding the regenerative potential of dental stem cells for the treatment of neurodegenerative disorders. As compared to existing treatment modalities, the stem cell therapy seems promising, and accumulating evidences about the differentiation of stem cells into various lineages are proving it. The incidence of neurodegenerative diseases such as Alzheimer's, Parkinson's, stroke, and peripheral neuropathy is increasing due to the rise in life expectancies of people which have put a huge burden on economies. Finding a promising treatment could benefit not only the patients but also the communities. Dental stem cells hold a great potential to differentiate into neuronal cells. Many studies have reported the differentiation potential of the dental stem cells with the presence of neuronal lineage markers. In this review, we conferred how the use of dental stem cells can benefit the above-mentioned bedridden diseases.
Collapse
Affiliation(s)
- Duaa Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan. .,Cell Therapy Center, The University of Jordan, Amman, Jordan.
| | - Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Bayan Almajali
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Leen Shajrawi
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman, Jordan.,School of Medicine, The University of Jordan, Amman, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan. .,School of Medicine, The University of Jordan, Amman, Jordan.
| |
Collapse
|
3
|
Lee SJ, Zhu W, Nowicki M, Lee G, Heo DN, Kim J, Zuo YY, Zhang LG. 3D printing nano conductive multi-walled carbon nanotube scaffolds for nerve regeneration. J Neural Eng 2019; 15:016018. [PMID: 29064377 DOI: 10.1088/1741-2552/aa95a5] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Nanomaterials, such as carbon nanotubes (CNTs), have been introduced to modify the surface properties of scaffolds, thus enhancing the interaction between the neural cells and biomaterials. In addition to superior electrical conductivity, CNTs can provide nanoscale structures similar to those present in the natural neural environment. The primary objective of this study is to investigate the proliferative capability and differential potential of neural stem cells (NSCs) seeded on a CNT incorporated scaffold. APPROACH Amine functionalized multi-walled carbon nanotubes (MWCNTs) were incorporated with a PEGDA polymer to provide enhanced electrical properties as well as nanofeatures on the surface of the scaffold. A stereolithography 3D printer was employed to fabricate a well-dispersed MWCNT-hydrogel composite neural scaffold with a tunable porous structure. 3D printing allows easy fabrication of complex 3D scaffolds with extremely intricate microarchitectures and controlled porosity. MAIN RESULTS Our results showed that MWCNT-incorporated scaffolds promoted neural stem cell proliferation and early neuronal differentiation when compared to those scaffolds without the MWCNTs. Furthermore, biphasic pulse stimulation with 500 µA current promoted neuronal maturity quantified through protein expression analysis by quantitative polymerase chain reaction. SIGNIFICANCE Results of this study demonstrated that an electroconductive MWCNT scaffold, coupled with electrical stimulation, may have a synergistic effect on promoting neurite outgrowth for therapeutic application in nerve regeneration.
Collapse
Affiliation(s)
- Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Porzionato A, Barbon S, Stocco E, Dalzoppo D, Contran M, De Rose E, Parnigotto PP, Macchi V, Grandi C, De Caro R. Development of Oxidized Polyvinyl Alcohol-Based Nerve Conduits Coupled with the Ciliary Neurotrophic Factor. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E1996. [PMID: 31234386 PMCID: PMC6631399 DOI: 10.3390/ma12121996] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/09/2019] [Accepted: 06/19/2019] [Indexed: 12/14/2022]
Abstract
Functionalized synthetic conduits represent a promising strategy to enhance peripheral nerve regeneration by guiding axon growth while delivering therapeutic neurotrophic factors. In this work, hollow nerve conduits made of polyvinyl alcohol partially oxidized with bromine (OxPVA_Br2) and potassium permanganate (OxPVA_KMnO4) were investigated for their structural/biological properties and ability to absorb/release the ciliary neurotrophic factor (CNTF). Chemical oxidation enhanced water uptake capacity of the polymer, with maximum swelling index of 60.5% ± 2.5%, 71.3% ± 3.6% and 19.5% ± 4.0% for OxPVA_Br2, OxPVA_KMnO4 and PVA, respectively. Accordingly, hydrogel porosity increased from 15.27% ± 1.16% (PVA) to 62.71% ± 8.63% (OxPVA_Br2) or 77.50% ± 3.39% (OxPVA_KMnO4) after oxidation. Besides proving that oxidized PVA conduits exhibited mechanical resistance and a suture holding ability, they did not exert a cytotoxic effect on SH-SY5Y and Schwann cells and biodegraded over time when subjected to enzymatic digestion, functionalization with CNTF was performed. Interestingly, higher amounts of neurotrophic factor were detected in the lumen of OxPVA_Br2 (0.22 ± 0.029 µg) and OxPVA_KMnO4 (0.29 ± 0.033 µg) guides rather than PVA (0.11 ± 0.021 µg) tubular scaffolds. In conclusion, we defined a promising technology to obtain drug delivery conduits based on functionalizable oxidized PVA hydrogels.
Collapse
Affiliation(s)
- Andrea Porzionato
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Silvia Barbon
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Elena Stocco
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Daniele Dalzoppo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35128 Padova, Italy.
| | - Martina Contran
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
| | - Enrico De Rose
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, 35030 Padua, Italy.
| | - Veronica Macchi
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Claudio Grandi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35128 Padova, Italy.
| | - Raffaele De Caro
- Department of Neurosciences, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| |
Collapse
|
5
|
Campos JM, Sousa AC, Caseiro AR, Pedrosa SS, Pinto PO, Branquinho MV, Amorim I, Santos JD, Pereira T, Mendonça CM, Afonso A, Atayde LM, Maurício AC. Dental pulp stem cells and Bonelike ® for bone regeneration in ovine model. Regen Biomater 2018; 6:49-59. [PMID: 30740242 PMCID: PMC6362823 DOI: 10.1093/rb/rby025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/01/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022] Open
Abstract
Development of synthetic bone substitutes has arisen as a major research interest in the need to find an alternative to autologous bone grafts. Using an ovine model, the present pre-clinical study presents a synthetic bone graft (Bonelike®) in combination with a cellular system as an alternative for the regeneration of non-critical defects. The association of biomaterials and cell-based therapies is a promising strategy for bone tissue engineering. Mesenchymal stem cells (MSCs) from human dental pulp have demonstrated both in vitro and in vivo to interact with diverse biomaterial systems and promote mineral deposition, aiming at the reconstruction of osseous defects. Moreover, these cells can be found and isolated from many species. Non-critical bone defects were treated with Bonelike® with or without MSCs obtained from the human dental pulp. Results showed that Bonelike® and MSCs treated defects showed improved bone regeneration compared with the defects treated with Bonelike® alone. Also, it was observed that the biomaterial matrix was reabsorbed and gradually replaced by new bone during the healing process. We therefore propose this combination as an efficient binomial strategy that promotes bone growth and vascularization in non-critical bone defects.
Collapse
Affiliation(s)
- J M Campos
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal.,Escola Universitária Vasco da Gama (EUVG), Hospital Veterinário Universitário de Coimbra (HVUC), Campo Universitário - Bloco B, Lordemão, Coimbra, Portugal
| | - A C Sousa
- REQUIMTE/LAQV - U. Porto - Porto/Portugal, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua, Dr. Roberto Frias, s/n, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Rua Dr. Roberto Frias, Porto, Portugal
| | - A R Caseiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal.,REQUIMTE/LAQV - U. Porto - Porto/Portugal, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua, Dr. Roberto Frias, s/n, Porto, Portugal
| | - S S Pedrosa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - P O Pinto
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal.,Escola Universitária Vasco da Gama (EUVG), Hospital Veterinário Universitário de Coimbra (HVUC), Campo Universitário - Bloco B, Lordemão, Coimbra, Portugal
| | - M V Branquinho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - I Amorim
- Department of Pathology and Molecular Immunology of the Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Institute for Research and Innovation in Health, (i3S), University of Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - J D Santos
- REQUIMTE/LAQV - U. Porto - Porto/Portugal, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua, Dr. Roberto Frias, s/n, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Rua Dr. Roberto Frias, Porto, Portugal
| | - T Pereira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - C M Mendonça
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - A Afonso
- Faculdade de Medicina Dentária da Universidade do Porto (FMDUP), Porto, Portugal
| | - L M Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - A C Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, no 228, Porto, Portugal.,Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| |
Collapse
|
6
|
Romanov YA, Volgina NE, Dugina TN, Kabaeva NV, Sukhikh GT. Human Umbilical Cord Mesenchymal Stromal Cell-Derived Microvesicles Express Surface Markers Identical to the Phenotype of Parental Cells. Bull Exp Biol Med 2018; 166:124-129. [DOI: 10.1007/s10517-018-4300-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Indexed: 02/06/2023]
|
7
|
Caseiro AR, Ivanova G, Pedrosa SS, Branquinho MV, Georgieva P, Barbosa PP, Santos JD, Magalhães R, Teixeira P, Pereira T, Maurício AC. Human umbilical cord blood plasma as an alternative to animal sera for mesenchymal stromal cells in vitro expansion - A multicomponent metabolomic analysis. PLoS One 2018; 13:e0203936. [PMID: 30304014 PMCID: PMC6179201 DOI: 10.1371/journal.pone.0203936] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 08/30/2018] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal Stromal cells (MSCs) have a potential role in cell-based therapies. Foetal bovine serum (FBS) is used to supplement the basal cell culture medium but presents several disadvantages and risks. Other alternatives have been studied, including human umbilical cord blood plasma (hUCBP), aiming at the development of xeno-free culturing protocols. A comparative characterization of multicomponent metabolic composition of hUCBP and commercial FBS based on Nuclear Magnetic Resonance (NMR) spectroscopy and multivariate statistical analysis was performed. The analysis of 1H-NMR spectra revealed both similarities and differences between the two proposed supplements. Similar metabolites (amino acids, glucose, lipids and nucleotides) were found in the hUCBP and FBS NMR spectra. The results show that the major difference between the metabolic profiles of the two proposed supplements are due to the significantly higher levels of glucose and lower levels of lactate, glutamate, alanine and branched chain amino acids in hUCBP. Similar or slightly different levels of important proteinogenic amino acids, as well as of nucleotides, lipids were found in the hUCBP and FBS. In order to validate it’s suitability for cell culture, umbilical cord-MSCs (UC-MSCs) and dental pulp stem cells (DPSCs) were expanded using hUCBP. In both hMSCs, in vitro culture with hUCBP supplementation presented similar to improved metabolic performances when compared to FBS. The two cell types tested expressed different optimum hUCBP percentage content. For DPSCs, the optimum hUCBP content was 6% and for UC-MSCs, 4%. Cultured hMSCs displayed no changes in senescence indicators, as well as maintained characteristic surface marker’s expression. FBS substitution was associated with an increase in early apoptosis events, in a dose dependent manner, as well as to slight up- and down-regulation of targeted gene’s expression. Tri-lineage differentiation capacity was also influenced by the substitution of FBS by hUCBP.
Collapse
Affiliation(s)
- A. R. Caseiro
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
- REQUIMTE/LAQV–U. Porto–Porto/Portugal, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, Porto, Portugal
| | - G. Ivanova
- REQUIMTE- LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre, Porto, Portugal
| | - S. S. Pedrosa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - M. V. Branquinho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - P. Georgieva
- Department of Electronics Telecommunications and Informatics, IEETA, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - P. P. Barbosa
- Biosckin, Molecular and Cell Therapies S.A., Laboratório Criovida, TecMaia, Rua Engenheiro Frederico Ulrich 2650, Moreira da Maia, Portugal
| | - J. D. Santos
- REQUIMTE/LAQV–U. Porto–Porto/Portugal, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, s/n, Porto, Portugal
| | - R. Magalhães
- Universidade Católica Portuguesa, CBQF—Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Rua Arquiteto Lobão Vital 172, Porto, Portugal
| | - P. Teixeira
- Universidade Católica Portuguesa, CBQF—Centro de Biotecnologia e Química Fina–Laboratório Associado, Escola Superior de Biotecnologia, Rua Arquiteto Lobão Vital 172, Porto, Portugal
| | - T. Pereira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
| | - A. C. Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, Portugal
- * E-mail: ,
| |
Collapse
|
8
|
Shaw KA, Parada SA, Gloystein DM, Devine JG. The Science and Clinical Applications of Placental Tissues in Spine Surgery. Global Spine J 2018; 8:629-637. [PMID: 30202718 PMCID: PMC6125928 DOI: 10.1177/2192568217747573] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
STUDY DESIGN Narrative literature review. OBJECTIVES Placental tissue, amniotic/chorionic membrane, and umbilical cord have seen a recent expansion in their clinical application in various fields of surgery. It is important for practicing surgeons to know the underlying science, especially as it relates to spine surgery, to understand the rationale and clinical indication, if any, for their usage. METHODS A literature search was performed using PubMed and MEDLINE databases to identify studies reporting the application of placental tissues as it relates to the practicing spine surgeon. Four areas of interest were identified and a comprehensive review was performed of available literature. RESULTS Clinical application of placental tissue holds promise with regard to treatment of intervertebral disc pathology, preventing epidural fibrosis, spinal dysraphism closure, and spinal cord injury; however, there is an overall paucity of high-quality evidence. As such, evidence-based guidelines for its clinical application are currently unavailable. CONCLUSIONS There is no high-level clinical evidence to support the application of placental tissue for spinal surgery, although it does hold promise for several areas of interest for the practicing spine surgeon. High-quality research is needed to define the clinical effectiveness and indications of placental tissue as it relates to spine surgery.
Collapse
Affiliation(s)
- K. Aaron Shaw
- Dwight D. Eisenhower Army Medical Center, Fort Gordon, GA, USA,K. Aaron Shaw, Department of Orthopaedic Surgery, Dwight D. Eisenhower Army Medical Center, 300 East Hospital Road, Fort Gordon, GA 30905, USA.
| | | | | | - John G. Devine
- Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
9
|
Potential Roles of Dental Pulp Stem Cells in Neural Regeneration and Repair. Stem Cells Int 2018; 2018:1731289. [PMID: 29853908 PMCID: PMC5964589 DOI: 10.1155/2018/1731289] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/22/2018] [Indexed: 12/22/2022] Open
Abstract
This review summarizes current advances in dental pulp stem cells (DPSCs) and their potential applications in the nervous diseases. Injured adult mammalian nervous system has a limited regenerative capacity due to an insufficient pool of precursor cells in both central and peripheral nervous systems. Nerve growth is also constrained by inhibitory factors (associated with central myelin) and barrier tissues (glial scarring). Stem cells, possessing the capacity of self-renewal and multicellular differentiation, promise new therapeutic strategies for overcoming these impediments to neural regeneration. Dental pulp stem cells (DPSCs) derive from a cranial neural crest lineage, retain a remarkable potential for neuronal differentiation, and additionally express multiple factors that are suitable for neuronal and axonal regeneration. DPSCs can also express immunomodulatory factors that stimulate formation of blood vessels and enhance regeneration and repair of injured nerve. These unique properties together with their ready accessibility make DPSCs an attractive cell source for tissue engineering in injured and diseased nervous systems. In this review, we interrogate the neuronal differentiation potential as well as the neuroprotective, neurotrophic, angiogenic, and immunomodulatory properties of DPSCs and its application in the injured nervous system. Taken together, DPSCs are an ideal stem cell resource for therapeutic approaches to neural repair and regeneration in nerve diseases.
Collapse
|
10
|
Devetzi M, Goulielmaki M, Khoury N, Spandidos DA, Sotiropoulou G, Christodoulou I, Zoumpourlis V. Genetically‑modified stem cells in treatment of human diseases: Tissue kallikrein (KLK1)‑based targeted therapy (Review). Int J Mol Med 2018; 41:1177-1186. [PMID: 29328364 PMCID: PMC5819898 DOI: 10.3892/ijmm.2018.3361] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
The tissue kallikrein-kinin system (KKS) is an endogenous multiprotein metabolic cascade which is implicated in the homeostasis of the cardiovascular, renal and central nervous system. Human tissue kallikrein (KLK1) is a serine protease, component of the KKS that has been demonstrated to exert pleiotropic beneficial effects in protection from tissue injury through its anti-inflammatory, anti-apoptotic, anti-fibrotic and anti-oxidative actions. Mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) constitute populations of well-characterized, readily obtainable multipotent cells with special immunomodulatory, migratory and paracrine properties rendering them appealing potential therapeutics in experimental animal models of various diseases. Genetic modification enhances their inherent properties. MSCs or EPCs are competent cellular vehicles for drug and/or gene delivery in the targeted treatment of diseases. KLK1 gene delivery using adenoviral vectors or KLK1 protein infusion into injured tissues of animal models has provided particularly encouraging results in attenuating or reversing myocardial, renal and cerebrovascular ischemic phenotype and tissue damage, thus paving the way for the administration of genetically modified MSCs or EPCs with the human tissue KLK1 gene. Engraftment of KLK1-modified MSCs and/or KLK1-modified EPCs resulted in advanced beneficial outcome regarding heart and kidney protection and recovery from ischemic insults. Collectively, findings from pre-clinical studies raise the possibility that tissue KLK1 may be a novel future therapeutic target in the treatment of a wide range of cardiovascular, cerebrovascular and renal disorders.
Collapse
Affiliation(s)
- Marina Devetzi
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Goulielmaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Nicolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
11
|
Sekuła M, Domalik-Pyzik P, Morawska-Chochół A, Bobis-Wozowicz S, Karnas E, Noga S, Boruczkowski D, Adamiak M, Madeja Z, Chłopek J, Zuba-Surma EK. Polylactide- and polycaprolactone-based substrates enhance angiogenic potential of human umbilical cord-derived mesenchymal stem cells in vitro - implications for cardiovascular repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 77:521-533. [PMID: 28532062 DOI: 10.1016/j.msec.2017.03.281] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/04/2017] [Accepted: 03/29/2017] [Indexed: 01/06/2023]
Abstract
Recent approaches in tissue regeneration focus on combining innovative achievements of stem cell biology and biomaterial sciences to develop novel therapeutic strategies for patients. Growing recent evidence indicates that mesenchymal stem cells harvested from human umbilical cord Wharton's jelly (hUC-MSCs) are a new valuable source of cells for autologous as well as allogeneic therapies in humans. hUC-MSCs are multipotent, highly proliferating cells with prominent immunoregulatory activity. In this study, we evaluated the impact of widely used FDA approved poly(α-esters) including polylactide (PLA) and polycaprolactone (PCL) on selected biological properties of hUC-MSCs in vitro. We found that both polymers can be used as non-toxic substrates for ex vivo propagation of hUC-MSCs as shown by no major impact on cell proliferation or viability. Moreover, PCL significantly enhanced the migratory capacity of hUC-MSCs. Importantly, genetic analysis indicated that both polymers promoted the angiogenic differentiation potential of hUC-MSCs with no additional chemical stimulation. These results indicate that PLA and PCL enhance selected biological properties of hUC-MSCs essential for their regenerative capacity including migratory and proangiogenic potential, which are required for effective vascular repair in vivo. Thus, PLA and PCL-based scaffolds combined with hUC-MSCs may be potentially employed as future novel grafts in tissue regeneration such as blood vessel reconstruction.
Collapse
Affiliation(s)
- Małgorzata Sekuła
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, 7A Gronostajowa St., 30-387 Krakow, Poland
| | - Patrycja Domalik-Pyzik
- Department of Biomaterials, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, 30 Mickiewicza Ave., 30-059 Krakow, Poland
| | - Anna Morawska-Chochół
- Department of Biomaterials, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, 30 Mickiewicza Ave., 30-059 Krakow, Poland
| | - Sylwia Bobis-Wozowicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, 7A Gronostajowa St., 30-387 Krakow, Poland
| | - Sylwia Noga
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, 7A Gronostajowa St., 30-387 Krakow, Poland
| | | | - Marta Adamiak
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland
| | - Jan Chłopek
- Department of Biomaterials, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, 30 Mickiewicza Ave., 30-059 Krakow, Poland.
| | - Ewa K Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Krakow, Poland.
| |
Collapse
|
12
|
Carnevale G, Pisciotta A, Riccio M, Bertoni L, De Biasi S, Gibellini L, Zordani A, Cavallini GM, La Sala GB, Bruzzesi G, Ferrari A, Cossarizza A, de Pol A. Human dental pulp stem cells expressing STRO-1, c-kit and CD34 markers in peripheral nerve regeneration. J Tissue Eng Regen Med 2017; 12:e774-e785. [PMID: 27943583 DOI: 10.1002/term.2378] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 10/10/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022]
Abstract
Peripheral nerve injuries are a commonly encountered clinical problem and often result in long-term functional defects. The application of stem cells able to differentiate in Schwann cell-like cells in vitro and in vivo, could represent an attractive therapeutic approach for the treatment of nerve injuries. Further, stem cells sources sharing the same embryological origin as Schwann cells might be considered a suitable tool. The aim of this study was to demonstrate the ability of a neuroectodermal subpopulation of human STRO-1+ /c-Kit+ /CD34+ DPSCs, expressing P75NTR , nestin and SOX-10, to differentiate into Schwann cell-like cells in vitro and to promote axonal regeneration in vivo, which led to functional recovery as measured by sustained gait improvement, in animal rat model of peripheral nerve injury. Transplanted human dental pulp stem cells (hDPSCs) engrafted into sciatic nerve defect, as revealed by the positive staining against human nuclei, showed the expression of typical Schwann cells markers, S100b and, noteworthy, a significant number of myelinated axons was detected. Moreover, hDPSCs promoted axonal regeneration from proximal to distal stumps 1 month after transplantation. This study demonstrates that STRO-1+ /c-Kit+ /CD34+ hDPSCs, associated with neural crest derivation, represent a promising source of stem cells for the treatment of demyelinating disorders and might provide a valid alternative tool for future clinical applications to achieve functional recovery after injury or peripheral neuropathies besides minimizing ethical issues. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Pisciotta
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Riccio
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Bertoni
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Lara Gibellini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessio Zordani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gian Maria Cavallini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giacomo Bruzzesi
- Oro-Maxillo-Facial Department, AUSL Baggiovara, Baggiovara, Modena, Italy
| | - Adriano Ferrari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy.,Children Rehabilitation Special Unit, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Andrea Cossarizza
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Anto de Pol
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
13
|
Ribeiro J, Caseiro AR, Pereira T, Armada-da-Silva PA, Pires I, Prada J, Amorim I, Leal Reis I, Amado S, Santos JD, Bompasso S, Raimondo S, Varejão ASP, Geuna S, Luís AL, Maurício AC. Evaluation of PVA biodegradable electric conductive membranes for nerve regeneration in axonotmesis injuries: the rat sciatic nerve animal model. J Biomed Mater Res A 2017; 105:1267-1280. [PMID: 28078802 DOI: 10.1002/jbm.a.35998] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/13/2016] [Accepted: 01/05/2017] [Indexed: 11/06/2022]
Abstract
The therapeutic effect of three polyvinyl alcohol (PVA) membranes loaded with electrically conductive materials - carbon nanotubes (PVA-CNTs) and polypyrrole (PVA-PPy) - were tested in vivo for neuro-muscular regeneration after an axonotmesis injury in the rat sciatic nerve. The membranes electrical conductivity measured was 1.5 ± 0.5 × 10-6 S/m, 579 ± 0.6 × 10-6 S/m, and 1837.5 ± 0.7 × 10-6 S/m, respectively. At week-12, a residual motor and nociceptive deficit were present in all treated groups, but at week-12, a better recovery to normal gait pattern of the PVA-CNTs and PVA-PPy treated groups was observed. Morphometrical analysis demonstrated that PVA-CNTs group presented higher myelin thickness and lower g-ratio. The tibialis anterior muscle, in the PVA-PPy and PVA-CNTs groups showed a 9% and 19% increase of average fiber size area and a 5% and 10% increase of the "minimal Feret's diameter," respectively. No inflammation, degeneration, fibrosis or necrosis were detected in lung, liver, kidneys, spleen, and regional lymph nodes and absence of carbon deposits was confirmed with Von Kossa and Masson-Fontana stains. In conclusion, the membranes of PVA-CNTs and PVA-PPy are biocompatible and have electrical conductivity. The higher electrical conductivity measured in PVA-CNTs membrane might be responsible for the positive results on maturation of myelinated fibers. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1267-1280, 2017.
Collapse
Affiliation(s)
- Jorge Ribeiro
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal.,UPVET, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal
| | - Ana Rita Caseiro
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal.,CEMUC, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
| | - Tiago Pereira
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
| | - Paulo Alexandre Armada-da-Silva
- Faculdade de Motricidade Humana (FMH), Universidade de Lisboa (ULisboa), Estrada da Costa, 1499-002, Dafundo, Cruz Quebrada, Portugal.,CIPER-FMH: Centro Interdisciplinar de Estudo de Performance Humana, Faculdade de Motricidade Humana (FMH), Universidade de Lisboa (ULisboa), Estrada da Costa, 1499-002, Cruz Quebrada - Dafundo, Portugal
| | - Isabel Pires
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.,CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, Vila Real, 5000-801, Portugal
| | - Justina Prada
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.,CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, Vila Real, 5000-801, Portugal
| | - Irina Amorim
- Departmento de Patologia e de Imunologia Molecular, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, 4200-465, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto (UP), Rua Alfredo Allen, Porto, 4200-135, Portugal
| | - Inês Leal Reis
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
| | - Sandra Amado
- Instituto Politécnico de Leiria, UIS-IPL: Unidade de Investigação em Saúde da Escola Superior de Saúde de Leiria, Portugal.,CDrsp - Centre for Rapid and Sustainable Product Development, Rua de Portugal 2430-028, Marinha, Grande, Portugal
| | - José Domingos Santos
- CEMUC, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, Porto, 4200-465, Portugal
| | - Simone Bompasso
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10126, Italy.,Neuroscience Institute of the Cavalieri Ottolenghi Foundation (NICO), Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Regione Gonzole 10, Orbassano, 10043, Turin, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10126, Italy.,Neuroscience Institute of the Cavalieri Ottolenghi Foundation (NICO), Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Regione Gonzole 10, Orbassano, 10043, Turin, Italy
| | - Artur Severo Proença Varejão
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal.,CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, Vila Real, 5000-801, Portugal
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, University of Turin, Turin, 10126, Italy.,Neuroscience Institute of the Cavalieri Ottolenghi Foundation (NICO), Azienda Ospedaliero-Universitaria San Luigi Gonzaga, Regione Gonzole 10, Orbassano, 10043, Turin, Italy
| | - Ana Lúcia Luís
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal.,UPVET, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal
| | - Ana Colette Maurício
- Departmento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, Porto, 4050-313, Portugal.,Sub-inidade de Cirurgia Experimental e Medicina Regenerativa, Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, Porto, 4051-401, Portugal
| |
Collapse
|
14
|
Zarbakhsh S, Goudarzi N, Shirmohammadi M, Safari M. Histological Study of Bone Marrow and Umbilical Cord Stromal Cell Transplantation in Regenerating Rat Peripheral Nerve. CELL JOURNAL 2016; 17:668-77. [PMID: 26862526 PMCID: PMC4746417 DOI: 10.22074/cellj.2016.3839] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/05/2015] [Indexed: 12/15/2022]
Abstract
Objective Bone marrow and umbilical cord stromal cells are multipotential stem cells
that have the ability to produce growth factors that play an important role in survival and
generation of axons. The goal of this study was to evaluate the effects of the two different
mesenchymal stem cells on peripheral nerve regeneration.
Materials and Methods In this experimental study, a 10 mm segment of the left sciatic
nerve of male Wistar rats (250-300 g) was removed with a silicone tube interposed into
this nerve gap. Bone marrow stromal cells (BMSCs) and human umbilical cord stromal
cells (HUCSCs) were respectively obtained from rat and human. The cells were sepa-
rately cultured and transplanted into the nerve gap. The sciatic nerve regeneration was
evaluated by immunohistochemistry, and light and electron microscopy. Moreover, histo-
morphology of the gastrocnemius muscle was observed.
Results The nerve regeneration in the BMSCs and HUCSCs groups that had received
the stem cells was significantly more favorable than the control group. In addition, the BM-
SCs group was significantly more favorable than the HUCSCs group (P<0.05).
Conclusion The results of this study suggest that both homograft BMSCs and het-
erograft HUCSCs may have the potential to regenerate peripheral nerve injury and
transplantation of BMSCs may be more effective than HUCSCs in rat.
Collapse
Affiliation(s)
- Sam Zarbakhsh
- Research Center of Nervous System Stem Cells, Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Nasim Goudarzi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shirmohammadi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Manouchehr Safari
- Research Center of Nervous System Stem Cells, Department of Anatomy, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
15
|
Neuromuscular Regeneration: Perspective on the Application of Mesenchymal Stem Cells and Their Secretion Products. Stem Cells Int 2016; 2016:9756973. [PMID: 26880998 PMCID: PMC4736584 DOI: 10.1155/2016/9756973] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/12/2015] [Accepted: 11/16/2015] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells are posing as a promising character in the most recent therapeutic strategies and, since their discovery, extensive knowledge on their features and functions has been gained. In recent years, innovative sources have been disclosed in alternative to the bone marrow, conveying their associated ethical concerns and ease of harvest, such as the umbilical cord tissue and the dental pulp. These are also amenable of cryopreservation and thawing for desired purposes, in benefit of the donor itself or other patients in pressing need. These sources present promising possibilities in becoming useful cell sources for therapeutic applications in the forthcoming years. Effective and potential applications of these cellular-based strategies for the regeneration of peripheral nerve are overviewed, documenting recent advances and identified issues for this research area in the near future. Finally, besides the differentiation capacities attributed to mesenchymal stem cells, advances in the recognition of their effective mode of action in the regenerative theatre have led to a new area of interest: the mesenchymal stem cells' secretome. The paracrine modulatory pathway appears to be a major mechanism by which these are beneficial to nerve regeneration and comprehension on the specific growth factors, cytokine, and extracellular molecules secretion profiles is therefore of great interest.
Collapse
|
16
|
Hosseini SM, Vasaghi A, Nakhlparvar N, Roshanravan R, Talaei-Khozani T, Razi Z. Differentiation of Wharton's jelly mesenchymal stem cells into neurons in alginate scaffold. Neural Regen Res 2015; 10:1312-6. [PMID: 26487861 PMCID: PMC4590246 DOI: 10.4103/1673-5374.162768] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alginate scaffold has been considered as an appropriate biomaterial for promoting the differentiation of embryonic stem cells toward neuronal cell lineage. We hypothesized that alginate scaffold is suitable for culturing Wharton's jelly mesenchymal stem cells (WJMSCs) and can promote the differentiation of WJMSCs into neuron-like cells. In this study, we cultured WJMSCs in a three-dimensional scaffold fabricated by 0.25% alginate and 50 mM CaCl2 in the presence of neurogenic medium containing 10 μM retinoic acid and 20 ng/mL basic fibroblast growth factor. These cells were also cultured in conventional two-dimensional culture condition in the presence of neurogenic medium as controls. After 10 days, immunofluorescence staining was performed for detecting β-tubulin (marker for WJMSCs-differentiated neuron) and CD271 (motor neuron marker). β-Tubulin and CD271 expression levels were significantly greater in the WJMSCs cultured in the three-dimensional alginate scaffold than in the conventional two-dimensional culture condition. These findings suggest that three-dimensional alginate scaffold cell culture system can induce neuronal differentiation of WJMSCs effectively.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran ; Stem Cell Laboratory, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Attiyeh Vasaghi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Newsha Nakhlparvar
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran ; Cell and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Roshanravan
- Colorectal Research Center, Department of Surgery, Shiraz University of Medical Science, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Tissue Engineering Laboratory, Department of Tissue Engineering, School of Advanced Medical Science and Technology, Shiraz University of Medical Sciences, Shiraz, Iran ; Laboratory for Stem Cell Research, Department of Anatomy, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Razi
- Cell and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran ; Department of Medical Physics, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Ribeiro J, Pereira T, Caseiro AR, Armada-da-Silva P, Pires I, Prada J, Amorim I, Amado S, França M, Gonçalves C, Lopes MA, Santos JD, Silva DM, Geuna S, Luís AL, Maurício AC. Evaluation of biodegradable electric conductive tube-guides and mesenchymal stem cells. World J Stem Cells 2015; 7:956-975. [PMID: 26240682 PMCID: PMC4515438 DOI: 10.4252/wjsc.v7.i6.956] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/19/2015] [Accepted: 05/06/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the therapeutic effect of three tube-guides with electrical conductivity associated to mesenchymal stem cells (MSCs) on neuro-muscular regeneration after neurotmesis.
METHODS: Rats with 10-mm gap nerve injury were tested using polyvinyl alcohol (PVA), PVA-carbon nanotubes (CNTs) and MSCs, and PVA-polypyrrole (PPy). The regenerated nerves and tibialis anterior muscles were processed for stereological studies after 20 wk. The functional recovery was assessed serially for gait biomechanical analysis, by extensor postural thrust, sciatic functional index and static sciatic functional index (SSI), and by withdrawal reflex latency (WRL). In vitro studies included cytocompatibility, flow cytometry, reverse transcriptase polymerase chain reaction and karyotype analysis of the MSCs. Histopathology of lung, liver, kidneys, and regional lymph nodes ensured the biomaterials biocompatibility.
RESULTS: SSI remained negative throughout and independently from treatment. Differences between treted groups in the severity of changes in WRL existed, showing a faster regeneration for PVA-CNTs-MSCs (P < 0.05). At toe-off, less acute ankle joint angles were seen for PVA-CNTs-MSCs group (P = 0.051) suggesting improved ankle muscles function during the push off phase of the gait cycle. In PVA-PPy and PVA-CNTs groups, there was a 25% and 42% increase of average fiber area and a 13% and 21% increase of the “minimal Feret’s diameter” respectively. Stereological analysis disclosed a significantly (P < 0.05) increased myelin thickness (M), ratio myelin thickness/axon diameter (M/d) and ratio axon diameter/fiber diameter (d/D; g-ratio) in PVA-CNT-MSCs group (P < 0.05).
CONCLUSION: Results revealed that treatment with MSCs and PVA-CNTs tube-guides induced better nerve fiber regeneration. Functional and kinematics analysis revealed positive synergistic effects brought by MSCs and PVA-CNTs. The PVA-CNTs and PVA-PPy are promising scaffolds with electric conductive properties, bio- and cytocompatible that might prevent the secondary neurogenic muscular atrophy by improving the reestablishment of the neuro-muscular junction.
Collapse
|
18
|
Guo ZY, Sun X, Xu XL, Zhao Q, Peng J, Wang Y. Human umbilical cord mesenchymal stem cells promote peripheral nerve repair via paracrine mechanisms. Neural Regen Res 2015; 10:651-8. [PMID: 26170829 PMCID: PMC4424761 DOI: 10.4103/1673-5374.155442] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2015] [Indexed: 12/17/2022] Open
Abstract
Human umbilical cord-derived mesenchymal stem cells (hUCMSCs) represent a promising young-state stem cell source for cell-based therapy. hUCMSC transplantation into the transected sciatic nerve promotes axonal regeneration and functional recovery. To further clarify the paracrine effects of hUCMSCs on nerve regeneration, we performed human cytokine antibody array analysis, which revealed that hUCMSCs express 14 important neurotrophic factors. Enzyme-linked immunosorbent assay and immunohistochemistry showed that brain-derived neurotrophic factor, glial-derived neurotrophic factor, hepatocyte growth factor, neurotrophin-3, basic fibroblast growth factor, type I collagen, fibronectin and laminin were highly expressed. Treatment with hUCMSC-conditioned medium enhanced Schwann cell viability and proliferation, increased nerve growth factor and brain-derived neurotrophic factor expression in Schwann cells, and enhanced neurite growth from dorsal root ganglion explants. These findings suggest that paracrine action may be a key mechanism underlying the effects of hUCMSCs in peripheral nerve repair.
Collapse
Affiliation(s)
- Zhi-Yuan Guo
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xun Sun
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xiao-Long Xu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qing Zhao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China ; The Neural Regeneration Co-innovation Center of Jiangsu Province, Nantong, Jiangsu Province, China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China ; The Neural Regeneration Co-innovation Center of Jiangsu Province, Nantong, Jiangsu Province, China
| |
Collapse
|
19
|
Askari N, Yaghoobi MM, Shamsara M, Esmaeili-Mahani S. Human Dental Pulp Stem Cells Differentiate into Oligodendrocyte Progenitors Using the Expression of Olig2 Transcription Factor. Cells Tissues Organs 2015; 200:93-103. [PMID: 25966902 DOI: 10.1159/000381668] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2015] [Indexed: 12/20/2022] Open
Abstract
The helix-loop-helix transcription factor Olig2 is essential for lineage determination of oligodendrocytes. Differentiation of stem cells into oligodendrocytes and transplanting them is a novel strategy for the repair of different demyelination diseases. Dental pulp stem cells (DPSCs) are of great interest in regenerative medicine due to their potential for repairing damaged tissues. In this study, DPSCs were isolated from human third molars and transfected with the human Olig2 gene as a differentiation inducer for the oligodendrogenic pathway. Following the differentiation procedure, the expression of Sox2, NG2, PDGFRα, Nestin, MBP, Olig2, Oct4, glial fibrillary acidic protein and A2B5 as stage-specific markers was studied by real-time RT-qPCR, immunocytochemistry and Western blot analysis. The cells were transplanted into a mouse model of local sciatic damage by lysolecithin as a model for demyelination. Oligodendrocyte progenitor cells (OPCs) actively remyelinated and recovered the lysolecithin-induced damages in the sciatic nerve as revealed by treadmill exercise, the von Frey filament test and hind paw withdrawal in response to a thermal stimulus. Recovery of behavioral reflexes occurred 2-6 weeks after OPC transplantation. The results demonstrate that the expression of Olig2 in DPSCs reduces the expression of stem cell markers and induces the development of oligodendrocyte progenitors as revealed by the emergence of oligodendrocyte markers. DPSCs could be programmed into oligodendrocyte progenitors and considered as a simple and valuable source for the cell therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nahid Askari
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | | | | |
Collapse
|
20
|
Neuro-muscular Regeneration Using Scaffolds with Mesenchymal Stem Cells (MSCs) Isolated from Human Umbilical Cord Wharton's Jelly: Functional and Morphological Analysis Using Rat Sciatic Nerve Neurotmesis Injury Model. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.proeng.2015.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Garzón I, Alfonso-Rodríguez CA, Martínez-Gómez C, Carriel V, Martin-Piedra MA, Fernández-Valadés R, Sánchez-Quevedo MC, Alaminos M. Expression of epithelial markers by human umbilical cord stem cells. A topographical analysis. Placenta 2014; 35:994-1000. [PMID: 25284359 DOI: 10.1016/j.placenta.2014.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 09/03/2014] [Accepted: 09/13/2014] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Human umbilical cord stem cells have inherent differentiation capabilities and potential usefulness in regenerative medicine. However, the epithelial differentiation capability and the heterogeneity of these cells have not been fully explored to the date. METHODS We analyzed the expression of several undifferentiation and epithelial markers in cells located in situ in different zones of the umbilical cord -in situ analysis- and in primary ex vivo cell cultures of Wharton's jelly stem cells by microarray and immunofluorescence. RESULTS Our results demonstrated that umbilical cord cells were heterogeneous and had intrinsic capability to express in situ stem cell markers, CD90 and CD105 and the epithelial markers cytokeratins 3, 4, 7, 8, 12, 13, 19, desmoplakin and zonula occludens 1 as determined by microarray and immunofluorescence, and most of these markers remained expressed after transferring the cells from the in situ to the ex vivo cell culture conditions. However, important differences were detected among some cell types in the umbilical cord, with subvascular zone cells showing less expression of stem cell markers and cells in Wharton's jelly and the amnioblastic zones showing the highest expression of stem cells and epithelial markers. CONCLUSIONS These results suggest that umbilical cord mesenchymal cells have intrinsic potential to express relevant epithelial markers, and support the idea that they could be used as alternative cell sources for epithelial tissue engineering.
Collapse
Affiliation(s)
- I Garzón
- Department of Histology (Tissue Engineering Group), University of Granada, Spain; Instituto de Investigación Biosanitaria ibs. Granada, Spain
| | - C A Alfonso-Rodríguez
- Department of Histology (Tissue Engineering Group), University of Granada, Spain; Instituto de Investigación Biosanitaria ibs. Granada, Spain; PhD programme in Biomedicine, University of Granada, Spain
| | - C Martínez-Gómez
- PhD programme in Clinical Medicine and Public Health, University of Granada, Spain
| | - V Carriel
- Department of Histology (Tissue Engineering Group), University of Granada, Spain; Instituto de Investigación Biosanitaria ibs. Granada, Spain
| | - M A Martin-Piedra
- Department of Histology (Tissue Engineering Group), University of Granada, Spain; Instituto de Investigación Biosanitaria ibs. Granada, Spain
| | - R Fernández-Valadés
- Instituto de Investigación Biosanitaria ibs. Granada, Spain; Division of Pediatric Surgery, University Hospital Virgen de las Nieves, Granada, Spain
| | - M C Sánchez-Quevedo
- Department of Histology (Tissue Engineering Group), University of Granada, Spain; Instituto de Investigación Biosanitaria ibs. Granada, Spain
| | - M Alaminos
- Department of Histology (Tissue Engineering Group), University of Granada, Spain; Instituto de Investigación Biosanitaria ibs. Granada, Spain.
| |
Collapse
|