101
|
Herrera LC, Shastri VP. Silencing of GFP expression in human mesenchymal stem cells using quaternary polyplexes of siRNA-PEI with glycosaminoglycans and albumin. Acta Biomater 2019; 99:397-411. [PMID: 31541736 DOI: 10.1016/j.actbio.2019.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/24/2019] [Accepted: 09/08/2019] [Indexed: 12/19/2022]
Abstract
In recent years evidence has been mounting for a role for mesenchymal stem cells (MSCs) in immunomodulation, anti-inflammatory processes, and paracrine signaling via secreted extracellular vesicles. In order to exploit these biological functions, systems to efficiently deliver genetic material into MSCs would therefore be highly desirable. In this study, efficient silencing of GFP expression by combining high N/P ratio siRNA and branched PEI (bPEI) polyplexes (siRNA-bPEI) polyplexes with glycosaminoglycans (GAGs), namely hyaluronic acid (HA), chondroitin sulfate (CS) and heparin sulfate (HS), and human serum albumin (HSA) is reported. These quaternary systems were characterized using surface charge, size and morphology and applied to MSCs, which represent a challenge due to their typically low transfection efficiency. The quaternary polyplexes promoted efficient charge shielding and release of siRNA in the cytoplasm with reduced toxicity. A high silencing efficiency of >90% (i.e., less than 10% remaining GFP expression) was achieved with noticeably reduced cellular toxicity, especially with siRNA-bPEI polyplexes modified with HA and HA + HSA. In general addition of GAGs led to more compact polyplexes. Endocytosis studies point to improved endosomal escape at high N/P ratios as a reason for high transfection efficiency and a role for hyaluronic acid in the uptake mechanism likely via CD44 interactions. Co-localization studies showed the polyplexes are stable in the cytosol over time, which correlates with a proper disassembly and subsequent silencing of GFP. Furthermore, GAG containing polyplexes were frequently co-localized with the nucleus. These findings in sum suggest that PEI/HSA/GAG based quaternary polyplexes are promising as transfection agents for MSCs. STATEMENT OF SIGNIFICANCE: Since mesenchymal stem cells (MSCs) are recruited to the site of tissue repair and play a role in immunomodulation, anti-inflammatory processes, and paracrine signaling, they present an excellent target for genetic engineering. However, delivery of genetic material into MSCs is challenging. In this study, >97% silencing of constitutive green fluorescent protein expression in human MSCs (hMSCs) using high N/P ratio polyplexes of branched-PEI-siRNA incorporating glycosaminoglycan as a charge neutralizer and human serum albumin as co-complexing agent is demonstrated. In addition to possessing good cytocompatibility and excellent cytosolic stability; polyplexes incorporating GAGs also showed altered endocytic uptake, with incorporation of hyaluronic acid promoting caveolae-mediated entry. Our system highlights the importance of physiologically derived macromolecules in delivery of genetic material into hMSCs.
Collapse
Affiliation(s)
- Laura C Herrera
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany
| | - V Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
102
|
Ashammakhi N, Kim HJ, Ehsanipour A, Bierman RD, Kaarela O, Xue C, Khademhosseini A, Seidlits SK. Regenerative Therapies for Spinal Cord Injury. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:471-491. [PMID: 31452463 DOI: 10.1089/ten.teb.2019.0182] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spinal cord injury (SCI) is a serious problem that primarily affects younger and middle-aged adults at its onset. To date, no effective regenerative treatment has been developed. Over the last decade, researchers have made significant advances in stem cell technology, biomaterials, nanotechnology, and immune engineering, which may be applied as regenerative therapies for the spinal cord. Although the results of clinical trials using specific cell-based therapies have proven safe, their efficacy has not yet been demonstrated. The pathophysiology of SCI is multifaceted, complex and yet to be fully understood. Thus, combinatorial therapies that simultaneously leverage multiple approaches will likely be required to achieve satisfactory outcomes. Although combinations of biomaterials with pharmacologic agents or cells have been explored, few studies have combined these modalities in a systematic way. For most strategies, clinical translation will be facilitated by the use of minimally invasive therapies, which are the focus of this review. In addition, this review discusses previously explored therapies designed to promote neuroregeneration and neuroprotection after SCI, while highlighting present challenges and future directions. Impact Statement To date there are no effective treatments that can regenerate the spinal cord after injury. Although there have been significant preclinical advances in bioengineering and regenerative medicine over the last decade, these have not translated into effective clinical therapies for spinal cord injury. This review focuses on minimally invasive therapies, providing extensive background as well as updates on recent technological developments and current clinical trials. This review is a comprehensive resource for researchers working towards regenerative therapies for spinal cord injury that will help guide future innovation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland.,Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Han-Jun Kim
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | | | | | - Outi Kaarela
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Chengbin Xue
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, P.R. China
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.,Center of Nanotechnology, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Chemical and Biological Engineering, University of California, Los Angeles, California
| | - Stephanie K Seidlits
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.,Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, California.,Brain Research Institute, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
103
|
Glenske K, Schuler G, Arnhold S, Elashry MI, Wagner AS, Barbeck M, Neumann E, Müller-Ladner U, Schnettler R, Wenisch S. Effects of testosterone and 17β-estradiol on osteogenic and adipogenic differentiation capacity of human bone-derived mesenchymal stromal cells of postmenopausal women. Bone Rep 2019; 11:100226. [PMID: 31709277 PMCID: PMC6833309 DOI: 10.1016/j.bonr.2019.100226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/11/2019] [Accepted: 10/02/2019] [Indexed: 12/31/2022] Open
Abstract
Progressive bone loss is a predominant symptom of aging and osteoporosis. Therefore, the effects of sex steroids (i.e. testosterone and 17β-estradiol) on the differentiation capacity of human bone-derived mesenchymal stromal cells (hMSCs), as progenitors of osteoblasts and adipocytes, are of particular interest. The objectives of the present study were, thus, to elucidate whether bone-derived hMSCs of postmenopausal women produce aromatase (CYP19A1) and, whether they modulate their differentiation behaviour in response to testosterone and 17β-estradiol (E2), in relation to their steroid receptor expression. Supplementation of testosterone resulted in a considerable formation of E2 under osteogenic and adipogenic culture conditions, whereas E2 synthesis remained minimal in the cells cultured in basal medium. Concomitant with high aromatase expression and 17β-estradiol formation of the cells cultured in osteogenic medium supplemented with testosterone, a distinct promotion of late-stage osteogenesis was found, as shown by significant matrix mineralization and a notable increase in osteogenic markers. These effects were abrogated by the aromatase inhibitor anastrozole. Under adipogenic conditions, testosterone reduced the occurrence of lipid droplets and led to a decrease in PPARγ and AR expression, independent of anastrozole. Regardless of the culture conditions, ERα was detectable whilst ERβ was not. In conclusion, aromatase activity is limited to differentiated hMSCs and the resulting 17β-estradiol enhances late osteogenic differentiation stages via ERα. Adipogenic differentiation, on the other hand, is reduced by both sex steroids: testosterone via AR and 17β-estradiol.
Collapse
Affiliation(s)
- Kristina Glenske
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, 35392, Germany
| | - Gerhard Schuler
- Veterinary Clinic of Obstetrics, Gynecology and Andrology, Justus-Liebig-University of Giessen, Giessen, 35392, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, 35392, Germany
| | - Mohamed I Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, 35392, Germany.,Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, 35516, Egypt
| | - Alena-Svenja Wagner
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, 35392, Germany
| | - Mike Barbeck
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine University Hospital Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Elena Neumann
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik, Bad Nauheim, 61231, Germany
| | - Ulf Müller-Ladner
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Kerckhoff-Klinik, Bad Nauheim, 61231, Germany
| | - Reinhard Schnettler
- Department of Oral and Maxillofacial Surgery, Division for Regenerative Orofacial Medicine University Hospital Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, 35392, Germany
| |
Collapse
|
104
|
Luna GLF, Russo TL, Sabadine MA, Estrada‐Bonilla YC, Andrade ALM, Brassolatti P, Anibal FF, Leal ÂMO. Effects of mesenchymal stromal cells on motor function and collagen in the skeletal muscles of rats with type I diabetes. Int J Exp Pathol 2019; 100:359-368. [PMID: 32026546 PMCID: PMC7042733 DOI: 10.1111/iep.12340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/12/2022] Open
Abstract
The present study aimed to evaluate the effects of mesenchymal stromal cell (MSC) transplantation on motor function and collagen organization in the muscles of rats with type 1 diabetes mellitus. Male Wistar rats were randomly assigned to three groups: control (C), diabetic (DM) and diabetic treated with MSCs (DM-MSCs). Diabetes was induced by streptozotocin (50 µg/kg). Bone marrow cells were isolated from the tibia and femur. After 10 weeks of DM induction, the DM-MSC rats received four i.p. injections of MSCs (1 × 106). Ten weeks after MSC transplantation, motor performance was evaluated by the rotarod test and the anterior tibial (TA) muscles were collected for morphometric and quantification of collagen birefringence by polarizing microscopy analysis. Motor performance of the DM group was significantly reduced when compared to the C group and increased significantly in the DM + MSC group. The TA muscle mass was significantly reduced in the DM and DM + MSC groups compared to the C group. The connective tissue increased in the DM group compared to the C group and decreased in the DM + MSC group. The percentage collagen birefringence decreased significantly in the DM group when compared to the C group and increased in the DM + MSC group. Motor performance was positively correlated with collagen birefringence and negatively correlated with percentage of connective tissue. The results indicate that MSC transplantation improves both motor function and the collagen macromolecular organization in type 1 DM.
Collapse
Affiliation(s)
- Genoveva L. F. Luna
- Department of MedicineFederal University of São Carlos (UFSCar)São CarlosBrazil
| | - Thiago L. Russo
- Department of Physical TherapyFederal University of São CarlosSão CarlosBrazil
| | - Maria A. Sabadine
- Department of MedicineFederal University of São Carlos (UFSCar)São CarlosBrazil
| | | | - Ana L. M. Andrade
- Department of Physical TherapyFederal University of São CarlosSão CarlosBrazil
| | - Patricia Brassolatti
- Department of Morphology and PathologyFederal University of São CarlosSão CarlosBrazil
| | - Fernanda F. Anibal
- Department of Morphology and PathologyFederal University of São CarlosSão CarlosBrazil
| | - Ângela M. O. Leal
- Department of Physical TherapyFederal University of São CarlosSão CarlosBrazil
| |
Collapse
|
105
|
High-throughput RNA-sequencing identifies mesenchymal stem cell-induced immunological signature in a rat model of corneal allograft rejection. PLoS One 2019; 14:e0222515. [PMID: 31545822 PMCID: PMC6756551 DOI: 10.1371/journal.pone.0222515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022] Open
Abstract
Objective The immune rejection mediated by CD4+ T cell and antigen presenting macrophages is the leading cause of corneal transplantation failure. Bone marrow-derived mesenchymal stem cells (BM-MSCs) possess robust immunomodulatory potentials, and have been shown by us and others to promote corneal allograft survival. However, the immunological mechanism underlying the protective effects of BM-MSCs remains unclear. Therefore, in the current study, this mechanism was investigated in a BM-MSC-treated rat model of corneal allograft rejection, in the hope to facilitate the search for novel interventional targets to corneal allograft rejection. Methods Lewis rats were subjected to corneal transplantation and then received subconjunctival injections of BM-MSCs (2×106 cells / 100 μl PBS) immediately and at day 3 post-transplantation. The control group received the injections of PBS with the same volume. The clinical parameters of the corneal allografts, including opacity, edema, and neovascularization, were regularly evaluated after transplantation. On day 10 post-transplantation, the corneal allografts were collected and subjected to flow cytometry and high-throughput RNA sequencing (RNA-seq). GO enrichment and KEGG pathways were analyzed. The quantitative realtime PCR (qPCR) and immunohistochemistry (IHC) were employed to validate the expression of the selected target genes at transcript and protein levels, respectively. Results BM-MSC subconjunctival administration prolonged the corneal allograft survival, with reduced opacity, alleviated edema, and diminished neovascularization. Flow cytometry showed reduced CD4+ T cells and CD68+ macrophages as well as boosted regulatory T cells (Tregs) in the BM-MSC-treated corneal allografts as compared with the PBS-treated counterparts. Moreover, the RNA-seq and qPCR results demonstrated that the transcript abundance of Cytotoxic T-Lymphocyte Associated Protein 4 (Ctla4), Protein Tyrosine Phosphatase, Receptor Type C (Ptprc), and C-X-C Motif Chemokine Ligand 9 (Cxcl9) genes were increased in the allografts of BM-MSC group compared with PBS group; whereas the expression of Heat Shock Protein Family A (Hsp70) Member 8 (Hspa8) gene was downregulated. The expression of these genes was confirmed by IHC at protein level. Conclusion Subconjunctival injections of BM-MSCs promoted corneal allograft survival, reduced CD4+ and CD68+ cell infiltration, and enriched Treg population in the allografts. The BM-MSC-induced upregulation of Ctla4, Ptprc, Cxcl9 genes and downregulation of Hspa8 gene might contribute to the protective effects of BM-MSCs and subserve the potential interventional targets to corneal allograft rejection.
Collapse
|
106
|
Jagiełło J, Sekuła-Stryjewska M, Noga S, Adamczyk E, Dźwigońska M, Kurcz M, Kurp K, Winkowska-Struzik M, Karnas E, Boruczkowski D, Madeja Z, Lipińska L, Zuba-Surma EK. Impact of Graphene-Based Surfaces on the Basic Biological Properties of Human Umbilical Cord Mesenchymal Stem Cells: Implications for Ex Vivo Cell Expansion Aimed at Tissue Repair. Int J Mol Sci 2019; 20:E4561. [PMID: 31540083 PMCID: PMC6770664 DOI: 10.3390/ijms20184561] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 01/20/2023] Open
Abstract
The potential therapeutic applications of mesenchymal stem/stromal cells (MSCs) and biomaterials have attracted a great amount of interest in the field of biomedical engineering. MSCs are multipotent adult stem cells characterized as cells with specific features, e.g., high differentiation potential, low immunogenicity, immunomodulatory properties, and efficient in vitro expansion ability. Human umbilical cord Wharton's jelly-derived MSCs (hUC-MSCs) are a new, important cell type that may be used for therapeutic purposes, i.e., for autologous and allogeneic transplantations. To improve the therapeutic efficiency of hUC-MSCs, novel biomaterials have been considered for use as scaffolds dedicated to the propagation and differentiation of these cells. Nowadays, some of the most promising materials for tissue engineering include graphene and its derivatives such as graphene oxide (GO) and reduced graphene oxide (rGO). Due to their physicochemical properties, they can be easily modified with biomolecules, which enable their interaction with different types of cells, including MSCs. In this study, we demonstrate the impact of graphene-based substrates (GO, rGO) on the biological properties of hUC-MSCs. The size of the GO flakes and the reduction level of GO have been considered as important factors determining the most favorable surface for hUC-MSCs growth. The obtained results revealed that GO and rGO are suitable scaffolds for hUC-MSCs. hUC-MSCs cultured on: (i) a thin layer of GO and (ii) an rGO surface with a low reduction level demonstrated a viability and proliferation rate comparable to those estimated under standard culture conditions. Interestingly, cell culture on a highly reduced GO substrate resulted in a decreased hUC-MSCs proliferation rate and induced cell apoptosis. Moreover, our analysis demonstrated that hUC-MSCs cultured on all the tested GO and rGO scaffolds showed no alterations of their typical mesenchymal phenotype, regardless of the reduction level and size of the GO flakes. Thus, GO scaffolds and rGO scaffolds with a low reduction level exhibit potential applicability as novel, safe, and biocompatible materials for utilization in regenerative medicine.
Collapse
Affiliation(s)
- Joanna Jagiełło
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, 01-919 Warsaw, Poland.
| | | | - Sylwia Noga
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| | - Edyta Adamczyk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| | - Monika Dźwigońska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| | - Magdalena Kurcz
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, 01-919 Warsaw, Poland.
| | - Katarzyna Kurp
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, 01-919 Warsaw, Poland.
| | - Magdalena Winkowska-Struzik
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, 01-919 Warsaw, Poland.
| | - Elżbieta Karnas
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| | | | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| | - Ludwika Lipińska
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, 01-919 Warsaw, Poland.
| | - Ewa K Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| |
Collapse
|
107
|
Airini R, Iordache F, Alexandru D, Savu L, Epureanu FB, Mihailescu D, Amuzescu B, Maniu H. Senescence-induced immunophenotype, gene expression and electrophysiology changes in human amniocytes. J Cell Mol Med 2019; 23:7233-7245. [PMID: 31478614 PMCID: PMC6815807 DOI: 10.1111/jcmm.14495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 12/23/2022] Open
Abstract
The aim of the study was to evidence replicative senescence‐induced changes in human amniocytes via flow cytometry, quantitative reverse‐transcription‐polymerase chain reaction (qRT‐PCR) and automated/manual patch‐clamp. Both cryopreserved and senescent amniocytes cultured in BIO‐AMF‐2 medium featured high percentages of pluripotency cell surface antigens SSEA‐1, SSEA‐4, TRA1‐60, TRA1‐81 (assessed by flow cytometry) and expression of pluripotency markers Oct4 (Pou5f1) and Nanog (by qRT‐PCR). We demonstrated in senescent vs cryopreserved amniocytes decreases in mesenchymal stem cell surface markers. Senescence‐associated β‐galactosidase stained only senescent amniocytes, and they showed no deoxyuridine incorporation. The gene expression profile revealed a secretory phenotype of senescent amniocytes (increased interleukin (IL)‐1α, IL‐6, IL‐8, transforming growth factor β, nuclear factor κB p65 expression), increases for cell cycle‐regulating genes (p16INK4A), cytoskeletal elements (β‐actin); HMGB1, c‐Myc, Bcl‐2 showed reduced changes and p21, MDM2 decreased. Via patch‐clamp we identified five ion current components: outward rectifier K+ current, an inactivatable component, big conductance Ca2+‐dependent K+ channels (BK) current fluctuations, Na+ current, and inward rectifier K+ current. Iberiotoxin 100 nmol/L blocked 71% of BK fluctuations, and lidocaine 200 μmol/L exerted use‐dependent Na+ current block. Transient receptor potential (TRP)M7‐like current density at −120 mV was significantly increased in senescent amniocytes. The proinflammatory profile acquired by senescent amniocytes in vitro may prevent their use in clinical therapies for immunosuppression, antiapoptotic and healing effects.
Collapse
Affiliation(s)
- Razvan Airini
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Florin Iordache
- Department of Regenerative Medicine, "N. Simionescu" Institute of Cell Biology and Pathology, Bucharest, Romania
| | - Dorin Alexandru
- Department of Regenerative Medicine, "N. Simionescu" Institute of Cell Biology and Pathology, Bucharest, Romania
| | - Lorand Savu
- Genetic Lab S.R.L., Bucharest, Romania.,Fundeni Clinical Institute, Bucharest, Romania
| | - Florin Bogdan Epureanu
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Dan Mihailescu
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Bogdan Amuzescu
- Department of Biophysics & Physiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Horia Maniu
- Department of Regenerative Medicine, "N. Simionescu" Institute of Cell Biology and Pathology, Bucharest, Romania
| |
Collapse
|
108
|
Novello S, Debouche A, Philippe M, Naudet F, Jeanne S. Clinical application of mesenchymal stem cells in periodontal regeneration: A systematic review and meta-analysis. J Periodontal Res 2019; 55:1-12. [PMID: 31378933 DOI: 10.1111/jre.12684] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/16/2019] [Accepted: 06/29/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate the potential efficacy of mesenchymal stem cells (MSCs) in periodontal regeneration in humans on the following main outcomes: clinical attachment level (CAL), probing depth (PD), and gingival recession (GR). BACKGROUND The clinical application of stem cells in periodontal regeneration has begun in recent years, but clinical practices are not yet standardized and no recommendations are available at this time. METHODS Electronic database searches and hand searches were conducted. All types of studies, case series, and case reports were qualitatively described. Double-blind randomized controlled trials (RCTs) evaluating MSCs in periodontal regeneration were included in a meta-analysis if they compared administration of MSCs vs application of stem cell-free therapy in the control group, in healthy patients with periodontal defects, with a minimum of three mo of follow-up. RESULTS Fifteen reports were included in qualitative analysis, involving 123 patients and 158 periodontal defects. Only two small RCTs at high risk of bias, with a total of 59 patients and 70 periodontal defects, were included in the meta-analysis. A small but significant difference between test and control groups was found for CAL at three mo (-0.90 mm, 95% CI [-1.51; -0.29]), but not for PD and GR. CONCLUSION Low-quality evidence suggests that MSC-based therapy may have a small impact on periodontal regeneration. However, due to the monocentric character, the small sample size, and potential heterogeneity across the two included RCTs, these results must not be considered as definitive. High-quality RCTs are needed before any clinical use of MSCs in periodontal regeneration.
Collapse
Affiliation(s)
- Solen Novello
- ISCR [(Institut des Sciences Chimiques de Rennes)] - UMR 6226, Univ Rennes, Rennes, France.,Unité de Formation et de Recherche d'Odontologie, Univ Rennes, Rennes, France.,Pôle d'Odontologie, UF Parodontologie, CHU Rennes, Rennes, France
| | - Alexandre Debouche
- Unité de Formation et de Recherche d'Odontologie, Univ Rennes, Rennes, France
| | - Marie Philippe
- Unité de Formation et de Recherche d'Odontologie, Univ Rennes, Rennes, France
| | - Florian Naudet
- CHU Rennes, Inserm, CIC 1414 [(Centre d'Investigation Clinique de Rennes)], Univ Rennes, Rennes, France
| | - Sylvie Jeanne
- ISCR [(Institut des Sciences Chimiques de Rennes)] - UMR 6226, Univ Rennes, Rennes, France.,Unité de Formation et de Recherche d'Odontologie, Univ Rennes, Rennes, France.,Pôle d'Odontologie, UF Parodontologie, CHU Rennes, Rennes, France
| |
Collapse
|
109
|
Batty BS, Bionaz M. Graduate Student Literature Review: The milk behind the mustache: A review of milk and bone biology. J Dairy Sci 2019; 102:7608-7617. [DOI: 10.3168/jds.2019-16421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/19/2019] [Indexed: 12/11/2022]
|
110
|
|
111
|
Caplan H, Olson SD, Kumar A, George M, Prabhakara KS, Wenzel P, Bedi S, Toledano-Furman NE, Triolo F, Kamhieh-Milz J, Moll G, Cox CS. Mesenchymal Stromal Cell Therapeutic Delivery: Translational Challenges to Clinical Application. Front Immunol 2019; 10:1645. [PMID: 31417542 PMCID: PMC6685059 DOI: 10.3389/fimmu.2019.01645] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
For several decades, multipotent mesenchymal stromal cells (MSCs) have been extensively studied for their therapeutic potential across a wide range of diseases. In the preclinical setting, MSCs demonstrate consistent ability to promote tissue healing, down-regulate excessive inflammation and improve outcomes in animal models. Several proposed mechanisms of action have been posited and demonstrated across an array of in vitro models. However, translation into clinical practice has proven considerably more difficult. A number of prominent well-funded late-phase clinical trials have failed, thus calling out for new efforts to optimize product delivery in the clinical setting. In this review, we discuss novel topics critical to the successful translation of MSCs from pre-clinical to clinical applications. In particular, we focus on the major routes of cell delivery, aspects related to hemocompatibility, and potential safety concerns associated with MSC therapy in the different settings.
Collapse
Affiliation(s)
- Henry Caplan
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Akshita Kumar
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mitchell George
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karthik S. Prabhakara
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pamela Wenzel
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Supinder Bedi
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Naama E. Toledano-Furman
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fabio Triolo
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Charles S. Cox
- Department of Pediatric Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
112
|
Vakhrushev IV, Yusupov VI, Raeva OS, Pyatnitskiy MA, Bagratashvili VN. Effect of Low-Level Laser Irradiation on Proliferative Activity of Wharton's Jelly Mesenchymal Stromal Cells. Bull Exp Biol Med 2019; 167:136-139. [PMID: 31183648 DOI: 10.1007/s10517-019-04477-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Indexed: 12/15/2022]
Abstract
We studied the effect of low-level laser irradiation on proliferative activity of cultured human Wharton's jelly mesenchymal stromal sells. Cells were irradiated with a solid-state laser emitting at 650 nm; irradiation doses were 0.04, 0.4, or 4 J/cm2. Laser irradiation was performed once at the start of the cell proliferation experiment or daily throughout the experiment. Cells were cultured for 7 days. The number of viable cells was assessed using the MTT test. An increase in cell proliferative activity was detected after daily laser irradiations; the maximum stimulating effect was achieved at a dose of 0.04 J/cm2. These results substantiate medical use of lasers for expansion of cells intended for transplantation.
Collapse
Affiliation(s)
- I V Vakhrushev
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia. .,Private Institution Laboratory for Biotechnological Research "3D Bioprinting Solutions", Moscow, Russia.
| | - V I Yusupov
- Institute of Photonics Technologies, Federal Research Center for Crystallography and Photonics, Russian Academy of Sciences, Troitsk, Russia
| | - O S Raeva
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - M A Pyatnitskiy
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia
| | - V N Bagratashvili
- Institute of Photonics Technologies, Federal Research Center for Crystallography and Photonics, Russian Academy of Sciences, Troitsk, Russia
| |
Collapse
|
113
|
Liu Q, Zhu Y, Qi J, Amadio PC, Moran SL, Gingery A, Zhao C. Isolation and characterization of turkey bone marrow-derived mesenchymal stem cells. J Orthop Res 2019; 37:1419-1428. [PMID: 30548886 DOI: 10.1002/jor.24203] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/06/2018] [Indexed: 02/04/2023]
Abstract
Flexor tendon injury is often associated with suboptimal outcomes and results in substantial digit dysfunction. Stem cells have been isolated from several experimental animals for the growing interest and needs of utilizing cell-based therapies. Recently, turkey has been developed as a new large animal model for flexor tendon research. In the present study, we reported the isolation and characterization of bone marrow-derived mesenchymal stem cells (BMSCs) from 8- to 12-month-old heritage-breed turkeys. The isolated cells demonstrated fibroblast-like morphology, clonogenic capacity, and high proliferation rate. These cells were positive for surface antigens CD90, CD105, and CD44, but were negative for CD45. The multipotency of turkey BMSCs was determined by differentiating cells into osteogenic, adipogenic, chondrogenic, and tenogenic lineages. There was upregulated gene expression of tenogenic markers, including mohawk, tenomodulin, and EGR1 as well as increased collagen synthesis in BMP12 induced cells. The successful isolation and verification of bone marrow-derived MSCs from turkey would provide opportunities of studying cell-based therapies and developing new treatments for tendon injuries using this novel preclinical large animal model. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1419-1428, 2019.
Collapse
Affiliation(s)
- Qian Liu
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Yaxi Zhu
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jun Qi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Peter C Amadio
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Steven L Moran
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Anne Gingery
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
114
|
Wilson A, Webster A, Genever P. Nomenclature and heterogeneity: consequences for the use of mesenchymal stem cells in regenerative medicine. Regen Med 2019; 14:595-611. [PMID: 31115266 PMCID: PMC7132560 DOI: 10.2217/rme-2018-0145] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are in development for many clinical indications, based both on ‘stem’ properties (tissue repair or regeneration) and on signaling repertoire (immunomodulatory and anti-inflammatory effects). Potential conflation of MSC properties with those of tissue-derived stromal cells presents difficulties in comparing study outcomes and represents a source of confusion in cell therapy development. Cultured MSCs demonstrate significant heterogeneity in clonogenicity and multi-lineage differentiation potential. However in vivo biology of MSCs includes native functions unrelated to regenerative medicine applications, so do nomenclature and heterogeneity matter? In this perspective we examine some consequences of the nomenclature debate and heterogeneity of MSCs. Regulatory expectations are considered, emphasizing that product development should prioritize detailed characterization of therapeutic cell populations for specific indications.
Collapse
Affiliation(s)
- Alison Wilson
- Department of Biology, University of York, York YO10 5DD, UK
| | - Andrew Webster
- Science & Technology Studies Unit, Department of Sociology, University of York, York YO10 5DD, UK
| | - Paul Genever
- Department of Biology, University of York, York YO10 5DD, UK
| |
Collapse
|
115
|
Vaes JEG, Vink MA, de Theije CGM, Hoebeek FE, Benders MJNL, Nijboer CHA. The Potential of Stem Cell Therapy to Repair White Matter Injury in Preterm Infants: Lessons Learned From Experimental Models. Front Physiol 2019; 10:540. [PMID: 31143126 PMCID: PMC6521595 DOI: 10.3389/fphys.2019.00540] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse white matter injury (dWMI) is a major cause of morbidity in the extremely preterm born infant leading to life-long neurological impairments, including deficits in cognitive, motor, sensory, psychological, and behavioral functioning. At present, no treatment options are clinically available to combat dWMI and therefore exploration of novel strategies is urgently needed. In recent years, the pathophysiology underlying dWMI has slowly started to be unraveled, pointing towards the disturbed maturation of oligodendrocytes (OLs) as a key mechanism. Immature OL precursor cells in the developing brain are believed to be highly sensitive to perinatal inflammation and cerebral oxygen fluctuations, leading to impaired OL differentiation and eventually myelination failure. OL lineage development under normal and pathological circumstances and the process of (re)myelination have been studied extensively over the years, often in the context of other adult and pediatric white matter pathologies such as stroke and multiple sclerosis (MS). Various studies have proposed stem cell-based therapeutic strategies to boost white matter regeneration as a potential strategy against a wide range of neurological diseases. In this review we will discuss experimental studies focusing on mesenchymal stem cell (MSC) therapy to reduce white matter injury (WMI) in multiple adult and neonatal neurological diseases. What lessons have been learned from these previous studies and how can we translate this knowledge to application of MSCs for the injured white matter in the preterm infant? A perspective on the current state of stem cell therapy will be given and we will discuss different important considerations of MSCs including cellular sources, timing of treatment and administration routes. Furthermore, we reflect on optimization strategies that could potentially reinforce stem cell therapy, including preconditioning and genetic engineering of stem cells or using cell-free stem cell products, to optimize cell-based strategy for vulnerable preterm infants in the near future.
Collapse
Affiliation(s)
- Josine E G Vaes
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marit A Vink
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Caroline G M de Theije
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Freek E Hoebeek
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Cora H A Nijboer
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
116
|
Lazarević JJ, Kukolj T, Bugarski D, Lazarević N, Bugarski B, Popović ZV. Probing primary mesenchymal stem cells differentiation status by micro-Raman spectroscopy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 213:384-390. [PMID: 30726762 DOI: 10.1016/j.saa.2019.01.069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/21/2019] [Indexed: 05/27/2023]
Abstract
We have employed micro-Raman spectroscopy to get insight into intrinsic biomolecular profile of individual mesenchymal stem cell isolated from periodontal ligament. Furthermore, these cells were stimulated towards adipogenic, chondrogenic, and osteogenic lineages and their status of differentiation was assessed using micro-Raman spectroscopy. In both cases, glass coverslips were used as substrates, due to their wide availability and cost effectiveness. In all sample groups, the same type of behavior was observed, manifested as changes in Raman spectra: the increase of relative intensity of protein/lipid bands and decrease of nucleic acid bands. Comprehensive statistical analysis in the form of principal component analysis was performed, which revealed noticeable grouping of cells with the similar features. Despite the inhomogeneity of primary stem cells and their differentiated lineages, we demonstrated that micro-Raman spectroscopy is sufficient for distinguishing cells' status, which can be valuable for medical and clinical application.
Collapse
Affiliation(s)
- J J Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia
| | - T Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11000, Serbia
| | - D Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade 11000, Serbia
| | - N Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia.
| | - B Bugarski
- Department of Chemical Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, Belgrade 11060, Serbia
| | - Z V Popović
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, Belgrade 11080, Serbia; Serbian Academy of Sciences and Arts, Knez Mihailova 35, Belgrade 11000, Serbia
| |
Collapse
|
117
|
Noguera R, Burgos-Panadero R, Gamero-Sandemetrio E, de la Cruz-Merino L, Álvaro Naranjo T. [An integral view of cancer (I). The study, classification and reprogramming of the tumoral microclimate]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2019; 52:92-102. [PMID: 30902384 DOI: 10.1016/j.patol.2018.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/09/2018] [Accepted: 11/27/2018] [Indexed: 11/16/2022]
Abstract
The group of diseases that we call cancer share a biological structure formed by a complex ecosystem, with altered intercellular communication, information fields, development and tissue function. Beyond the genetic alterations of the tumor cell, the demonstration of an altered ecosystem, with interconnections at systemic levels, opens up a new perspective on cancer biology and behavior. Different tumor facets, such as morphology, classification, clinical aggressiveness, prognosis and response to treatment now appear under a comprehensive vision that offers a new horizon of study, research and clinical management. The Somatic Mutation Theory in cancer, in force for more than one hundred years, is now completed by the study of the tumor microenvironment, the extracellular matrix, the stromal cells, the immune response, the innervation, the nutrition, the mitochondria, the metabolism, the interstitial fluid, the mechanical and electromagnetic properties of the tissue and many other areas of emerging knowledge; thus opening the door to a reprogramming exercise of the tumor phenotype through the modification of the keys offered by this new paradigm. Its recognition makes it possible to go from considering the oncological process as a cellular problem to a supracellular alteration based on the disorganization of tissues, immersed in the relationships of the complex system of the living being.
Collapse
Affiliation(s)
- Rosa Noguera
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia/Instituto de Investigaciones Sanitarias INCLIVA, Valencia, España; CIBERONC, Madrid, España
| | - Rebeca Burgos-Panadero
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia/Instituto de Investigaciones Sanitarias INCLIVA, Valencia, España; CIBERONC, Madrid, España
| | - Esther Gamero-Sandemetrio
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia/Instituto de Investigaciones Sanitarias INCLIVA, Valencia, España; CIBERONC, Madrid, España
| | | | - Tomás Álvaro Naranjo
- CIBERONC, Madrid, España; Hospital Verge de la Cinta, Tortosa, Tarragona, España.
| |
Collapse
|
118
|
Pedrosa CR, Arl D, Grysan P, Khan I, Durrieu S, Krishnamoorthy S, Durrieu MC. Controlled Nanoscale Topographies for Osteogenic Differentiation of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:8858-8866. [PMID: 30785254 DOI: 10.1021/acsami.8b21393] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanotopography with length scales of the order of extracellular matrix elements offers the possibility of regulating cell behavior. Investigation of the impact of nanotopography on cell response has been limited by the inability to precisely control geometries, especially at high spatial resolutions and across practically large areas. In this paper, we demonstrate well-controlled and periodic nanopillar arrays of silicon and investigate their impact on osteogenic differentiation of human mesenchymal stem cells (hMSCs). Silicon nanopillar arrays with critical dimensions in the range of 40-200 nm, exhibiting standard deviations below 15% across full wafers, were realized using the self-assembly of block copolymer colloids. Immunofluorescence and quantitative polymerase chain reaction measurements reveal clear dependence of osteogenic differentiation of hMSCs on the diameter and periodicity of the arrays. Further, the differentiation of hMSCs was found to be dependent on the age of the donor. While osteoblastic differentiation was found to be promoted by the pillars with larger diameters and heights independent of donor age, they were found to be different for different spacings. Pillar arrays with smaller pitch promoted differentiation from a young donor, while a larger spacing promoted those of an old donor. These findings can contribute for the development of personalized treatments of bone diseases, namely, novel implant nanostructuring depending on patient age.
Collapse
Affiliation(s)
- Catarina R Pedrosa
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN) , 33600 Pessac , France
- CNRS, CBMN UMR5248 , 33600 Pessac , France
- Bordeaux INP, CBMN UMR5248 , 33600 Pessac , France
- Materials Research and Technology (MRT) Department , Luxembourg Institute of Science and Technology (LIST) , L-4422 Belvaux , Luxembourg
| | - Didier Arl
- Materials Research and Technology (MRT) Department , Luxembourg Institute of Science and Technology (LIST) , L-4422 Belvaux , Luxembourg
| | - Patrick Grysan
- Materials Research and Technology (MRT) Department , Luxembourg Institute of Science and Technology (LIST) , L-4422 Belvaux , Luxembourg
| | - Irfan Khan
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN) , 33600 Pessac , France
- CNRS, CBMN UMR5248 , 33600 Pessac , France
- Bordeaux INP, CBMN UMR5248 , 33600 Pessac , France
| | - Stéphanie Durrieu
- ARNA Laboratory , Université de Bordeaux , 33076 Bordeaux , France
- ARNA Laboratory , INSERM, U1212-CNRS UMR 5320 , 33000 Bordeaux , France
| | - Sivashankar Krishnamoorthy
- Materials Research and Technology (MRT) Department , Luxembourg Institute of Science and Technology (LIST) , L-4422 Belvaux , Luxembourg
| | - Marie-Christine Durrieu
- Université de Bordeaux, Chimie et Biologie des Membranes et Nano-Objets (UMR5248 CBMN) , 33600 Pessac , France
- CNRS, CBMN UMR5248 , 33600 Pessac , France
- Bordeaux INP, CBMN UMR5248 , 33600 Pessac , France
| |
Collapse
|
119
|
Mardones R, Camacho D, Monsalvo F, Zulch N, Jofre C, Minguell JJ. Treatment of osteonecrosis of the femoral head by core decompression and implantation of fully functional ex vivo-expanded bone marrow-derived mesenchymal stem cells: a proof-of-concept study. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2019; 12:11-16. [PMID: 30881048 PMCID: PMC6402444 DOI: 10.2147/sccaa.s181883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Based on several attributes involved in bone formation, bone marrow-resident mesenchymal stem cells (MSCs) have been employed in the treatment of patients suffering from femoral head osteonecrosis. Due to the low content of MSCs in the bone marrow, ex vivo expansion procedures are utilized to increase the cell number. Customarily, before administration of the resulting expanded cell product MSCs to the patient, its cellular identity is usually evaluated according to a set of “minimal phenotypic” markers, which are not modified by ex vivo processing. However, MSC functional (“reparative”) markers, which are severely impaired along the ex vivo expansion routine, are usually not assessed. Patients and methods In this proof-of-concept study, a cohort of five avascular osteonecrosis patients received an instillation of ex vivo-expanded autologous MSCs, manufactured under controlled conditions, with an aim to protect their functional (“reparative”) capacity. Results and conclusion Outcomes of this study confirmed the safety and effectiveness of the MSC-based therapy used. After a follow-up period (19–54 months), in all patients, the hip function was significantly improved and pain intensity markedly reduced. As a corollary, no patient required hip arthroplasty.
Collapse
Affiliation(s)
| | - Daniel Camacho
- Department of Orthopedics, Clínica Las Condes, Santiago, Chile.,Department of Orthopedics, Instituto Traumatológico, Santiago, Chile
| | | | - Nicolás Zulch
- Department of Orthopedics, Clínica Las Condes, Santiago, Chile
| | - Claudio Jofre
- Centro de Terapia Regenerativa Celular, Clínica Las Condes, Santiago, Chile,
| | - José J Minguell
- Centro de Terapia Regenerativa Celular, Clínica Las Condes, Santiago, Chile,
| |
Collapse
|
120
|
Wang J, Sun M, Liu W, Li Y, Li M. Stem Cell-Based Therapies for Liver Diseases: An Overview and Update. Tissue Eng Regen Med 2019; 16:107-118. [PMID: 30989038 DOI: 10.1007/s13770-019-00178-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver disease is one of the top causes of death globally. Although liver transplantation is a very effective treatment strategy, the shortage of available donor organs, waiting list mortality, and high costs of surgery remain huge problems. Stem cells are undifferentiated cells that can differentiate into a variety of cell types. Scientists are exploring the possibilities of generating hepatocytes from stem cells as an alternative for the treatment of liver diseases. METHODS In this review, we summarized the updated researches in the field of stem cell-based therapies for liver diseases as well as the current challenges and future expectations for a successful cell-based liver therapy. RESULTS Several cell types have been investigated for liver regeneration, such as embryonic stem cells, induced pluripotent stem cells, liver stem cells, mesenchymal stem cells, and hematopoietic stem cells. In vitro and in vivo studies have demonstrated that stem cells are promising cell sources for the liver regeneration. CONCLUSION Stem cell-based therapy could be a promising therapeutic method for patients with end-stage liver disease, which may alleviate the need for liver transplantation in the future.
Collapse
Affiliation(s)
- Jie Wang
- 1Department of Neurology, The China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, 130033 Jilin China
| | - Meiyan Sun
- Medical Examination College, Jilin Medical University, No. 5 Jilin Street, Changchun, Jilin, 132013 China
| | - Wei Liu
- Medical Examination College, Jilin Medical University, No. 5 Jilin Street, Changchun, Jilin, 132013 China
| | - Yan Li
- Medical Examination College, Jilin Medical University, No. 5 Jilin Street, Changchun, Jilin, 132013 China
| | - Miao Li
- 3Department of Neurosurgery, The China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, 130033 China
| |
Collapse
|
121
|
Naderain H, Khanlarkhani N, Ragerdi Kashani I, Atlasi A, Atlasi MA. Comparison of the effects of progesterone and 17 β-estradiol on Schwann cell markers expression in rat adipose-derived stem cells. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2019; 9:307-313. [PMID: 30713608 PMCID: PMC6346486 DOI: 10.30466/vrf.2018.33103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/21/2018] [Indexed: 11/22/2022]
Abstract
Steroids promote the myelination and regeneration in the peripheral nervous system. Whereas, little is known about the inducing effects by which the hormones exert their effects on Schwann cells differentiation. This could be revealed by the expression of Schwann cell markers in adipose-derived stem cells (ADSCs). The purpose of this study was to present the effects of progesterone and 17 β-estradiol on the Schwann cell markers in rat ADSCs. The mesenchymal stem cell markers (CD73, and CD90) were assayed by flow cytometry. Rat ADSCs were sequentially treated with β-mercaptoethanol, and all-trans-retinoic acid, followed by a mixture of basic fibrobroblast growth factor, platelet-derived growth factor, forskolin and heregulin. In experimental groups, forskolin and heregulin were substituted by progesterone and 17 β-estradiol. After induction, the expression of Schwann cell markers P0, and S-100 and the cellular immunocytochemical staining positive rate of anti-S100 and anti-glial fibrillary acidic protein (GFAP) antibodies were compared in the experimental and control groups. Progesterone and 17 β-estradiol triggered P0 and S-100 genes expression and induced a cellular immunocytochemical staining positive rate of S-100 and GFAP in rats ADSCs. Progesterone induced these changes stronger than 17 β-estradiol. Thus, progesterone may induce rat ADSCs toward Schwann-like cells by expression of Schwann cell markers and is more potent than 17 β-estradiol in the expression of these markers.
Collapse
Affiliation(s)
- Homayoun Naderain
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Khanlarkhani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirabbas Atlasi
- Student Research Committee, Faculty of Dentistry, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
122
|
Ansari MO, Gauthaman K, Essa A, Bencherif SA, Memic A. Graphene and Graphene-Based Materials in Biomedical Applications. Curr Med Chem 2019; 26:6834-6850. [DOI: 10.2174/0929867326666190705155854] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/06/2018] [Accepted: 10/26/2018] [Indexed: 12/27/2022]
Abstract
:
Nanobiotechnology has huge potential in the field of regenerative medicine. One of the main
drivers has been the development of novel nanomaterials. One developing class of materials is graphene
and its derivatives recognized for their novel properties present on the nanoscale. In particular,
graphene and graphene-based nanomaterials have been shown to have excellent electrical, mechanical,
optical and thermal properties. Due to these unique properties coupled with the ability to tune their
biocompatibility, these nanomaterials have been propelled for various applications. Most recently, these
two-dimensional nanomaterials have been widely recognized for their utility in biomedical research. In
this review, a brief overview of the strategies to synthesize graphene and its derivatives are discussed.
Next, the biocompatibility profile of these nanomaterials as a precursor to their biomedical application
is reviewed. Finally, recent applications of graphene-based nanomaterials in various biomedical fields
including tissue engineering, drug and gene delivery, biosensing and bioimaging as well as other biorelated
studies are highlighted.
Collapse
Affiliation(s)
| | - Kalamegam Gauthaman
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States
| | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
123
|
Ashammakhi N, Ahadian S, Darabi MA, El Tahchi M, Lee J, Suthiwanich K, Sheikhi A, Dokmeci MR, Oklu R, Khademhosseini A. Minimally Invasive and Regenerative Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1804041. [PMID: 30565732 PMCID: PMC6709364 DOI: 10.1002/adma.201804041] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/20/2018] [Indexed: 05/03/2023]
Abstract
Advances in biomaterial synthesis and fabrication, stem cell biology, bioimaging, microsurgery procedures, and microscale technologies have made minimally invasive therapeutics a viable tool in regenerative medicine. Therapeutics, herein defined as cells, biomaterials, biomolecules, and their combinations, can be delivered in a minimally invasive way to regenerate different tissues in the body, such as bone, cartilage, pancreas, cardiac, skeletal muscle, liver, skin, and neural tissues. Sophisticated methods of tracking, sensing, and stimulation of therapeutics in vivo using nano-biomaterials and soft bioelectronic devices provide great opportunities to further develop minimally invasive and regenerative therapeutics (MIRET). In general, minimally invasive delivery methods offer high yield with low risk of complications and reduced costs compared to conventional delivery methods. Here, minimally invasive approaches for delivering regenerative therapeutics into the body are reviewed. The use of MIRET to treat different tissues and organs is described. Although some clinical trials have been performed using MIRET, it is hoped that such therapeutics find wider applications to treat patients. Finally, some future perspective and challenges for this emerging field are highlighted.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mohammad Ali Darabi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mario El Tahchi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- LBMI, Department of Physics, Lebanese University - Faculty of Sciences 2, PO Box 90656, Jdeidet, Lebanon
| | - Junmin Lee
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Kasinan Suthiwanich
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Amir Sheikhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Mehmet R. Dokmeci
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, California, USA
- Department of Radiological Sciences, University of California - Los Angeles, Los Angeles, California, USA
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, California, USA
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
124
|
Dhaliwal A, Pelka S, Gray DS, Moghe PV. Engineering Lineage Potency and Plasticity of Stem Cells using Epigenetic Molecules. Sci Rep 2018; 8:16289. [PMID: 30389989 PMCID: PMC6215020 DOI: 10.1038/s41598-018-34511-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 10/11/2018] [Indexed: 12/17/2022] Open
Abstract
Stem cells are considered as a multipotent regenerative source for diseased and dysfunctional tissues. Despite the promise of stem cells, the inherent capacity of stem cells to convert to tissue-specific lineages can present a major challenge to the use of stem cells for regenerative medicine. We hypothesized that epigenetic regulating molecules can modulate the stem cell’s developmental program, and thus potentially overcome the limited lineage differentiation that human stem cells exhibit based on the source and processing of stem cells. In this study, we screened a library of 84 small molecule pharmacological agents indicated in nucleosomal modification and identified a sub-set of specific molecules that influenced osteogenesis in human mesenchymal stem cells (hMSCs) while maintaining cell viability in-vitro. Pre-treatment with five candidate hits, Gemcitabine, Decitabine, I-CBP112, Chidamide, and SIRT1/2 inhibitor IV, maximally enhanced osteogenesis in-vitro. In contrast, five distinct molecules, 4-Iodo-SAHA, Scriptaid, AGK2, CI-amidine and Delphidine Chloride maximally inhibited osteogenesis. We then tested the role of these molecules on hMSCs derived from aged human donors and report that small epigenetic molecules, namely Gemcitabine and Chidamide, can significantly promote osteogenic differentiation by 5.9- and 2.3-fold, respectively. Taken together, this study demonstrates new applications of identified small molecule drugs for sensitively regulating the lineage plasticity fates of bone-marrow derived mesenchymal stem cells through modulating the epigenetic profile of the cells.
Collapse
Affiliation(s)
- Anandika Dhaliwal
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States
| | - Sandra Pelka
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States
| | - David S Gray
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States. .,Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ, United States.
| |
Collapse
|
125
|
Jiao Y, Li XY, Liu J. A New Approach to Cerebral Palsy Treatment: Discussion of the Effective Components of Umbilical Cord Blood and its Mechanisms of Action. Cell Transplant 2018; 28:497-509. [PMID: 30384766 PMCID: PMC7103597 DOI: 10.1177/0963689718809658] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cerebral palsy (CP) includes a group of persistent non-progressive disorders
affecting movement, muscle tone, and/or posture. The total economic loss during
the life-span of an individual with CP places a heavy financial burden on such
patients and their families worldwide; however, a complete cure is still
lacking. Umbilical cord blood (UCB)-based interventions are emerging as a
scientifically plausible treatment and possible cure for CP. Stem cells have
been used in many experimental CP animal models and achieved good results.
Compared with other types of stem cells, those from UCB have advantages in terms
of treatment safety and efficacy, ethics, non-neoplastic proliferation,
accessibility, ease of preservation, and regulation of immune responses, based
on findings in animal models and clinical trials. Currently, the use of
UCB-based interventions for CP is limited as the components of UCB are complex
and possess different therapeutic mechanisms. These can be categorized by three
aspects: homing and neuroregeneration, trophic factor secretion, and
neuroprotective effects. Our review summarizes the features of active components
of UCB and their therapeutic mechanism of action. This review highlights current
research findings and clinical evidence regarding UCB that contribute to
treatment suggestions, inform decision-making for therapeutic interventions, and
help to direct future research.
Collapse
Affiliation(s)
- Yang Jiao
- 1 Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Xiao-Yan Li
- 1 Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Jing Liu
- 1 Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| |
Collapse
|
126
|
Rafaiee R, Ahmadiankia N. Bone Marrow Derived Mesenchymal Stem Cells in Addiction Related Hippocampal Damages. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:69-79. [PMID: 30276162 PMCID: PMC6148505 DOI: 10.22088/ijmcm.bums.7.2.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/13/2018] [Indexed: 11/30/2022]
Abstract
The brain is an important organ that controls all sensory and motor actions, memory, and emotions. Each anatomical and physiological modulation in various brain centers, results in psychological, behavioral, and sensory-motor changes. Alcohol and addictive drugs such as opioids and amphetamines have been shown to exert a great impact on brain, specifically on the hippocampus. Emerging evidence has indicated that altered hippocampal neurogenesis is associated with the pathophysiology of neuropsychological disorders including addiction. The addictive drugs impair neurogenesis and undermine the function of neural stem/progenitor cells in hippocampus. This feature was claimed to be one of the underlying mechanisms of behavioral changes in patients with addiction. As the impairment of stem cells’ function has been proven to be the underlying cause of pathologic neuroadaptations in the brain, the administration of stem cell populations has shown promising results for re-modulating of neuronal status in the brain and especially in the hippocampus. Among the different types of stem cells, bone marrow derived mesenchymal stem cells are the most proper candidates for stem cell therapies. In this review article, the recent studies on the effects of addictive drugs on brain neurogenesis, and also the promising potential effects of stem cells in curing addiction related hippocampal damages are discussed.
Collapse
Affiliation(s)
- Raheleh Rafaiee
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | |
Collapse
|
127
|
Assessment of the hepatocytic differentiation ability of human skin-derived ABCB5+ stem cells. Exp Cell Res 2018; 369:335-347. [DOI: 10.1016/j.yexcr.2018.05.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 02/06/2023]
|
128
|
Wang JJ, Zhang WX, Wang KF, Zhang S, Han X, Guan WJ, Ma YH. Isolation and biological characteristics of multipotent mesenchymal stromal cells derived from chick embryo intestine. Br Poult Sci 2018; 59:521-530. [PMID: 29914266 DOI: 10.1080/00071668.2018.1490495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
1. Over the past decade, rapid advancement in isolation methods for identifying markers of the once elusive intestinal stem cell (ISC) populations has laid the foundation for unravelling their complex interrelationships during homeostasis. Study on ISC in avian intestinal tissue might play a pivotal foundation for further studies on the epithelial-to-mesenchymal transition (EMT) in gastrointestinal disease and cell-based therapy as well as intestinal tissue engineering. 2. The following experiment isolated a population of fibroblast-like, plastic adhering cells derived from chick embryo intestine, showing a strong self-renewing and proliferative ability, which was maintained in vitro up to passage 25. The findings included growth characteristics, detected expression of cell surface markers and characterised the capability of these cells to differentiate towards the osteogenic, adipogenic, and chondrogenic cell lineages. 3. RT-PCR analysis showed that these cells from chick embryos expressed mesenchymal stromal cell markers CD44, CD90 and VIMENTIN as well as ISC-specific genes LGR5, MI1, SMOC2, BMI1, and HOPX. Immunofluorescence and flow cytometry confirmed this biology characterisation further. 4. In conclusion, cells were isolated from the intestine of 18-day-old chicken embryos that exhibited the biological characteristics of mesenchymal stromal cells as well as markers of intestinal stem cells. Our findings may provide a novel insight for in vitro cell culture and characteristics of ISCs in avian species, which may also indicate a benefit for obtaining cell source for intestinal tissue engineering as well as cell-based investigation for gastrointestinal disease and treatment.
Collapse
Affiliation(s)
- J J Wang
- a Institute of Animal Science , Chinese Academy of Agricultural Sciences , Beijing , China.,b Department of Kinesiology and Health , Harbin Sport University , Harbin , Heilongjiang , China
| | - W X Zhang
- a Institute of Animal Science , Chinese Academy of Agricultural Sciences , Beijing , China
| | - K F Wang
- a Institute of Animal Science , Chinese Academy of Agricultural Sciences , Beijing , China
| | - S Zhang
- c Research Center for Sports Scientific Experiment , Harbin Sport University , Harbin , Heilongjiang , China
| | - X Han
- a Institute of Animal Science , Chinese Academy of Agricultural Sciences , Beijing , China
| | - W J Guan
- a Institute of Animal Science , Chinese Academy of Agricultural Sciences , Beijing , China
| | - Y H Ma
- a Institute of Animal Science , Chinese Academy of Agricultural Sciences , Beijing , China
| |
Collapse
|
129
|
Lv Y, Liu B, Liu Y, Wang H, Wang H. TGF-β1 combined with Sal-B promotes cardiomyocyte differentiation of rat mesenchymal stem cells. Exp Ther Med 2018; 15:5359-5364. [PMID: 29904415 DOI: 10.3892/etm.2018.6105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/27/2017] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor β1 (TGF-β1) and salvianolic acid B (Sal-B) are key signaling factors for stem cell differentiation into cardiomyocytes (CMs). The present study compared the biological effect of TGF-β1 and Sal-B, alone or in combination, on bone marrow mesenchymal stromal cells (BMSCs) that differentiate into myocardial-like cells in a simulated myocardial microenvironment in vitro. BMSCs were isolated from bones of limbs of 10 male Sprague Dawley rats and cultured. The 2nd-generation BMSCs were co-incubated with TGF-β1 and Sal-B, alone or in combination, for 72 h. The control group was BMSCs cultured without any inductive substance. The levels GATA binding protein 4 (GATA4) and homeobox protein NKx2.5 were determined by reverse-transcription quantitative polymerase chain reaction and immunofluorescence staining was used to evaluate α-sarcomeric actin and cardiac troponin I (cTNI) as cardiomyogenic differentiation markers. The ultrastructure of BMSCs in each group was also observed. BMSCs were initially spindle-shaped with irregular processes. The cells gradually increased in number 24 h post-inoculation and proliferated 7 days later. Compared with the control group, BMSCs in the treatment groups had fusiform shapes, orientating with one accord and were connected with adjoining cells forming myotube-like structures on day 28. The morphology and architecture/myotubes of BMSCs was similar among the treatment groups, but the amount of cells in the combined group was comparatively higher. The results of immunofluorescence staining revealed the expression of the CM-specific proteins α-sarcomeric actin and cTNI in these cells. The expression of these cardiac-specific markers in the combined group was significantly higher than that in the other groups (P<0.01 or P<0.05). In addition, the transcriptional expression of GATA4 and NKx2.5 in the treatment groups was stable and significantly higher than that in the control group on day 7. Transmission electron microscopy showed that BMSCs in the treatment groups all had myofilaments, rough endoplasmic reticulum and mitochondria in the cytoplasm when compared with the control group. Taken together, these results indicated that the combination of TGF-β1 and Sal-B effectively promotes cardiomyogenic differentiation of BMSCs in vitro and their application may represent a therapeutic strategy for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Yang Lv
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Bo Liu
- Department of Pathology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Yuan Liu
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Haoyu Wang
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Haiping Wang
- Department of Histology and Embryology, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|
130
|
Isolation and Characterization of Multipotent Turkey Tendon-Derived Stem Cells. Stem Cells Int 2018; 2018:3697971. [PMID: 29977306 PMCID: PMC6011053 DOI: 10.1155/2018/3697971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
Abstract
Tendon injuries are among the most common and severe hand injuries with a high demand for functional recovery. Stem cells have been identified and isolated from different species and a variety of tissues for the sake of regenerative medicine. Recently, turkey has been suggested as a potential new large animal model for flexor tendon-related research. However, turkey tissue-specific stem cells have not been investigated. Here, we presented the isolation and verification of tendon-derived stem cells (TDSCs) from 6- to 8-month-old heritage-breed turkey. TDSCs were isolated from turkey flexor tendon by plating nucleated cells at the determined optimal density. Approximately 4% of the nucleated cells demonstrated clonogenicity, high proliferation rate, and trilineage differentiation potential after induction culturing. These cells expressed surface antigens CD90, CD105, and CD44, but did not express CD45. There was a high level of gene expression of tenogenic markers in TDSCs, including mohawk, collagen type I, tenascin C, and elastin. Turkey TDSCs also expressed transcription factors PouV, Nanog, and Sox2, which are critically involved in the regulation of stemness. The successful isolation of tendon-derived stem cells from turkey was beneficial for future studies in tendon tissue engineering and would help in the development of new treatment for tendon diseases using this novel animal model.
Collapse
|
131
|
Supplementation with IL-6 and Muscle Cell Culture Conditioned Media Enhances Myogenic Differentiation of Adipose Tissue-Derived Stem Cells through STAT3 Activation. Int J Mol Sci 2018; 19:ijms19061557. [PMID: 29882916 PMCID: PMC6032255 DOI: 10.3390/ijms19061557] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 12/25/2022] Open
Abstract
Mature skeletal muscle cells cannot be expanded in culture systems. Therefore, it is difficult to construct an in vitro model for muscle diseases. To establish an efficient protocol for myogenic differentiation of human adipose tissue-derived stem cells (hADSCs), we investigated whether addition of IL-6 and/or myocyte-conditioned media (CM) to conventional differentiation media can shorten the differentiation period. hADSCs were differentiated to myocytes using the conventional protocol or modified with the addition of 25 pg/mL IL-6 and/or C2C12 CM (25% v/v). The expression of MyoD and myogenine mRNA was significantly higher at 5⁻6 days after differentiation using the modified protocol than with the conventional protocol. mRNA and protein expression of myosin heavy chain, a marker of myotubes, was significantly upregulated at 28 and 42 days of differentiation using the modified protocol, and the level achieved after a 4-week differentiation period was similar to that achieved at 6 weeks using the conventional protocol. The expression of p-STAT3 was significantly increased when the modified protocol was used. Similarly, addition of colivelin, a STAT3 activator, instead of IL-6 and C2C12 CM, promoted the myogenic differentiation of ADSCs. The modified protocol improved differentiation efficiency and reduced the time required for differentiation of myocytes. It might be helpful to save cost and time when preparing myocytes for cell therapies and drug discovery.
Collapse
|
132
|
Mussano F, Genova T, Petrillo S, Roato I, Ferracini R, Munaron L. Osteogenic Differentiation Modulates the Cytokine, Chemokine, and Growth Factor Profile of ASCs and SHED. Int J Mol Sci 2018; 19:ijms19051454. [PMID: 29757956 PMCID: PMC5983594 DOI: 10.3390/ijms19051454] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023] Open
Abstract
Great efforts have been made to improve bone regeneration techniques owing to a growing variety of sources of stem cells suitable for autologous transplants. Specifically, adipose-derived stem cells (ASCs) and stems cells from human exfoliated deciduous teeth (SHED) hold great potential for bone tissue engineering and cell therapy. After a preliminary characterization of the main biomolecules ASCs and SHED released in their conditioned media, cells were kept both in normal and osteo-inducing conditions. Conventional assays were performed to prove their osteogenic potential such as quantitative real-time polymerase chain reaction (qRT-PCR) (for RUNX-2, collagen type I, osteopontin and osteonectin), alkaline phosphatase activity, osteocalcin production, and von Kossa staining. Conditioned media were tested again after the osteogenic induction and compared to maintaining condition both at base line and after 14 days of culture. The osteogenic condition inhibited the release of all the biomolecules, with the exception, concerning SHED, of growth-regulated alpha protein precursor (GROα), and, to a lesser extent, interleukin (IL)-8. In conclusion, our data support that undifferentiated ASCs and SHED may be preferable to committed ones for general cell therapy approaches, due to their higher paracrine activity. Osteoinduction significantly affects the cytokine, chemokine, and growth factor profile in a differential way, as SHED kept a more pronounced pro-angiogenic signature than ASCs.
Collapse
Affiliation(s)
- Federico Mussano
- CIR Dental School, Department of Surgical Sciences UNITO, via Nizza 230, 10126 Turin, Italy.
| | - Tullio Genova
- CIR Dental School, Department of Surgical Sciences UNITO, via Nizza 230, 10126 Turin, Italy.
- Department of Life Sciences and Systems Biology, UNITO, via Accademia Albertina 13, 10123 Turin, Italy.
| | - Sara Petrillo
- Department of Molecular Biotechnology and Health Sciences, UNITO, Via Nizza 52, 10126 Turin, Italy.
| | - Ilaria Roato
- Center for Research and Medical Studies, A.O.U. Città della Salute e della Scienza, 10126 Turin, Italy.
| | - Riccardo Ferracini
- Department of Surgical Sciences (DISC), Orthopaedic Clinic-IRCCS A.O.U. San Martino, 16132 Genoa, Italy.
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, UNITO, via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
133
|
Alvites R, Rita Caseiro A, Santos Pedrosa S, Vieira Branquinho M, Ronchi G, Geuna S, Varejão AS, Colette Maurício A. Peripheral nerve injury and axonotmesis: State of the art and recent advances. COGENT MEDICINE 2018. [DOI: 10.1080/2331205x.2018.1466404] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
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
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Ana Rita Caseiro
- 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
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, Universidade do Porto (REQUIMTE/LAQV), R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sílvia Santos Pedrosa
- 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
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Mariana Vieira 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
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Giulia Ronchi
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Stefano Geuna
- Departamento de Ciências Veterinárias, Universidade de Trás-os-Montes e Alto Douro, UTAD, Quinta de Prados, 5000-801 Vila Real, Portugal
| | - Artur S.P. Varejão
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal
- Department of Clinical and Biological Sciences, and Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Ospedale San Luigi, 10043 Orbassano, Turin, Italy
| | - 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
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| |
Collapse
|
134
|
Silva MDA, Leite YKDC, de Carvalho CES, Feitosa MLT, Alves MMDM, Carvalho FADA, Neto BCV, Miglino MA, Jozala AF, de Carvalho MAM. Behavior and biocompatibility of rabbit bone marrow mesenchymal stem cells with bacterial cellulose membrane. PeerJ 2018; 6:e4656. [PMID: 29736332 PMCID: PMC5933324 DOI: 10.7717/peerj.4656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/01/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tissue engineering has been shown to exhibit great potential for the creation of biomaterials capable of developing into functional tissues. Cellular expansion and integration depends on the quality and surface-determinant factors of the scaffold, which are required for successful biological implants. The objective of this research was to characterize and evaluate the in vitro characteristics of rabbit bone marrow mesenchymal stem cells (BM-MSCs) associated with a bacterial cellulose membrane (BCM). We assessed the adhesion, expansion, and integration of the biomaterial as well as its ability to induce macrophage activation. Finally, we evaluated the cytotoxicity and toxicity of the BCM. METHODS Samples of rabbit bone marrow were collected. Mesenchymal stem cells were isolated from medullary aspirates to establish fibroblast colony-forming unit assay. Osteogenic, chondrogenic, and adipogenic differentiation was performed. Integration with the BCM was assessed by scanning electron microscopy at 1, 7, and 14 days. Cytotoxicity was assessed via the production of nitric oxide, and BCM toxicity was assessed with the MTT assay; phagocytic activity was also determined. RESULTS The fibroblastoid colony-forming unit (CFU-F) assay showed cells with a fibroblastoid morphology organized into colonies, and distributed across the culture area surface. In the growth curve, two distinct phases, lag and log phase, were observed at 15 days. Multipotentiality of the cells was evident after induction of osteogenic, chondrogenic, and adipogenic lineages. Regarding the BM-MSCs' bioelectrical integration with the BCM, BM-MSCs were anchored in the BCM in the first 24 h. On day 7 of culture, the cytoplasm was scattered, and on day 14, the cells were fully integrated with the biomaterial. We also observed significant macrophage activation; analysis of the MTT assay and the concentration of nitric oxide revealed no cytotoxicity of the biomaterial. CONCLUSION The BCM allowed the expansion and biointegration of bone marrow progenitor cells with a stable cytotoxic profile, thus presenting itself as a biomaterial with potential for tissue engineering.
Collapse
Affiliation(s)
- Marcello de Alencar Silva
- Integrated Nucleus of Morphology and Stem Cell Research, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Matheus Levi Tajra Feitosa
- Integrated Nucleus of Morphology and Stem Cell Research, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Bartolomeu Cruz Viana Neto
- Department of Physics/Advanced Microscopy Multiuser Laboratory/Laboratory of Physics Material, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Maria Angélica Miglino
- Departament of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Angela Faustino Jozala
- Laboratory of Industrial Microbiology and Fermentation Process, University of Sorocaba, Sorocaba, São Paulo, Brazil
| | | |
Collapse
|
135
|
The Differentiation Balance of Bone Marrow Mesenchymal Stem Cells Is Crucial to Hematopoiesis. Stem Cells Int 2018; 2018:1540148. [PMID: 29765406 PMCID: PMC5903338 DOI: 10.1155/2018/1540148] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/21/2018] [Indexed: 01/20/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs), the important component and regulator of bone marrow microenvironment, give rise to hematopoietic-supporting stromal cells and form hematopoietic niches for hematopoietic stem cells (HSCs). However, how BMSC differentiation affects hematopoiesis is poorly understood. In this review, we focus on the role of BMSC differentiation in hematopoiesis. We discussed the role of BMSCs and their progeny in hematopoiesis. We also examine the mechanisms that cause differentiation bias of BMSCs in stress conditions including aging, irradiation, and chemotherapy. Moreover, the differentiation balance of BMSCs is crucial to hematopoiesis. We highlight the negative effects of differentiation bias of BMSCs on hematopoietic recovery after bone marrow transplantation. Keeping the differentiation balance of BMSCs is critical for hematopoietic recovery. This review summarises current understanding about how BMSC differentiation affects hematopoiesis and its potential application in improving hematopoietic recovery after bone marrow transplantation.
Collapse
|
136
|
Shin HS, Lee S, Hong HJ, Lim YC, Koh WG, Lim JY. Stem cell properties of human clonal salivary gland stem cells are enhanced by three-dimensional priming culture in nanofibrous microwells. Stem Cell Res Ther 2018; 9:74. [PMID: 29566770 PMCID: PMC5863805 DOI: 10.1186/s13287-018-0829-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/05/2018] [Accepted: 03/07/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cultures recapitulate the microenvironment of tissue-resident stem cells and enable them to modulate their properties. We determined whether salivary gland-resident stem cells (SGSCs) are primed by a 3D spheroid culture prior to treating irradiation-induced salivary hypofunction using in-vitro coculture and in-vivo transplant models. METHODS 3D spheroid-derived SGSCs (SGSCs3D) were obtained from 3D culture in microwells consisting of a nanofiber bottom and cell-repellent hydrogel walls, and were examined for salivary stem or epithelial gene/protein expression, differentiation potential, and paracrine secretory function compared with monolayer-cultured SGSCs (SGSCs2D) in vitro and in vivo. RESULTS SGSCs3D expressed increased salivary stem cell markers (LGR5 and THY1) and pluripotency markers (POU5F1 and NANOG) compared with SGSCs2D. Also, SGSCs3D exhibited enhanced potential to differentiate into salivary epithelial cells upon differentiation induction and increased paracrine secretion as compared to SGSCs2D. Wnt signaling was activated by 3D spheroid formation in the microwells and suppression of the Wnt/β-catenin pathway led to reduced stemness of SGSCs3D. Enhanced radioprotective properties of SGSCs3D against radiation-induced salivary hypofunction was confirmed by an organotypic 3D coculture and in-vivo transplantation experiments. CONCLUSION The 3D spheroid culture of SGSCs in nanofibrous microwells promotes stem cell properties via activation of Wnt signaling. This may contribute to SGSC priming prior to regenerative therapy to restore salivary hypofunction after radiotherapy.
Collapse
Affiliation(s)
- Hyun-Soo Shin
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Songyi Lee
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Hye Jin Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Young Chang Lim
- Department of Otorhinolaryngology, Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
137
|
Sphingosine 1-Phosphate Receptor 1 Is Required for MMP-2 Function in Bone Marrow Mesenchymal Stromal Cells: Implications for Cytoskeleton Assembly and Proliferation. Stem Cells Int 2018; 2018:5034679. [PMID: 29713350 PMCID: PMC5866864 DOI: 10.1155/2018/5034679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cell- (BM-MSC-) based therapy is a promising option for regenerative medicine. An important role in the control of the processes influencing the BM-MSC therapeutic efficacy, namely, extracellular matrix remodelling and proliferation and secretion ability, is played by matrix metalloproteinase- (MMP-) 2. Therefore, the identification of paracrine/autocrine regulators of MMP-2 function may be of great relevance for improving BM-MSC therapeutic potential. We recently reported that BM-MSCs release the bioactive lipid sphingosine 1-phosphate (S1P) and, here, we demonstrated an impairment of MMP-2 expression/release when the S1P receptor subtype S1PR1 is blocked. Notably, active S1PR1/MMP-2 signalling is required for F-actin structure assembly (lamellipodia, microspikes, and stress fibers) and, in turn, cell proliferation. Moreover, in experimental conditions resembling the damaged/regenerating tissue microenvironment (hypoxia), S1P/S1PR1 system is also required for HIF-1α expression and vinculin reduction. Our findings demonstrate for the first time the trophic role of S1P/S1PR1 signalling in maintaining BM-MSCs' ability to modulate MMP-2 function, necessary for cytoskeleton reorganization and cell proliferation in both normoxia and hypoxia. Altogether, these data provide new perspectives for considering S1P/S1PR1 signalling a pharmacological target to preserve BM-MSC properties and to potentiate their beneficial potential in tissue repair.
Collapse
|
138
|
Peptide modified nanofibrous scaffold promotes human mesenchymal stem cell proliferation and long-term passaging. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018. [DOI: 10.1016/j.msec.2017.11.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
139
|
Fregni G, Quinodoz M, Möller E, Vuille J, Galland S, Fusco C, Martin P, Letovanec I, Provero P, Rivolta C, Riggi N, Stamenkovic I. Reciprocal modulation of mesenchymal stem cells and tumor cells promotes lung cancer metastasis. EBioMedicine 2018; 29:128-145. [PMID: 29503225 PMCID: PMC5925622 DOI: 10.1016/j.ebiom.2018.02.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/14/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022] Open
Abstract
Metastasis is a multi-step process in which direct crosstalk between cancer cells and their microenvironment plays a key role. Here, we assessed the effect of paired tumor-associated and normal lung tissue mesenchymal stem cells (MSCs) on the growth and dissemination of primary human lung carcinoma cells isolated from the same patients. We show that the tumor microenvironment modulates MSC gene expression and identify a four-gene MSC signature that is functionally implicated in promoting metastasis. We also demonstrate that tumor-associated MSCs induce the expression of genes associated with an aggressive phenotype in primary lung cancer cells and selectively promote their dissemination rather than local growth. Our observations provide insight into mechanisms by which the stroma promotes lung cancer metastasis. Distinct gene expression profiles distinguish normal lung and tumor-associated MSCs. MSCs induce EMT- and hypoxia-related genes in primary tumor cells and promote their metastatic potential. A 4-gene T-MSC signature is involved in MSC-induced metastasis promotion.
The tumor microenvironment, which includes mesenchymal stem cells (MSCs) among many other stromal cell types, plays a fundamental role in cancer metastasis. Although MSCs are suggested to participate in tumor progression, most studies thus far have been performed on bone marrow-derived MSCs and cancer cell lines. Using primary human pulmonary MSCs and paired lung cancer cells, we show that tumor cells modulate MSCs to acquire properties, including a four-gene signature, which allow them to promote tumor dissemination. Our results provide insight into the mutual cancer cell-stromal cell modulation that drives tumor dissemination.
Collapse
Affiliation(s)
- Giulia Fregni
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Mathieu Quinodoz
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, 1011, Switzerland
| | - Emely Möller
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Joanna Vuille
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Sabine Galland
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Carlo Fusco
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Patricia Martin
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Igor Letovanec
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Paolo Provero
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, IRCCS, Milan20132, Italy; Dept. of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Carlo Rivolta
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, 1011, Switzerland; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 9HN, UK
| | - Nicolo Riggi
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Ivan Stamenkovic
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland.
| |
Collapse
|
140
|
Jakubikova J, Cholujova D, Hideshima T, Gronesova P, Soltysova A, Harada T, Joo J, Kong SY, Szalat RE, Richardson PG, Munshi NC, Dorfman DM, Anderson KC. A novel 3D mesenchymal stem cell model of the multiple myeloma bone marrow niche: biologic and clinical applications. Oncotarget 2018; 7:77326-77341. [PMID: 27764795 PMCID: PMC5357212 DOI: 10.18632/oncotarget.12643] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/28/2016] [Indexed: 12/19/2022] Open
Abstract
Specific niches within the tumor bone marrow (BM) microenvironment afford a sanctuary for multiple myeloma (MM) clones due to stromal cell-tumor cell interactions, which confer survival advantage and drug resistance. Defining the sequelae of tumor cell interactions within the MM niches on an individualized basis may provide the rationale for personalized therapies. To mimic the MM niche, we here describe a new 3D co-culture ex-vivo model in which primary MM patient BM cells are co-cultured with mesenchymal stem cells (MSC) in a hydrogel 3D system. In the 3D model, MSC with conserved phenotype (CD73+CD90+CD105+) formed compact clusters with active fibrous connections, and retained lineage differentiation capacity. Extracellular matrix molecules, integrins, and niche related molecules including N-cadherin and CXCL12 are expressed in 3D MSC model. Furthermore, activation of osteogenesis (MMP13, SPP1, ADAMTS4, and MGP genes) and osteoblastogenic differentiation was confirmed in 3D MSC model. Co-culture of patient-derived BM mononuclear cells with either autologous or allogeneic MSC in 3D model increased proliferation of MM cells, CXCR4 expression, and SP cells. We carried out immune profiling to show that distribution of immune cell subsets was similar in 3D and 2D MSC model systems. Importantly, resistance to novel agents (IMiDs, bortezomib, carfilzomib) and conventional agents (doxorubicin, dexamethasone, melphalan) was observed in 3D MSC system, reflective of clinical resistance. This 3D MSC model may therefore allow for studies of MM pathogenesis and drug resistance within the BM niche. Importantly, ongoing prospective trials are evaluating its utility to inform personalized targeted and immune therapy in MM.
Collapse
Affiliation(s)
- Jana Jakubikova
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Danka Cholujova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Teru Hideshima
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paulina Gronesova
- Cancer Research Institute, Biomedical Research Center SAS, Bratislava, Slovak Republic
| | - Andrea Soltysova
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Takeshi Harada
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jungnam Joo
- Biometric Research Branch, Division of Cancer Epidemiology and Prevention, Research Institute & Hospital, National Cancer Center, Goyang-si Gyeonggi-do, South Korea
| | - Sun-Young Kong
- Department of Laboratory Medicine and Translational Epidemiology Branch, Research Institute & Hospital, National Cancer Center, Goyang-si Gyeonggi-do, South Korea
| | - Raphael E Szalat
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nikhil C Munshi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana Farber Cancer Institute, Department of Medical Oncology, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
141
|
Desancé M, Contentin R, Bertoni L, Gomez-Leduc T, Branly T, Jacquet S, Betsch JM, Batho A, Legendre F, Audigié F, Galéra P, Demoor M. Chondrogenic Differentiation of Defined Equine Mesenchymal Stem Cells Derived from Umbilical Cord Blood for Use in Cartilage Repair Therapy. Int J Mol Sci 2018; 19:ijms19020537. [PMID: 29439436 PMCID: PMC5855759 DOI: 10.3390/ijms19020537] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/03/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022] Open
Abstract
Cartilage engineering is a new strategy for the treatment of cartilage damage due to osteoarthritis or trauma in humans. Racehorses are exposed to the same type of cartilage damage and the anatomical, cellular, and biochemical properties of their cartilage are comparable to those of human cartilage, making the horse an excellent model for the development of cartilage engineering. Human mesenchymal stem cells (MSCs) differentiated into chondrocytes with chondrogenic factors in a biomaterial appears to be a promising therapeutic approach for direct implantation and cartilage repair. Here, we characterized equine umbilical cord blood-derived MSCs (eUCB-MSCs) and evaluated their potential for chondrocyte differentiation for use in cartilage repair therapy. Our results show that isolated eUCB-MSCs had high proliferative capacity and differentiated easily into osteoblasts and chondrocytes, but not into adipocytes. A three-dimensional (3D) culture approach with the chondrogenic factors BMP-2 and TGF-β1 potentiated chondrogenic differentiation with a significant increase in cartilage-specific markers at the mRNA level (Col2a1, Acan, Snorc) and the protein level (type II and IIB collagen) without an increase in hypertrophic chondrocyte markers (Col10a1 and Mmp13) in normoxia and in hypoxia. However, these chondrogenic factors caused an increase in type I collagen, which can be reduced using small interfering RNA targeting Col1a2. This study provides robust data on MSCs characterization and demonstrates that eUCB-MSCs have a great potential for cartilage tissue engineering.
Collapse
Affiliation(s)
- Mélanie Desancé
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| | | | - Lélia Bertoni
- Center of Imaging and Research on Locomotor Affections in Equines, Ecole Vétérinaire d'Alfort, Université Paris-Est, 14430 Goustranville, France.
| | | | - Thomas Branly
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| | - Sandrine Jacquet
- Center of Imaging and Research on Locomotor Affections in Equines, Ecole Vétérinaire d'Alfort, Université Paris-Est, 14430 Goustranville, France.
| | - Jean-Marc Betsch
- Clinique Vétérinaire Equine de Méheudin, Méheudin, 61150 Ecouché, France.
| | - Agnès Batho
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
- EFS Caen, 14000 Caen, France.
| | | | - Fabrice Audigié
- Center of Imaging and Research on Locomotor Affections in Equines, Ecole Vétérinaire d'Alfort, Université Paris-Est, 14430 Goustranville, France.
| | - Philippe Galéra
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| | - Magali Demoor
- Normandie University, UNICAEN, BIOTARGEN, 14000 Caen, France.
| |
Collapse
|
142
|
Combined use of bone marrow-derived mesenchymal stromal cells (BM-MSCs) and platelet rich plasma (PRP) stimulates proliferation and differentiation of myoblasts in vitro: new therapeutic perspectives for skeletal muscle repair/regeneration. Cell Tissue Res 2018; 372:549-570. [PMID: 29404727 DOI: 10.1007/s00441-018-2792-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/06/2018] [Indexed: 10/18/2022]
Abstract
Satellite cell-mediated skeletal muscle repair/regeneration is compromised in cases of extended damage. Bone marrow mesenchymal stromal cells (BM-MSCs) hold promise for muscle healing but some criticisms hamper their clinical application, including the need to avoid animal serum contamination for expansion and the scarce survival after transplant. In this context, platelet-rich plasma (PRP) could offer advantages. Here, we compare the effects of PRP or standard culture media on C2C12 myoblast, satellite cell and BM-MSC viability, survival, proliferation and myogenic differentiation and evaluate PRP/BM-MSC combination effects in promoting myogenic differentiation. PRP induced an increase of mitochondrial activity and Ki67 expression comparable or even greater than that elicited by standard media and promoted AKT signaling activation in myoblasts and BM-MSCs and Notch-1 pathway activation in BM-MSCs. It stimulated MyoD, myogenin, α-sarcomeric actin and MMP-2 expression in myoblasts and satellite cell activation. Notably, PRP/BM-MSC combination was more effective than PRP alone. We found that BM-MSCs influenced myoblast responses through a paracrine activation of AKT signaling, contributing to shed light on BM-MSC action mechanisms. Our results suggest that PRP represents a good serum substitute for BM-MSC manipulation in vitro and could be beneficial towards transplanted cells in vivo. Moreover, it might influence muscle resident progenitors' fate, thus favoring the endogenous repair/regeneration mechanisms. Finally, within the limitations of an in vitro experimentation, this study provides an experimental background for considering the PRP/BM-MSC combination as a potential therapeutic tool for skeletal muscle damage, combining the beneficial effects of BM-MSCs and PRP on muscle tissue, while potentiating BM-MSC functionality.
Collapse
|
143
|
Klomps LV, Zomorodi N, Kim HM. Role of transplanted bone marrow cells in development of rotator cuff muscle fatty degeneration in mice. J Shoulder Elbow Surg 2017; 26:2177-2186. [PMID: 28869071 DOI: 10.1016/j.jse.2017.06.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/12/2017] [Accepted: 06/19/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND Rotator cuff muscle fatty degeneration after a chronic tendon tear is an irreversible pathologic change associated with poor clinical outcomes of tendon repair, and its exact pathogenesis remains unknown. We sought to investigate the role of transplanted bone marrow cells in the development of fatty degeneration, specifically in adipocyte accumulation, using a mouse model. METHODS Fourteen mice were divided into 2 bone marrow chimeric animal groups: bone marrow transplantation (BMT) group and reverse BMT group. For the BMT group, C57BL/6J wild-type mice underwent whole body irradiation followed by BMT into the retro-orbital sinus from green fluorescent protein (GFP)-transgenic donor mice. For the reverse BMT group, GFP-transgenic mice received BMT from C57BL/6J wild-type donor mice after irradiation. The supraspinatus tendon, infraspinatus tendon, and suprascapular nerve were surgically transected 3 weeks after transplantation. The rotator cuff muscles were harvested 13 weeks after transplantation for histologic analysis and GFP immunohistochemistry. RESULTS On histologic examination, both groups showed substantial fatty degeneration, fibrosis, and atrophy of the cuff muscles. The BMT group showed no noticeable GFP immunostaining, whereas the reverse BMT group showed significantly stronger GFP staining in most adipocytes (P < .001). However, both groups also showed that a small number of adipocytes originated from transplanted bone marrow cells. A small number of myocytes showed a large cytoplasmic lipid vacuole resembling adipocytes. CONCLUSIONS This study's findings suggest that most adipocytes in fatty degeneration of the rotator cuff muscles originate from sources other than bone marrow-derived stem cells, and there may be more than 1 source for the adipocytes.
Collapse
Affiliation(s)
- Lawrence V Klomps
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Naseem Zomorodi
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine Milton S. Hershey Medical Center, Hershey, PA, USA
| | - H Mike Kim
- Department of Orthopaedics and Rehabilitation, Penn State College of Medicine Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
144
|
Zhou H, Zhu J, Liu M, Wu Q, Dong N. Role of the protease corin in chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells. J Tissue Eng Regen Med 2017; 12:973-982. [PMID: 28714548 DOI: 10.1002/term.2514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/30/2017] [Accepted: 07/11/2017] [Indexed: 01/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have the potency to differentiate into chondrocytes, osteocytes and adipocytes. Corin is a cardiac protease that activates the natriuretic peptides, thereby regulating blood volume and pressure. In addition to the heart, corin gene upregulation was reported in bone marrow- and adipose tissue-derived MSCs that underwent osteogenic differentiation. To date, the biological significance of corin expression in MSC differentiation remains unknown. In this study we isolated and cultured human bone marrow-derived MSCs that were capable of undergoing chondrogenic, osteogenic and adipogenic lineage differentiation. By reverse transcription polymerase chain reaction (RT-PCR) and immunostaining, we found that corin expression was upregulated when these MSCs underwent chondrogenic, osteogenic and adipogenic differentiation. The upregulation of corin expression was most significant in the cells undergoing chondrogenic lineage differentiation. Silencing corin gene expression by small hairpin RNA in the MSCs inhibited chondrogenic, but not osteogenic and adipogenic, differentiation. These results suggest a novel function of corin in MSC differentiation and chondrocyte development.
Collapse
Affiliation(s)
- Haibin Zhou
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinsong Zhu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Liu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
- Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
145
|
Mortada I, Mortada R. Epigenetic changes in mesenchymal stem cells differentiation. Eur J Med Genet 2017; 61:114-118. [PMID: 29079547 DOI: 10.1016/j.ejmg.2017.10.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/30/2017] [Accepted: 10/22/2017] [Indexed: 01/09/2023]
Abstract
Epigenetic factors are known to play a major role in determining stem cell fate and differentiation. Mesenchymal stem cells are the most studied population of stem cells due to their important applications in experimental biology and regenerative medicine. After a brief overview on mesenchymal stem cells, this review aims to highlight the role of epigenetic changes on mesenchymal stem cells biology and differentiation protocols with a focus on osteocytic, chondrocytic and adipocytic differentiation. Chromatin remodeling, DNA methylation, histone modifications and miRNA expression will be investigated. The impact of epigenetics on transdifferentiation of mesenchymal stem cells will also be discussed. Indeed, epigenetic modulation appears to constitute a promising experimental target in stem cell basic and translational research.
Collapse
|
146
|
Secretion of interleukin-6 by bone marrow mesenchymal stem cells promotes metastasis in hepatocellular carcinoma. Biosci Rep 2017; 37:BSR20170181. [PMID: 28659496 PMCID: PMC5567045 DOI: 10.1042/bsr20170181] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/15/2017] [Accepted: 06/28/2017] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) interact with tumor cells and regulate
tumorigenesis and metastasis. As one of the important components of the tumor
microenvironment, MSC-secreted cytokines play a critical role in cancer
development. However, whether and how bone marrow MSCs (BMSCs) and their
secreted cytokines participate in hepatocellular carcinoma (HCC) progression,
still remains largely unknown. In the present study, we first measured the
concentration of interleukin-6 (IL-6) in BMSC conditioned medium (BMSC-CM).
Next, we assessed the changes of invasion ability in response to treatment of
BMSC-CM or recombinant IL-6 in two human HCC cell lines Bel-7404 and HepG2. Then
we analyzed the level of key components of the IL-6 signal pathway, including
IL-6 receptor and signal transducer (i.e. IL-6R and gp130), a transcription
factor STAT3 (signal transducer and activator of transcription 3), as well as
its target genes BCL2, CCND1, MCL1 and MMP2, in BMSC-CM or recombinant IL-6
treated Bel-7404 and HepG2 cells. Results showed that a considerable amount of
IL-6 was secreted by BMSCs, and BMSC-CM markedly elevated Bel-7404 cell invasion
rate and stimulated the signal transduction of IL-6/STAT3 pathway.
Neutralizing the secreted IL-6 bioactivity by the anti-IL-6 antibody diminished
the invasion-promoting effect and down-regulated IL-6/STAT3 pathway of
BMSC-CM treated Bel-7404 cells. In conclusion, we found that BMSCs may activate
the IL-6/STAT3 signaling pathway and promote cell invasion in Bel-7404
cells, suggesting that this protumor effect should be seriously considered
before clinical application of MSC-mediated cancer therapy.
Collapse
|
147
|
Zhou P, Liu Z, Li X, Zhang B, Wang X, Lan J, Shi Q, Li D, Ju X. Migration ability and Toll-like receptor expression of human mesenchymal stem cells improves significantly after three-dimensional culture. Biochem Biophys Res Commun 2017; 491:323-328. [PMID: 28734835 DOI: 10.1016/j.bbrc.2017.07.102] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
While the conventional two-dimensional (2D) culture protocol is well accepted for the culture of mesenchymal stem cells (MSCs), this method fails to recapitulate the in vivo native three-dimensional (3D) cellular microenvironment, and may result in phenotypic changes, and homing and migration capacity impairments. MSC preparation in 3D culture systems has been considered an attractive preparatory and delivery method recently. We seeded human umbilical cord-derived MSCs (hUCMSCs) in a 3D culture system with porcine acellular dermal matrix (PADM), and investigated the phenotypic changes, the expression changes of some important receptors, including Toll-like receptors (TLRs) and C-X-C chemokine receptor type 4 (CXCR4) when hUCMSCs were transferred from 2D to 3D systems, as well as the alterations in in vivo homing and migration potential. It was found that the percentage of CD105-positive cells decreased significantly, whereas that of CD34- and CD271-positive cells increased significantly in 3D culture, compared to that in 2D culture. The mRNA and protein expression levels of TLR2, TLR3, TLR4, TLR6, and CXCR4 in hUCMSCs were increased significantly upon culturing with PADM for 3 days, compared to the levels in 2D culture. The numbers of migratory 3D hUCMSCs in the heart, liver, spleen, and bone marrow were significantly greater than the numbers of 2D hUCMSCs, and the worst migration occurred in 3D + AMD3100 (CXCR4 antagonist) hUCMSCs. These results suggested that 3D culture of hUCMSCs with PADM could alter the phenotypic characteristics of hUCMSCs, increase their TLR and CXCR4 expression levels, and promote their migratory and homing capacity in which CXCR4 plays an important role.
Collapse
Affiliation(s)
- Panpan Zhou
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Zilin Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xue Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Bing Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiaoyuan Wang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jing Lan
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Qing Shi
- Cryomedicine Lab, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Dong Li
- Cryomedicine Lab, Qilu Hospital of Shandong University, Jinan 250012, China.
| | - Xiuli Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|
148
|
Abstract
White adipose tissue is a remarkably expandable organ with results in the last decade showing that human white adipocytes are continuously turned over during the entire life-span. Data primarily in murine models have demonstrated that adipocytes are derived from precursors present mainly in the perivascular areas of adipose tissue but their precise origin remains unclear. Subsets of cells present in bone marrow display a multipotent differentiation capacity which has prompted the hypothesis that bone marrow-derived cells (BMDCs) may also contribute to the adipocyte pool present in peripheral fat depots. This notion was initially demonstrated in a murine transplantation model, however, subsequent animal studies have been conflicting resulting in a debate of whether BMDCs actually differentiate into adipocytes or just fuse with resident fat cells. This controversy was recently resolved in 2 studies of human subjects undergoing bone marrow transplantation. Using a combination of different assays these data suggest that bone marrow contributes to at least 10% of the adipocyte pool. This proportion is doubled in obesity, suggesting that BMDCs may constitute a reserve pool for adipogenesis, particularly upon weight gain. This review discusses the possible mechanisms and relevance of these findings for human pathophysiology.
Collapse
Affiliation(s)
- Peter Arner
- Karolinska Institutet, Department of Medicine (H7), Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine (H7), Karolinska University Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
149
|
Adhesion profile and differentiation capacity of human adipose tissue derived mesenchymal stem cells grown on metal ion (Zn, Ag and Cu) doped hydroxyapatite nano-coated surfaces. Colloids Surf B Biointerfaces 2017; 155:415-428. [DOI: 10.1016/j.colsurfb.2017.04.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 01/31/2023]
|
150
|
Wobma HM, Liu D, Vunjak-Novakovic G. Paracrine Effects of Mesenchymal Stromal Cells Cultured in Three-Dimensional Settings on Tissue Repair. ACS Biomater Sci Eng 2017; 4:1162-1175. [PMID: 33418654 DOI: 10.1021/acsbiomaterials.7b00005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a promising cell source for promoting tissue repair, due to their ability to release growth, angiogenic, and immunomodulatory factors. However, when injected as a suspension, these cells suffer from poor survival and localization, and suboptimal release of paracrine factors. While there have been attempts to overcome these limitations by modifying MSCs themselves, a more versatile solution is to grow them in three dimensions, as aggregates or embedded into biomaterials. Here we review the mechanisms by which 3D culture can influence the regenerative capacity of undifferentiated MSCs, focusing on recent examples from the literature. We further discuss how knowledge of these mechanisms can lead to strategic design of MSC therapies that overcome some of the challenges to their effective translation.
Collapse
|