1
|
Plissonneau C, Santosa S. Regional primary preadipocyte characteristics in humans with obesity and type 2 diabetes mellitus. Heliyon 2024; 10:e39710. [PMID: 39553621 PMCID: PMC11564010 DOI: 10.1016/j.heliyon.2024.e39710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
The excessive accumulation of adipose tissue in obesity appears to result in adipose tissue dysfunction perpetuating the onset of obesity-related diseases, including type 2 diabetes (T2DM). In humans, adipose tissue is stored in several depots including subcutaneous and visceral. These depots contribute to the pathology of obesity differently owing to differences in the tissue microenvironment, a main one being preadipocyte function. In examining adipocyte and preadipocyte characteristics, many have used the 3T3-L1 murine cell lines. Though these cell lines provide valuable mechanistic data, the results remain to be translated to humans. Experiments using primary human preadipocytes has shown that obesity and T2DM impact preadipocyte phenotypes. The objective of this review is to describe the differences in regional characteristics of primary preadipocytes collected from humans with obesity and to discuss how these characteristics might be affected in type 2 diabetes mellitus. In doing so, we will show that the characteristics of regional primary preadipocytes in humans are differentially affected by obesity and the development of T2DM.
Collapse
Affiliation(s)
- Claire Plissonneau
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada
- Metabolism, Obesity, and Nutrition Lab, School of Health, Concordia University, Montreal, Quebec, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Sylvia Santosa
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada
- Metabolism, Obesity, and Nutrition Lab, School of Health, Concordia University, Montreal, Quebec, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
2
|
Christy BA, Herzig MC, Wu X, Mohammadipoor A, McDaniel JS, Bynum JA. Cell Therapies for Acute Radiation Syndrome. Int J Mol Sci 2024; 25:6973. [PMID: 39000080 PMCID: PMC11241804 DOI: 10.3390/ijms25136973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
The risks of severe ionizing radiation exposure are increasing due to the involvement of nuclear powers in combat operations, the increasing use of nuclear power, and the existence of terrorist threats. Exposure to a whole-body radiation dose above about 0.7 Gy results in H-ARS (hematopoietic acute radiation syndrome), which is characterized by damage to the hematopoietic system; higher doses result in further damage to the gastrointestinal and nervous systems. Only a few medical countermeasures for ARS are currently available and approved for use, although others are in development. Cell therapies (cells or products produced by cells) are complex therapeutics that show promise for the treatment of radiation injury and have been shown to reduce mortality and morbidity in animal models. Since clinical trials for ARS cannot be ethically conducted, animal testing is extremely important. Here, we describe cell therapies that have been tested in animal models. Both cells and cell products appear to promote survival and lessen tissue damage after whole-body irradiation, although the mechanisms are not clear. Because radiation exposure often occurs in conjunction with other traumatic injuries, animal models of combined injury involving radiation and future countermeasure testing for these complex medical problems are also discussed.
Collapse
Affiliation(s)
- Barbara A Christy
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Maryanne C Herzig
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Xiaowu Wu
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Arezoo Mohammadipoor
- Hemorrhage and Vascular Dysfunction, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Jennifer S McDaniel
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - James A Bynum
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
- Department of Surgery, UT Health San Antonio, San Antonio, TX 78229, USA
- Trauma Research and Combat Casualty Care Collaborative, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
3
|
Nakamura Y, Niho S, Shimizu Y. Cell-Based Therapy for Fibrosing Interstitial Lung Diseases, Current Status, and Potential Applications of iPSC-Derived Cells. Cells 2024; 13:893. [PMID: 38891026 PMCID: PMC11172081 DOI: 10.3390/cells13110893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Fibrosing interstitial lung diseases (FILDs), e.g., due to idiopathic pulmonary fibrosis (IPF), are chronic progressive diseases with a poor prognosis. The management of these diseases is challenging and focuses mainly on the suppression of progression with anti-fibrotic drugs. Therefore, novel FILD treatments are needed. In recent years, cell-based therapy with various stem cells has been investigated for FILD, and the use of mesenchymal stem cells (MSCs) has been widely reported and clinical studies are also ongoing. Induced pluripotent stem cells (iPSCs) have also been reported to have an anti-fibrotic effect in FILD; however, these have not been as well studied as MSCs in terms of the mechanisms and side effects. While MSCs show a potent anti-fibrotic effect, the possibility of quality differences between donors and a stable supply in the case of donor shortage or reduced proliferative capacity after cell passaging needs to be considered. The application of iPSC-derived cells has the potential to overcome these problems and may lead to consistent quality of the cell product and stable product supply. This review provides an overview of iPSCs and FILD, followed by the current status of cell-based therapy for FILD, and then discusses the possibilities and perspectives of FILD therapy with iPSC-derived cells.
Collapse
Affiliation(s)
- Yusuke Nakamura
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
- Center of Regenerative Medicine, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
| | - Seiji Niho
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
| | - Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
- Center of Regenerative Medicine, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
- Respiratory Endoscopy Center, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
| |
Collapse
|
4
|
Haskell A, White BP, Rogers RE, Goebel E, Lopez MG, Syvyk AE, de Oliveira DA, Barreda HA, Benton J, Benavides OR, Dalal S, Bae E, Zhang Y, Maitland K, Nikolov Z, Liu F, Lee RH, Kaunas R, Gregory CA. Scalable manufacture of therapeutic mesenchymal stromal cell products on customizable microcarriers in vertical wheel bioreactors that improve direct visualization, product harvest, and cost. Cytotherapy 2024; 26:372-382. [PMID: 38363250 PMCID: PMC11057043 DOI: 10.1016/j.jcyt.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Human mesenchymal stromal cells (hMSCs) and their secreted products show great promise for treatment of musculoskeletal injury and inflammatory or immune diseases. However, the path to clinical utilization is hampered by donor-tissue variation and the inability to manufacture clinically relevant yields of cells or their products in a cost-effective manner. Previously we described a method to produce chemically and mechanically customizable gelatin methacryloyl (GelMA) microcarriers for culture of hMSCs. Herein, we demonstrate scalable GelMA microcarrier-mediated expansion of induced pluripotent stem cell (iPSC)-derived hMSCs (ihMSCs) in 500 mL and 3L vertical wheel bioreactors, offering several advantages over conventional microcarrier and monolayer-based expansion strategies. METHODS Human mesenchymal stromal cells derived from induced pluripotent cells were cultured on custom-made spherical gelatin methacryloyl microcarriers in single-use vertical wheel bioreactors (PBS Biotech). Cell-laden microcarriers were visualized using confocal microscopy and elastic light scattering methodologies. Cells were assayed for viability and differentiation potential in vitro by standard methods. Osteogenic cell matrix derived from cells was tested in vitro for osteogenic healing using a rodent calvarial defect assay. Immune modulation was assayed with an in vivo peritonitis model using Zymozan A. RESULTS The optical properties of GelMA microcarriers permit noninvasive visualization of cells with elastic light scattering modalities, and harvest of product is streamlined by microcarrier digestion. At volumes above 500 mL, the process is significantly more cost-effective than monolayer culture. Osteogenic cell matrix derived from ihMSCs expanded on GelMA microcarriers exhibited enhanced in vivo bone regenerative capacity when compared to bone morphogenic protein 2, and the ihMSCs exhibited superior immunosuppressive properties in vivo when compared to monolayer-generated ihMSCs. CONCLUSIONS These results indicate that the cell expansion strategy described here represents a superior approach for efficient generation, monitoring and harvest of therapeutic MSCs and their products.
Collapse
Affiliation(s)
- Andrew Haskell
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Berkley P White
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Robert E Rogers
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Erin Goebel
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA; Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Megan G Lopez
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Andrew E Syvyk
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA
| | - Daniela A de Oliveira
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA; Biological and Agricultural Engineering, Texas A&M University, College Station, Texas, USA
| | - Heather A Barreda
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Joshua Benton
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Oscar R Benavides
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Sujata Dalal
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - EunHye Bae
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Yu Zhang
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Kristen Maitland
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA; Imaging Program, Chan Zuckerberg Initiative, Redwood City, California, USA
| | - Zivko Nikolov
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA; Biological and Agricultural Engineering, Texas A&M University, College Station, Texas, USA
| | - Fei Liu
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA
| | - Roland Kaunas
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA.
| | - Carl A Gregory
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, Texas, USA.
| |
Collapse
|
5
|
Bai X, Chen T, Li Y, Ge X, Qiu C, Gou H, Wei S, Liu T, Yang W, Yang L, Liang Y, Jia Z, Lv L, Li T. PD-L1 expression levels in mesenchymal stromal cells predict their therapeutic values for autoimmune hepatitis. Stem Cell Res Ther 2023; 14:370. [PMID: 38111045 PMCID: PMC10729378 DOI: 10.1186/s13287-023-03594-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Autoimmune hepatitis is a chronic inflammatory hepatic disorder with no effective treatment. Mesenchymal stromal cells (MSCs) have emerged as a promising treatment owing to their unique advantages. However, their heterogeneity is hampering use in clinical applications. METHODS Wharton's jelly derived MSCs (WJ-MSCs) were isolated from 58 human donors using current good manufacturing practice conditions. Gene expression profiles of the WJ-MSCs were analyzed by transcriptome and single-cell RNA-sequencing (scRNA-seq), and subsequent functional differences were assessed. Expression levels of programmed death-ligand 1 (PD-L1) were used as an indicator to screen WJ-MSCs with varied immunomodulation activities and assessed their corresponding therapeutic effects in a mouse model of concanavalin A-induced autoimmune hepatitis. RESULTS The 58 different donor-derived WJ-MSCs were grouped into six gene expression profile clusters. The gene in different clusters displayed obvious variations in cell proliferation, differentiation bias, trophic factor secretion, and immunoregulation. Data of scRNA-seq revealed four distinct WJ-MSCs subpopulations. Notably, the different immunosuppression capacities of WJ-MSCs were positively correlated with PD-L1 expression. WJ-MSCs with high expression of PD-L1 were therapeutically superior to WJ-MSCs with low PD-L1 expression in treating autoimmune hepatitis. CONCLUSION PD-L1 expression levels of WJ-MSCs could be regarded as an indicator to choose optimal MSCs for treating autoimmune disease. These findings provided novel insights into the quality control of MSCs and will inform improvements in the therapeutic benefits of MSCs.
Collapse
Affiliation(s)
- Xilong Bai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
- Department of Hematology, Xi'an International Medical Center Hospital, Xi'an, 710100, Shaanxi, China
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Yuqi Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China
| | - Xiaofan Ge
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Caie Qiu
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Huili Gou
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Sili Wei
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Tingting Liu
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Wei Yang
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Liting Yang
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China
| | - Yingmin Liang
- Department of Hematology, Xi'an International Medical Center Hospital, Xi'an, 710100, Shaanxi, China
| | - Zhansheng Jia
- Department of Infection and Liver Disease, Xi'an International Medical Center Hospital, Xi'an, 710100, Shaanxi, China
| | - Liangshan Lv
- Department of Minimally Invasive Interventional Radiology, Xi'an Gaoxin Hospital, Xi'an, , 710075, Shaanxi, China
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, Yunnan, China.
- Xi'an ChaoYue Stem Cell Co., Ltd, Xi'an, 710100, Shaanxi, China.
| |
Collapse
|
6
|
Burns JS. The Evolving Landscape of Potency Assays. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:165-189. [PMID: 37258790 DOI: 10.1007/978-3-031-30040-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
There is a "goldilocks" aspect to potency assays. On the one hand, a comprehensive evaluation of the cell product with detailed quantitative measurement of the critical quality attribute/s of the desired biological activity is required. On the other hand, the potency assay benefits from simplification and lean approaches that avoid unnecessary complication and enhance robustness, to provide a reproducible and scalable product. There is a need to balance insightful knowledge of complex biological healing processes with straightforward manufacture of an advanced therapeutic medicinal product (ATMP) that can be administered in a trustworthy cost-effective manner. While earlier chapters within this book have highlighted numerous challenges facing the potency assay conundrum, this chapter offers a forward-looking perspective regarding the many recent advances concerning acellular products, cryopreservation, induced MSC, cell priming, nanotechnology, 3D culture, regulatory guidelines and evolving institutional roles, that are likely to facilitate potency assay development in the future.
Collapse
Affiliation(s)
- Jorge S Burns
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
7
|
Steens J, Klein D. HOX genes in stem cells: Maintaining cellular identity and regulation of differentiation. Front Cell Dev Biol 2022; 10:1002909. [PMID: 36176275 PMCID: PMC9514042 DOI: 10.3389/fcell.2022.1002909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Stem cells display a unique cell type within the body that has the capacity to self-renew and differentiate into specialized cell types. Compared to pluripotent stem cells, adult stem cells (ASC) such as mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) exhibit restricted differentiation capabilities that are limited to cell types typically found in the tissue of origin, which implicates that there must be a certain code or priming determined by the tissue of origin. HOX genes, a subset of homeobox genes encoding transcription factors that are generally repressed in undifferentiated pluripotent stem cells, emerged here as master regulators of cell identity and cell fate during embryogenesis, and in maintaining this positional identity throughout life as well as specifying various regional properties of respective tissues. Concurrently, intricate molecular circuits regulated by diverse stem cell-typical signaling pathways, balance stem cell maintenance, proliferation and differentiation. However, it still needs to be unraveled how stem cell-related signaling pathways establish and regulate ASC-specific HOX expression pattern with different temporal-spatial topography, known as the HOX code. This comprehensive review therefore summarizes the current knowledge of specific ASC-related HOX expression patterns and how these were integrated into stem cell-related signaling pathways. Understanding the mechanism of HOX gene regulation in stem cells may provide new ways to manipulate stem cell fate and function leading to improved and new approaches in the field of regenerative medicine.
Collapse
|
8
|
Szewc M, Radzikowska-Bűchner E, Wdowiak P, Kozak J, Kuszta P, Niezabitowska E, Matysiak J, Kubiński K, Masłyk M. MSCs as Tumor-Specific Vectors for the Delivery of Anticancer Agents-A Potential Therapeutic Strategy in Cancer Diseases: Perspectives for Quinazoline Derivatives. Int J Mol Sci 2022; 23:2745. [PMID: 35269887 PMCID: PMC8911180 DOI: 10.3390/ijms23052745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered to be a powerful tool in the treatment of various diseases. Scientists are particularly interested in the possibility of using MSCs in cancer therapy. The research carried out so far has shown that MSCs possess both potential pro-oncogenic and anti-oncogenic properties. It has been confirmed that MSCs can regulate tumor cell growth through a paracrine mechanism, and molecules secreted by MSCs can promote or block a variety of signaling pathways. These findings may be crucial in the development of new MSC-based cell therapeutic strategies. The abilities of MSCs such as tumor tropism, deep migration and immune evasion have evoked considerable interest in their use as tumor-specific vectors for small-molecule anticancer agents. Studies have shown that MSCs can be successfully loaded with chemotherapeutic drugs such as gemcitabine and paclitaxel, and can release them at the site of primary and metastatic neoplasms. The inhibitory effect of MSCs loaded with anti-cancer agents on the proliferation of cancer cells has also been observed. However, not all known chemotherapeutic agents can be used in this approach, mainly due to their cytotoxicity towards MSCs and insufficient loading and release capacity. Quinazoline derivatives appear to be an attractive choice for this therapeutic solution due to their biological and pharmacological properties. There are several quinazolines that have been approved for clinical use as anticancer drugs by the US Food and Drug Administration (FDA). It gives hope that the synthesis of new quinazoline derivatives and the development of methods of their application may contribute to the establishment of highly effective therapies for oncological patients. However, a deeper understanding of interactions between MSCs and tumor cells, and the exploration of the possibilities of using quinazoline derivatives in MSC-based therapy is necessary to achieve this goal. The aim of this review is to discuss the prospects for using MSC-based cell therapy in cancer treatment and the potential use of quinazolines in this procedure.
Collapse
Affiliation(s)
- Monika Szewc
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Elżbieta Radzikowska-Bűchner
- Department of Plastic, Reconstructive and Maxillary Surgery, Central Clinical Hospital MSWiA, 02-507 Warsaw, Poland;
| | - Paulina Wdowiak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Joanna Kozak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Piotr Kuszta
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Ewa Niezabitowska
- Department of Urology and Urological Oncology, Multidisciplinary Hospital in Lublin, 20-400 Lublin, Poland;
| | - Joanna Matysiak
- Department of Chemistry, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Konrad Kubiński
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland;
| | - Maciej Masłyk
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland;
| |
Collapse
|
9
|
Zhang C, Han X, Liu J, Chen L, Lei Y, Chen K, Si J, Wang TY, Zhou H, Zhao X, Zhang X, An Y, Li Y, Wang QF. Single-cell Transcriptomic Analysis Reveals the Cellular Heterogeneity of Mesenchymal Stem Cells. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:70-86. [PMID: 35123072 PMCID: PMC9510874 DOI: 10.1016/j.gpb.2022.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 01/04/2022] [Accepted: 01/27/2022] [Indexed: 01/04/2023]
Abstract
Ex vivo-expanded mesenchymal stem cells (MSCs) have been demonstrated to be a heterogeneous mixture of cells exhibiting varying proliferative, multipotential, and immunomodulatory capacities. However, the exact characteristics of MSCs remain largely unknown. By single-cell RNA sequencing of 61,296 MSCs derived from bone marrow and Wharton’s jelly, we revealed five distinct subpopulations. The developmental trajectory of these five MSC subpopulations was mapped, revealing a differentiation path from stem-like active proliferative cells (APCs) to multipotent progenitor cells, followed by branching into two paths: 1) unipotent preadipocytes or 2) bipotent prechondro-osteoblasts that were subsequently differentiated into unipotent prechondrocytes. The stem-like APCs, expressing the perivascular mesodermal progenitor markers CSPG4/MCAM/NES, uniquely exhibited strong proliferation and stemness signatures. Remarkably, the prechondrocyte subpopulation specifically expressed immunomodulatory genes and was able to suppress activated CD3+ T cell proliferation in vitro, supporting the role of this population in immunoregulation. In summary, our analysis mapped the heterogeneous subpopulations of MSCs and identified two subpopulations with potential functions in self-renewal and immunoregulation. Our findings advance the definition of MSCs by identifying the specific functions of their heterogeneous cellular composition, allowing for more specific and effective MSC application through the purification of their functional subpopulations.
Collapse
Affiliation(s)
- Chen Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China; Qingdao Key Lab of Mitochondrial Medicine, Qingdao 266035, China
| | - Xueshuai Han
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingkun Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Lei
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kunying Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia Si
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tian-Yi Wang
- International Department, Liangxiang Campus, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Hui Zhou
- Yihua Biotechnology Co., Ltd., Beijing 100041, China
| | - Xiaoyun Zhao
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China; Qingdao Key Lab of Mitochondrial Medicine, Qingdao 266035, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China
| | - Yihua An
- Department of Functional Neurosurgery, Third Medical Center, General Hospital of Chinese PLA, Beijing 100039, China
| | - Yueying Li
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qian-Fei Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
10
|
Gallo A, Cuscino N, Contino F, Bulati M, Pampalone M, Amico G, Zito G, Carcione C, Centi C, Bertani A, Conaldi PG, Miceli V. Changes in the Transcriptome Profiles of Human Amnion-Derived Mesenchymal Stromal/Stem Cells Induced by Three-Dimensional Culture: A Potential Priming Strategy to Improve Their Properties. Int J Mol Sci 2022; 23:ijms23020863. [PMID: 35055049 PMCID: PMC8778321 DOI: 10.3390/ijms23020863] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/08/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are believed to function in vivo as a homeostatic tool that shows therapeutic properties for tissue repair/regeneration. Conventionally, these cells are expanded in two-dimensional (2D) cultures, and, in that case, MSCs undergo genotypic/phenotypic changes resulting in a loss of their therapeutic capabilities. Moreover, several clinical trials using MSCs have shown controversial results with moderate/insufficient therapeutic responses. Different priming methods were tested to improve MSC effects, and three-dimensional (3D) culturing techniques were also examined. MSC spheroids display increased therapeutic properties, and, in this context, it is crucial to understand molecular changes underlying spheroid generation. To address these limitations, we performed RNA-seq on human amnion-derived MSCs (hAMSCs) cultured in both 2D and 3D conditions and examined the transcriptome changes associated with hAMSC spheroid formation. We found a large number of 3D culture-sensitive genes and identified selected genes related to 3D hAMSC therapeutic effects. In particular, we observed that these genes can regulate proliferation/differentiation, as well as immunomodulatory and angiogenic processes. We validated RNA-seq results by qRT-PCR and methylome analysis and investigation of secreted factors. Overall, our results showed that hAMSC spheroid culture represents a promising approach to cell-based therapy that could significantly impact hAMSC application in the field of regenerative medicine.
Collapse
Affiliation(s)
- Alessia Gallo
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Nicola Cuscino
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Flavia Contino
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Matteo Bulati
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Mariangela Pampalone
- Fondazione Ri.MED, 90127 Palermo, Italy; (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Giandomenico Amico
- Fondazione Ri.MED, 90127 Palermo, Italy; (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Giovanni Zito
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | | | - Claudio Centi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Alessandro Bertani
- Thoracic Surgery and Lung Transplantation Unit, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy;
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (A.G.); (N.C.); (F.C.); (M.B.); (G.Z.); (C.C.); (P.G.C.)
- Correspondence: ; Tel.: +39-09-1219-2430
| |
Collapse
|
11
|
Sun C, Zhang K, Yue J, Meng S, Zhang X. Deconstructing transcriptional variations and their effects on immunomodulatory function among human mesenchymal stromal cells. Stem Cell Res Ther 2021; 12:53. [PMID: 33422149 PMCID: PMC7796611 DOI: 10.1186/s13287-020-02121-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Background Mesenchymal stromal cell (MSC)-based therapies are being actively investigated in various inflammatory disorders. However, functional variability among MSCs cultured in vitro will lead to distinct therapeutic efficacies. Until now, the mechanisms behind immunomodulatory functional variability in MSCs are still unclear. Methods We systemically investigated transcriptomic variations among MSC samples derived from multiple tissues to reveal their effects on immunomodulatory functions of MSCs. We then analyzed transcriptomic changes of MSCs licensed with INFγ to identify potential molecular mechanisms that result in distinct MSC samples with different immunomodulatory potency. Results MSCs were clustered into distinct groups showing different functional enrichment according to transcriptomic patterns. Differential expression analysis indicated that different groups of MSCs deploy common regulation networks in response to inflammatory stimulation, while expression variation of genes in the networks could lead to different immunosuppressive capability. These different responsive genes also showed high expression variability among unlicensed MSC samples. Finally, a gene panel was derived from these different responsive genes and was able to regroup unlicensed MSCs with different immunosuppressive potencies. Conclusion This study revealed genes with expression variation that contribute to immunomodulatory functional variability of MSCs and provided us a strategy to identify candidate markers for functional variability assessment of MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02121-8.
Collapse
Affiliation(s)
- Changbin Sun
- BGI-Shenzhen, Jinsha Road, Dapeng New District, Shenzhen, 518083, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China
| | - Kehua Zhang
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Jianhui Yue
- BGI-Shenzhen, Jinsha Road, Dapeng New District, Shenzhen, 518083, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.,Section of Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Shufang Meng
- Cell Collection and Research Center, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Xi Zhang
- BGI-Shenzhen, Jinsha Road, Dapeng New District, Shenzhen, 518083, China. .,China National GeneBank, BGI-Shenzhen, Shenzhen, 518120, China.
| |
Collapse
|
12
|
Höving AL, Sielemann K, Greiner JFW, Kaltschmidt B, Knabbe C, Kaltschmidt C. Transcriptome Analysis Reveals High Similarities between Adult Human Cardiac Stem Cells and Neural Crest-Derived Stem Cells. BIOLOGY 2020; 9:biology9120435. [PMID: 33271866 PMCID: PMC7761507 DOI: 10.3390/biology9120435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
For the identification of a stem cell population, the comparison of transcriptome data enables the simultaneous analysis of tens of thousands of molecular markers and thus enables the precise distinction of even closely related populations. Here, we utilized global gene expression profiling to compare two adult human stem cell populations, namely neural crest-derived inferior turbinate stem cells (ITSCs) of the nasal cavity and human cardiac stem cells (hCSCs) from the heart auricle. We detected high similarities between the transcriptomes of both stem cell populations, particularly including a range of neural crest-associated genes. However, global gene expression likewise reflected differences between the stem cell populations with regard to their niches of origin. In a broader analysis, we further identified clear similarities between ITSCs, hCSCs and other adherent stem cell populations compared to non-adherent hematopoietic progenitor cells. In summary, our observations reveal high similarities between adult human cardiac stem cells and neural crest-derived stem cells from the nasal cavity, which include a shared relation to the neural crest. The analyses provided here may help to understand underlying molecular regulators determining differences between adult human stem cell populations.
Collapse
Affiliation(s)
- Anna L. Höving
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
- Correspondence: (A.L.H.); (C.K.)
| | - Katharina Sielemann
- Genetics and Genomics of Plants, Center for Biotechnology (CeBiTec), Bielefeld University, 33615 Bielefeld, Germany;
- Graduate School DILS, Bielefeld Institute for Bioinformatics Infrastructure (BIBI), Bielefeld University, 33615 Bielefeld, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
- AG Molecular Neurobiology, Bielefeld University, 33615 Bielefeld, Germany
| | - Cornelius Knabbe
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany;
| | - Christian Kaltschmidt
- Department of Cell Biology, Bielefeld University, 33615 Bielefeld, Germany; (J.F.W.G.); (B.K.)
- Correspondence: (A.L.H.); (C.K.)
| |
Collapse
|
13
|
Baker EK, Jacobs SE, Lim R, Wallace EM, Davis PG. Cell therapy for the preterm infant: promise and practicalities. Arch Dis Child Fetal Neonatal Ed 2020; 105:563-568. [PMID: 32253200 DOI: 10.1136/archdischild-2019-317896] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023]
Abstract
Recent decades have seen the rapid progress of neonatal intensive care, and the survival rates of the most preterm infants are improving. This improvement is associated with changing patterns of morbidity and new phenotypes of bronchopulmonary dysplasia and preterm brain injury are recognised. Inflammation and immaturity are known contributors to their pathogenesis. However, a new phenomenon, the exhaustion of progenitor cells is emerging as an important factor. Current therapeutic approaches do not adequately address these new mechanisms of injury. Cell therapy, that is the use of stem and stem-like cells, with its potential to both repair and prevent injury, offers a new approach to these challenging conditions. This review will examine the rationale for cell therapy in the extremely preterm infant, the preclinical and early clinical evidence to support its use in bronchopulmonary dysplasia and preterm brain injury. Finally, it will address the challenges in translating cell therapy from the laboratory to early clinical trials.
Collapse
Affiliation(s)
- Elizabeth K Baker
- Newborn Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia .,Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Susan E Jacobs
- Newborn Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Euan M Wallace
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Peter G Davis
- Newborn Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Miranda VHS, Gomes TR, Eller DE, Ferraz LDCN, Chaves AT, Bicalho KA, Silva CEC, Birbrair A, Pascoal Xavier MA, de Goes AM, Corrêa-Oliveira R, Alves ÉAR, Bozzi A. Liver damage in schistosomiasis is reduced by adipose tissue-derived stem cell therapy after praziquantel treatment. PLoS Negl Trop Dis 2020; 14:e0008635. [PMID: 32853206 PMCID: PMC7480869 DOI: 10.1371/journal.pntd.0008635] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 09/09/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
Background In view of the potential immunosuppressive and regenerative properties of mesenchymal stem cells (MSC), we investigated whether transplantation of adipose tissue-derived stem cells (ASC) could be used to control the granulomatous reaction in the liver of mice infected with Schistosoma mansoni after Praziquantel (PZQ) treatment. Methodology/Prinicpal findings C57BL/6 mice infected with S. mansoni were treated with PZQ and transplanted intravenously with ASC from uninfected mice. Liver morpho-physiological and immunological analyses were performed. The combined PZQ/ASC therapy significantly reduced the volume of hepatic granulomas, as well as liver damage as measured by ALT levels. We also observed that ASC accelerated the progression of the granulomatous inflammation to the advanced/curative phase. The faster healing interfered with the expression of CD28 and CTLA-4 molecules in CD4+ T lymphocytes, and the levels of IL-10 and IL-17 cytokines, mainly in the livers of PZQ/ASC-treated mice. Conclusions Our results show that ASC therapy after PZQ treatment results in smaller granulomas with little tissue damage, suggesting the potential of ASC for the development of novel therapeutic approaches to minimize hepatic lesions as well as a granulomatous reaction following S. mansoni infection. Further studies using the chronic model of schistosomiasis are required to corroborate the therapeutic use of ASC for schistosomiasis. Schistosomiasis is the second most prevalent parasitic disease in the world and is caused by the Schistosoma trematode. This disease is characterized by a granulomatous reaction around parasite eggs trapped in the tissues. The liver is one of the most affected organs and can develop severe fibrosis. Praziquantel (PZQ) is the treatment for schistosomiasis and kills the adult the worm; however, inflammation still persists around the eggs in the tissues. Mesenchymal stem cells (MSC) have been extensively studied as an alternative therapy to repair tissues and to stop inflammation due to their potential to differentiate in several cells types (bone, cartilage, fat, tendon, muscle, and marrow stroma), and to interfere with immune responses. This scenario has motivated the authors to investigate the use of MSC extract from adipose tissue (ASC) associated with PZQ to treat schistosomiasis. Briefly, mice were treated with PZQ followed by ASC injection showing significant reduction of the granulomas and normal levels of the enzyme alanine aminotransferase, an indicator of liver damage. These results suggest that ASC has the potential to be used as a novel therapeutic approach to control inflammation following infection by S. mansoni or liver disorders. Although the findings are promising, further studies using the chronic model of schistosomiasis are required to confirm using ASC for schistosomiasis therapy.
Collapse
Affiliation(s)
| | - Talita Rocha Gomes
- Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Morfologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | - Alexander Birbrair
- Departamento de Patologia, ICB, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Antônio Pascoal Xavier
- Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
- Faculdade de Medicina, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Adriana Bozzi
- Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, Bahia, Brazil
- * E-mail:
| |
Collapse
|
15
|
Therapeutic Mesenchymal Stromal Cells for Immunotherapy and for Gene and Drug Delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:204-224. [PMID: 32071924 PMCID: PMC7012781 DOI: 10.1016/j.omtm.2020.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) possess several fairly unique properties that, when combined, make them ideally suited for cellular-based immunotherapy and as vehicles for gene and drug delivery for a wide range of diseases and disorders. Key among these are: (1) their relative ease of isolation from a variety of tissues; (2) the ability to be expanded in culture without a loss of functionality, a property that varies to some degree with tissue source; (3) they are relatively immune-inert, perhaps obviating the need for precise donor/recipient matching; (4) they possess potent immunomodulatory functions that can be tailored by so-called licensing in vitro and in vivo; (5) the efficiency with which they can be modified with viral-based vectors; and (6) their almost uncanny ability to selectively home to damaged tissues, tumors, and metastases following systemic administration. In this review, we summarize the latest research in the immunological properties of MSCs, their use as immunomodulatory/anti-inflammatory agents, methods for licensing MSCs to customize their immunological profile, and their use as vehicles for transferring both therapeutic genes in genetic disease and drugs and genes designed to destroy tumor cells.
Collapse
|
16
|
Rockel JS, Rabani R, Viswanathan S. Anti-fibrotic mechanisms of exogenously-expanded mesenchymal stromal cells for fibrotic diseases. Semin Cell Dev Biol 2019; 101:87-103. [PMID: 31757583 DOI: 10.1016/j.semcdb.2019.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/11/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022]
Abstract
Most chronic diseases involving inflammation have a fibrotic component that involves remodeling and excess accumulation of extracellular matrix components. Left unchecked, fibrosis leads to organ failure and death. Mesenchymal stromal cells (MSCs) are emerging as a potent cell-based therapy for a wide spectrum of fibrotic conditions due to their immunomodulatory, anti-inflammatory and anti-fibrotic properties. This review provides an overview of known mechanisms by which MSCs mediate their anti-fibrotic actions and in relation to animal models of pulmonary, liver, renal and cardiac fibrosis. Recent MSC clinical trials results in liver, lung, skin, kidney and hearts are discussed and next steps for future MSC-based therapies including pre-activated or genetically-modified cells, or extracellular vesicles are also considered.
Collapse
Affiliation(s)
- Jason S Rockel
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| | - Razieh Rabani
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
17
|
Mammadova A, Carels CEL, Zhou J, Gilissen C, Helmich MPAC, Bian Z, Zhou H, Von den Hoff JW. Deregulated Adhesion Program in Palatal Keratinocytes of Orofacial Cleft Patients. Genes (Basel) 2019; 10:genes10110836. [PMID: 31652793 PMCID: PMC6895790 DOI: 10.3390/genes10110836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/17/2019] [Accepted: 10/19/2019] [Indexed: 01/10/2023] Open
Abstract
Orofacial clefts (OFCs) are the most frequent craniofacial birth defects. An orofacial cleft (OFC) occurs as a result of deviations in palatogenesis. Cell proliferation, differentiation, adhesion, migration and apoptosis are crucial in palatogenesis. We hypothesized that deregulation of these processes in oral keratinocytes contributes to OFC. We performed microarray expression analysis on palatal keratinocytes from OFC and non-OFC individuals. Principal component analysis showed a clear difference in gene expression with 24% and 17% for the first and second component, respectively. In OFC cells, 228 genes were differentially expressed (p < 0.001). Gene ontology analysis showed enrichment of genes involved in β1 integrin-mediated adhesion and migration, as well as in P-cadherin expression. A scratch assay demonstrated reduced migration of OFC keratinocytes (343.6 ± 29.62 μm) vs. non-OFC keratinocytes (503.4 ± 41.81 μm, p < 0.05). Our results indicate that adhesion and migration are deregulated in OFC keratinocytes, which might contribute to OFC pathogenesis.
Collapse
Affiliation(s)
- Aysel Mammadova
- Department of Dentistry, Section Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Carine E L Carels
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium.
- Department of Oral Health Sciences, KU Leuven, 3000 Leuven, Belgium.
| | - Jie Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan 430079, China.
| | - Christian Gilissen
- Department of Human Genetics, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Maria P A C Helmich
- Department of Dentistry, Section Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Zhuan Bian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan 430079, China.
| | - Huiqing Zhou
- Department of Human Genetics, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences (RIMLS), P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Johannes W Von den Hoff
- Department of Dentistry, Section Orthodontics and Craniofacial Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Ménard C, Dulong J, Roulois D, Hébraud B, Verdière L, Pangault C, Sibut V, Bezier I, Bescher N, Monvoisin C, Gadelorge M, Bertheuil N, Flécher E, Casteilla L, Collas P, Sensebé L, Bourin P, Espagnolle N, Tarte K. Integrated transcriptomic, phenotypic, and functional study reveals tissue-specific immune properties of mesenchymal stromal cells. Stem Cells 2019; 38:146-159. [PMID: 31502731 DOI: 10.1002/stem.3077] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/08/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
Clinical-grade mesenchymal stromal cells (MSCs) can be expanded from bone marrow and adipose tissue to treat inflammatory diseases and degenerative disorders. However, the influence of their tissue of origin on their functional properties, including their immunosuppressive activity, remains unsolved. In this study, we produced paired bone marrow-derived mesenchymal stromal cell (BM-MSC) and adipose-derived stromal cell (ASC) batches from 14 healthy donors. We then compared them using transcriptomic, phenotypic, and functional analyses and validated our results on purified native MSCs to infer which differences were really endowed by tissue of origin. Cultured MSCs segregated together owing to their tissue of origin based on their gene expression profile analyzed using differential expression and weighted gene coexpression network analysis. This translated into distinct immune-related gene signatures, phenotypes, and functional cell interactions. Importantly, sorted native BM-MSCs and ASCs essentially displayed the same distinctive patterns than their in vitro-expanded counterparts. As a whole, ASCs exhibited an immune profile consistent with a stronger inhibition of immune response and a lower immunogenicity, supporting the use of adipose tissue as a valuable source for clinical applications.
Collapse
Affiliation(s)
- Cédric Ménard
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Joëlle Dulong
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - David Roulois
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Benjamin Hébraud
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Léa Verdière
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Céline Pangault
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,Pôle Biologie, CHU Rennes, Rennes, France
| | - Vonick Sibut
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Isabelle Bezier
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Nadège Bescher
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| | - Céline Monvoisin
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Mélanie Gadelorge
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Nicolas Bertheuil
- SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France.,Department of Plastic Surgery, CHU Rennes, Rennes, France
| | - Erwan Flécher
- Department of Thoracic and Cardiac Surgery, CHU Rennes, Rennes, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Luc Sensebé
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | | | - Nicolas Espagnolle
- STROMALab, Etablissement Français du Sang-Occitanie (EFS), Inserm 1031, University of Toulouse, National Veterinary School of Toulouse (ENVT), ERL5311 CNRS, Toulouse, France
| | - Karin Tarte
- UMR 1236, University of Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.,SITI Laboratory, Etablissement Français du Sang Bretagne, CHU Rennes, Rennes, France
| |
Collapse
|
19
|
Brennen WN, Isaacs JT. Mesenchymal stem cells and the embryonic reawakening theory of BPH. Nat Rev Urol 2019; 15:703-715. [PMID: 30214054 DOI: 10.1038/s41585-018-0087-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The prostate is the only organ in a man that continues to grow with age. John McNeal proposed, 40 years ago, that this BPH is characterized by an age-related reinitiation of benign neoplastic growth selectively in developmentally abortive distal ducts within the prostate transition-periurethral zone (TPZ), owing to a reawakening of inductive stroma selectively within these zones. An innovative variant of this hypothesis is that, owing to its location, the TPZ is continuously exposed to urinary components and/or autoantigens, which produces an inflammatory TPZ microenvironment that promotes recruitment of bone marrow-derived mesenchymal stem cells (MSCs) and generates a paracrine-inductive stroma that reinitiates benign neoplastic nodular growth. In support of this hypothesis, MSCs infiltrate human BPH tissue and have the ability to stimulate epithelial stem cell growth. These results provide a framework for defining both the aetiology of BPH in ageing men and insights into new therapeutic approaches.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA. .,Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Seo Y, Shin TH, Kim HS. Current Strategies to Enhance Adipose Stem Cell Function: An Update. Int J Mol Sci 2019; 20:E3827. [PMID: 31387282 PMCID: PMC6696067 DOI: 10.3390/ijms20153827] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) emerged as a promising therapeutic tool targeting a variety of inflammatory disorders due to their multiple remarkable properties, such as superior immunomodulatory function and tissue-regenerative capacity. Although bone marrow (BM) is a dominant source for adult MSCs, increasing evidence suggests that adipose tissue-derived stem cells (ASCs), which can be easily obtained at a relatively high yield, have potent therapeutic advantages comparable with BM-MSCs. Despite its outstanding benefits in pre-clinical settings, the practical efficacy of ASCs remains controversial since clinical trials with ASC application often resulted in unsatisfactory outcomes. To overcome this challenge, scientists established several strategies to generate highly functional ASCs beyond the naïve cells, including (1) pre-conditioning of ASCs with various stimulants such as inflammatory agents, (2) genetic manipulation of ASCs and (3) modification of culture conditions with three-dimensional (3D) aggregate formation and hypoxic culture. Also, exosomes and other extracellular vesicles secreted from ASCs can be applied directly to recapitulate the beneficial performance of ASCs. This review summarizes the current strategies to improve the therapeutic features of ASCs for successful clinical implementation.
Collapse
Affiliation(s)
- Yoojin Seo
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Tae-Hoon Shin
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hyung-Sik Kim
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea.
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
21
|
Zhai W, Yong D, El-Jawhari JJ, Cuthbert R, McGonagle D, Win Naing M, Jones E. Identification of senescent cells in multipotent mesenchymal stromal cell cultures: Current methods and future directions. Cytotherapy 2019; 21:803-819. [PMID: 31138507 DOI: 10.1016/j.jcyt.2019.05.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/30/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
Regardless of their tissue of origin, multipotent mesenchymal stromal cells (MSCs) are commonly expanded in vitro for several population doublings to achieve a sufficient number of cells for therapy. Prolonged MSC expansion has been shown to result in phenotypical, morphological and gene expression changes in MSCs, which ultimately lead to the state of senescence. The presence of senescent cells in therapeutic MSC batches is undesirable because it reduces their viability, differentiation potential and trophic capabilities. Additionally, senescent cells acquire senescence-activated secretory phenotype, which may not only induce apoptosis in the neighboring host cells following MSC transplantation, but also trigger local inflammatory reactions. This review outlines the current and promising new methodologies for the identification of senescent cells in MSC cultures, with a particular emphasis on non-destructive and label-free methodologies. Technologies allowing identification of individual senescent cells, based on new surface markers, offer potential advantage for targeted senescent cell removal using new-generation senolytic agents, and subsequent production of therapeutic MSC batches fully devoid of senescent cells. Methods or a combination of methods that are non-destructive and label-free, for example, involving cell size and spectroscopic measurements, could be the best way forward because they do not modify the cells of interest, thus maximizing the final output of therapeutic-grade MSC cultures. The further incorporation of machine learning methods has also recently shown promise in facilitating, automating and enhancing the analysis of these measured data.
Collapse
Affiliation(s)
- Weichao Zhai
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK; Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Derrick Yong
- Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Jehan Jomaa El-Jawhari
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK; Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Richard Cuthbert
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK
| | - May Win Naing
- Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Elena Jones
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK.
| |
Collapse
|
22
|
Abstract
Adipogenesis is a complex process whereby the multipotent adipose-derived stem cell is converted to a preadipocyte before terminal differentiation into the mature adipocyte. Preadipocytes are present throughout adult life, exhibit adipose fat depot specificity, and differentiate and proliferate from distinct progenitor cells. The mechanisms that promote preadipocyte commitment and maturation involve numerous protein factor regulators, epigenetic factors, and miRNAs. Detailed characterization of this process is currently an area of intense research and understanding the roles of preadipocytes in tissue plasticity may provide insight into novel approaches for tissue engineering, regenerative medicine and treating a host of obesity-related conditions. In the current study, we analyzed the current literature and present a review of the characteristics of transitioning adipocytes and detail how local microenvironments influence their progression towards terminal differentiation and maturation. Specifically, we detail the characterization of preadipocyte via surface markers, examine the signaling cascades and regulation behind adipogenesis and cell maturation, and survey their role in tissue plasticity and health and disease.
Collapse
|
23
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|
24
|
Abstract
With the advancements in antenatal steroid therapies and surfactant replacement, current clinical practices in neonatal intensive care units allow the survival of infants at very low gestational age. Despite these advances, there continues to be significant morbidity associated with extreme preterm birth that includes both short-term and long-term cardiorespiratory impairment. With no effective single therapy in preventing or treating developmental lung injuries, the need for new tools to treat and reduce risk of complications associated with extreme preterm birth is urgent. Stem cell-based therapies, in particular therapies utilizing mesenchymal stem (stromal) cells (MSCs), have shown promise in a number of animal models of lung pathologies relevant to neonatology. Recent studies in this field have consolidated the concept that the therapeutic mechanism of MSC action is paracrine, and this led to wide acceptance of the concept that the delivery of the MSC secretome rather than live cells may provide an alternative therapeutic approach for many complex diseases. Here, we summarize the significance and application of cell-free based therapies in preclinical models of neonatal lung injury. We emphasize the development of extracellular vesicle (EV)-based therapeutics and focus on the challenges that remain to be addressed before their application to clinical practice.
Collapse
|
25
|
Negri T, Brich S, Bozzi F, Volpi CV, Gualeni AV, Stacchiotti S, De Cecco L, Canevari S, Gloghini A, Pilotti S. New transcriptional-based insights into the pathogenesis of desmoplastic small round cell tumors (DSRCTs). Oncotarget 2018; 8:32492-32504. [PMID: 28415643 PMCID: PMC5464804 DOI: 10.18632/oncotarget.16477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022] Open
Abstract
To gain new insights into desmoplastic small round cell tumors (DSRCTs) by means of gene expression profiling (GEP). Formalin-fixed, paraffin-embedded surgical specimens obtained from seven pretreated DSRCT patients were interrogated using GEP complemented by immunohistochemistry, a cancer stem cell array, and miRNA in situ hybridisation, including the combined chimera modules miRNA-200/ZEB1 and miRNA-34/SLUG. The chimera modules divided the cases into three classes that respectively recapitulated the traits of mesenchymal epithelial reverse transition (MErT), epithelial mesenchymal transition (EMT), and hybrid/partial EMT. This indicates a close correlation between the reprogramming governed by EMT regulators and DSRCT biology, which was further confirmed by miRNA-21 and is consistent with the broad morphological spectrum of DSRCTs. Starting from the miRNA-200/ZEB1 axis, we also found that DSRCTs carry a signature of immunological ignorance that is not responsive to PD-L1 blockade. Evidence that the up-regulation of miRNA-200 and E-cadherin, and quite a high level of miRNA-21 expression segregate with the MErT supports the idea that, in addition to the hybrid/partial state, MErT is also enriched in stemness: the androgen-positive cases, whose stemness traits were confirmed by stem cell arrays, all fell into these two classes. Our findings also confirmed that tumoral cell PDGFRA expression correlates with desmoplasia, and demonstrated the co-expression of PDGFRA and ISLR/Meflin, another marker of pluripotency. Despite the limited number of cases, these findings provide unexpectedly relevant information concerning the pathogenesis of DSRCTs, and prove the validity of miRNA-based chimera circuit modelling in the clinico-pathological setting.
Collapse
Affiliation(s)
- Tiziana Negri
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Brich
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,MOSE-DEA, University of Trieste, Trieste, Italy
| | - Fabio Bozzi
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara V Volpi
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ambra V Gualeni
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor and Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics and Bioinformatics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Canevari
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics and Bioinformatics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
26
|
Dolbashid AS, Mokhtar MS, Muhamad F, Ibrahim F. Potential applications of human artificial skin and electronic skin (e-skin): a review. BIOINSPIRED BIOMIMETIC AND NANOBIOMATERIALS 2018. [DOI: 10.1680/jbibn.17.00002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Asdani Saifullah Dolbashid
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia; Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Mas Sahidayana Mokhtar
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia; Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Farina Muhamad
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Fatimah Ibrahim
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia; Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Corrêa NCR, Kuligovski C, Paschoal ACC, Abud APR, Rebelatto CLK, Leite LMB, Senegaglia AC, Dallagiovanna B, Aguiar AMD. Human adipose-derived stem cells (ADSC) and human periodontal ligament stem cells (PDLSC) as cellular substrates of a toxicity prediction assay. Regul Toxicol Pharmacol 2018; 92:75-82. [DOI: 10.1016/j.yrtph.2017.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023]
|
28
|
Fahy N, Alini M, Stoddart MJ. Mechanical stimulation of mesenchymal stem cells: Implications for cartilage tissue engineering. J Orthop Res 2018; 36:52-63. [PMID: 28763118 DOI: 10.1002/jor.23670] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/24/2017] [Indexed: 02/04/2023]
Abstract
Articular cartilage is a load-bearing tissue playing a crucial mechanical role in diarthrodial joints, facilitating joint articulation, and minimizing wear. The significance of biomechanical stimuli in the development of cartilage and maintenance of chondrocyte phenotype in adult tissues has been well documented. Furthermore, dysregulated loading is associated with cartilage pathology highlighting the importance of mechanical cues in cartilage homeostasis. The repair of damaged articular cartilage resulting from trauma or degenerative joint disease poses a major challenge due to a low intrinsic capacity of cartilage for self-renewal, attributable to its avascular nature. Bone marrow-derived mesenchymal stem cells (MSCs) are considered a promising cell type for cartilage replacement strategies due to their chondrogenic differentiation potential. Chondrogenesis of MSCs is influenced not only by biological factors but also by the environment itself, and various efforts to date have focused on harnessing biomechanics to enhance chondrogenic differentiation of MSCs. Furthermore, recapitulating mechanical cues associated with cartilage development and homeostasis in vivo, may facilitate the development of a cellular phenotype resembling native articular cartilage. The goal of this review is to summarize current literature examining the effect of mechanical cues on cartilage homeostasis, disease, and MSC chondrogenesis. The role of biological factors produced by MSCs in response to mechanical loading will also be examined. An in-depth understanding of the impact of mechanical stimulation on the chondrogenic differentiation of MSCs in terms of endogenous bioactive factor production and signaling pathways involved, may identify therapeutic targets and facilitate the development of more robust strategies for cartilage replacement using MSCs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:52-63, 2018.
Collapse
Affiliation(s)
- Niamh Fahy
- AO Research Institute Davos, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | |
Collapse
|
29
|
Medeiros Tavares Marques JC, Cornélio DA, Nogueira Silbiger V, Ducati Luchessi A, de Souza S, Batistuzzo de Medeiros SR. Identification of new genes associated to senescent and tumorigenic phenotypes in mesenchymal stem cells. Sci Rep 2017; 7:17837. [PMID: 29259202 PMCID: PMC5736717 DOI: 10.1038/s41598-017-16224-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023] Open
Abstract
Although human mesenchymal stem cells (hMSCs) are a powerful tool for cell therapy, prolonged culture times result in replicative senescence or acquisition of tumorigenic features. To identify a molecular signature for senescence, we compared the transcriptome of senescent and young hMSCs with normal karyotype (hMSCs/n) and with a constitutional inversion of chromosome 3 (hMSC/inv). Senescent and young cells from both lineages showed differentially expressed genes (DEGs), with higher levels in senescent hMSCs/inv. Among the 30 DEGs in senescent hMSC/inv, 11 are new candidates for biomarkers of cellular senescence. The functional categories most represented in senescent hMSCs were related to cellular development, cell growth/proliferation, cell death, cell signaling/interaction, and cell movement. Mapping of DEGs onto biological networks revealed matrix metalloproteinase-1, thrombospondin 1, and epidermal growth factor acting as topological bottlenecks. In the comparison between senescent hMSCs/n and senescent hMSCs/inv, other functional annotations such as segregation of chromosomes, mitotic spindle formation, and mitosis and proliferation of tumor lines were most represented. We found that many genes categorized into functional annotations related to tumors in both comparisons, with relation to tumors being highest in senescent hMSCs/inv. The data presented here improves our understanding of the molecular mechanisms underlying the onset of cellular senescence as well as tumorigenesis.
Collapse
Affiliation(s)
- Joana Cristina Medeiros Tavares Marques
- Faculdade de Ciências da Saúde do Trairi (FACISA), Universidade Federal do Rio Grande do Norte (UFRN), Rua Traíri, S/N, Centro, Santa Cruz, Rio Grande do Norte (RN), 59200-000, Brazil
| | - Déborah Afonso Cornélio
- Laboratório de Biologia Molecular e Genômica, Centro de Biociências, UFRN, Campus Universitário, Avenida Senador Salgado Filho, 3000, Lagoa nova, Natal, RN, 59078-900, Brazil
| | - Vivian Nogueira Silbiger
- Departamento de Análises Clínicas e Toxicológicas, Centro de Ciências da Saúde, CCS/UFRN, Av General Cordeiro de Farias S/N, Petropolis, Natal, 59010-115, RN, Brazil
| | - André Ducati Luchessi
- Departamento de Análises Clínicas e Toxicológicas, Centro de Ciências da Saúde, CCS/UFRN, Av General Cordeiro de Farias S/N, Petropolis, Natal, 59010-115, RN, Brazil
| | - Sandro de Souza
- Instituto do Cérebro, Instituto de Metrópole Digital, UFRN, Av. Nascimento de Castro, 2155, UFRN, 59056-450, RN, Brazil
| | - Silvia Regina Batistuzzo de Medeiros
- Laboratório de Biologia Molecular e Genômica, Centro de Biociências, UFRN, Campus Universitário, Avenida Senador Salgado Filho, 3000, Lagoa nova, Natal, RN, 59078-900, Brazil.
| |
Collapse
|
30
|
RNA sequencing reveals a transcriptomic portrait of human mesenchymal stem cells from bone marrow, adipose tissue, and palatine tonsils. Sci Rep 2017; 7:17114. [PMID: 29214990 PMCID: PMC5719355 DOI: 10.1038/s41598-017-16788-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/17/2017] [Indexed: 12/28/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) are adult multipotent cells that have plasticity and inhabit the stroma of diverse tissues. The potential utility of MSCs has been heavily investigated in the fields of regenerative medicine and cell therapy. However, MSCs represent diverse populations that may depend on the tissue of origin. Thus, the ability to identify specific MSC populations has remained difficult. Using RNA sequencing, we analyzed the whole transcriptomes of bone marrow-derived MSCs (BMs), adipose tissue-derived MSCs (AMs), and tonsil-derived MSCs (TMs). We categorized highly regulated genes from these MSC groups according to functional gene ontology (GO) classification. AMs and TMs showed higher expression of genes encoding proteins that function in protein binding, growth factor, or cytokine activity in extracellular compartments than BMs. Interestingly, TM were highly enriched for genes coding extracellular, protein-binding proteins compared with AMs. Functional Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis also showed differentially enriched signaling pathways between the three MSC groups. Further, we confirmed surface antigens expressed in common and in a tissue-specific manner on BMs, AMs, and TMs by flow cytometry analysis. This study provides comprehensive characteristics of MSCs derived from different tissues to better understand their cellular and molecular biology.
Collapse
|
31
|
Kasoju N, Wang H, Zhang B, George J, Gao S, Triffitt JT, Cui Z, Ye H. Transcriptomics of human multipotent mesenchymal stromal cells: Retrospective analysis and future prospects. Biotechnol Adv 2017; 35:407-418. [DOI: 10.1016/j.biotechadv.2017.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 12/28/2022]
|
32
|
Togarrati PP, Sasaki RT, Abdel-Mohsen M, Dinglasan N, Deng X, Desai S, Emmerson E, Yee E, Ryan WR, da Silva MCP, Knox SM, Pillai SK, Muench MO. Identification and characterization of a rich population of CD34 + mesenchymal stem/stromal cells in human parotid, sublingual and submandibular glands. Sci Rep 2017; 7:3484. [PMID: 28615711 PMCID: PMC5471181 DOI: 10.1038/s41598-017-03681-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 05/03/2017] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) play crucial roles in maintaining tissue homeostasis during physiological turnovers and injuries. Very little is known about the phenotype, distribution and molecular nature of MSCs in freshly isolated human salivary glands (SGs) as most reports have focused on the analysis of cultured MSCs. Our results demonstrate that the cell adhesion molecule CD34 was widely expressed by the MSCs of human major SGs, namely parotid (PAG), sublingual (SLG) and submandibular (SMG) glands. Further, gene expression analysis of CD34+ cells derived from fetal SMGs showed significant upregulation of genes involved in cellular adhesion, proliferation, branching, extracellular matrix remodeling and organ development. Moreover, CD34+ SMG cells exhibited elevated expression of genes encoding extracellular matrix, basement membrane proteins, and members of ERK, FGF and PDGF signaling pathways, which play key roles in glandular development, branching and homeostasis. In vitro CD34+ cell derived SG-MSCs revealed multilineage differentiation potential. Intraglandular transplantation of cultured MSCs in immunodeficient mice led to their engraftment in the injected and uninjected contralateral and ipsilateral glands. Engrafted cells could be localized to the stroma surrounding acini and ducts. In summary, our data show that CD34+ derived SG-MSCs could be a promising cell source for adoptive cell-based SG therapies, and bioengineering of artificial SGs.
Collapse
Affiliation(s)
| | - Robson T Sasaki
- Department of Morphology and Genetics - Discipline of Descriptive and Topographic Anatomy, Federal University of São Paulo, Brazil, CEP, USA
| | - Mohamed Abdel-Mohsen
- Blood Systems Research Institute, San Francisco, CA, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA.,The Wistar Institute, Philadelphia, PA, USA
| | | | - Xutao Deng
- Blood Systems Research Institute, San Francisco, CA, USA
| | - Shivani Desai
- Blood Systems Research Institute, San Francisco, CA, USA
| | - Elaine Emmerson
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Elizabeth Yee
- Blood Systems Research Institute, San Francisco, CA, USA
| | - William R Ryan
- Division of Head and Neck Oncologic/Endocrine/Salivary Surgery, Department of Otolaryngology, University of California San Francisco, San Francisco, CA, USA
| | - Marcelo C P da Silva
- Department of Morphology and Genetics - Discipline of Descriptive and Topographic Anatomy, Federal University of São Paulo, Brazil, CEP, USA
| | - Sarah M Knox
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Satish K Pillai
- Blood Systems Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Marcus O Muench
- Blood Systems Research Institute, San Francisco, CA, USA. .,Department of Laboratory Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
33
|
Clark KC, Fierro FA, Ko EM, Walker NJ, Arzi B, Tepper CG, Dahlenburg H, Cicchetto A, Kol A, Marsh L, Murphy WJ, Fazel N, Borjesson DL. Human and feline adipose-derived mesenchymal stem cells have comparable phenotype, immunomodulatory functions, and transcriptome. Stem Cell Res Ther 2017; 8:69. [PMID: 28320483 PMCID: PMC5360077 DOI: 10.1186/s13287-017-0528-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/10/2017] [Accepted: 03/03/2017] [Indexed: 12/18/2022] Open
Abstract
Background Adipose-derived mesenchymal stem cells (ASCs) are a promising cell therapy to treat inflammatory and immune-mediated diseases. Development of appropriate pre-clinical animal models is critical to determine safety and attain early efficacy data for the most promising therapeutic candidates. Naturally occurring diseases in cats already serve as valuable models to inform human clinical trials in oncologic, cardiovascular, and genetic diseases. The objective of this study was to complete a comprehensive side-by-side comparison of human and feline ASCs, with an emphasis on their immunomodulatory capacity and transcriptome. Methods Human and feline ASCs were evaluated for phenotype, immunomodulatory profile, and transcriptome. Additionally, transwells were used to determine the role of cell-cell contact in ASC-mediated inhibition of lymphocyte proliferation in both humans and cats. Results Similar to human ASCs, feline ASCs were highly proliferative at low passages and fit the minimal criteria of multipotent stem cells including a compatible surface protein phenotype, osteogenic capacity, and normal karyotype. Like ASCs from all species, feline ASCs inhibited mitogen-activated lymphocyte proliferation in vitro, with or without direct ASC-lymphocyte contact. Feline ASCs mimic human ASCs in their mediator secretion pattern, including prostaglandin E2, indoleamine 2,3 dioxygenase, transforming growth factor beta, and interleukin-6, all augmented by interferon gamma secretion by lymphocytes. The transcriptome of three unactivated feline ASC lines were highly similar. Functional analysis of the most highly expressed genes highlighted processes including: 1) the regulation of apoptosis; 2) cell adhesion; 3) response to oxidative stress; and 4) regulation of cell differentiation. Finally, feline ASCs had a similar gene expression profile to noninduced human ASCs. Conclusions Findings suggest that feline ASCs modulate lymphocyte proliferation using soluble mediators that mirror the human ASC secretion pattern. Uninduced feline ASCs have similar gene expression profiles to uninduced human ASCs, as revealed by transcriptome analysis. These data will help inform clinical trials using cats with naturally occurring diseases as surrogate models for human clinical trials in the regenerative medicine arena. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0528-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaitlin C Clark
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, 95816, USA
| | - Fernando A Fierro
- Institute for Regenerative Cures and Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95816, USA
| | - Emily Mills Ko
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, 95816, USA
| | - Naomi J Walker
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, 95816, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, 95816, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, 95816, USA
| | - Heather Dahlenburg
- Institute for Regenerative Cures and Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95816, USA
| | - Andrew Cicchetto
- Institute for Regenerative Cures and Department of Cell Biology and Human Anatomy, University of California, Davis, CA, 95816, USA
| | - Amir Kol
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, 95816, USA
| | - Lyndsey Marsh
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, 95816, USA
| | - William J Murphy
- Department of Dermatology, School of Medicine, University of California, Davis, CA, 95816, USA
| | - Nasim Fazel
- Department of Dermatology, School of Medicine, University of California, Davis, CA, 95816, USA
| | - Dori L Borjesson
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, 95816, USA.
| |
Collapse
|
34
|
Rubessa M, Polkoff K, Bionaz M, Monaco E, Milner DJ, Holllister SJ, Goldwasser MS, Wheeler MB. Use of Pig as a Model for Mesenchymal Stem Cell Therapies for Bone Regeneration. Anim Biotechnol 2017; 28:275-287. [PMID: 28267421 DOI: 10.1080/10495398.2017.1279169] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bone is a plastic tissue with a large healing capability. However, extensive bone loss due to disease or trauma requires extreme therapy such as bone grafting or tissue-engineering applications. Presently, bone grafting is the gold standard for bone repair, but presents serious limitations including donor site morbidity, rejection, and limited tissue regeneration. The use of stem cells appears to be a means to overcome such limitations. Bone marrow mesenchymal stem cells (BMSC) have been the choice thus far for stem cell therapy for bone regeneration. However, adipose-derived stem cells (ASC) have similar immunophenotype, morphology, multilineage potential, and transcriptome compared to BMSC, and both types have demonstrated extensive osteogenic capacity both in vitro and in vivo in several species. The use of scaffolds in combination with stem cells and growth factors provides a valuable tool for guided bone regeneration, especially for complex anatomic defects. Before translation to human medicine, regenerative strategies must be developed in animal models to improve effectiveness and efficiency. The pig presents as a useful model due to similar macro- and microanatomy and favorable logistics of use. This review examines data that provides strong support for the clinical translation of the pig model for bone regeneration.
Collapse
Key Words
- ASC, adipose-derived stem cells
- BMP, bone morphogenetic protein
- BMSC, bone marrow mesenchymal stem cells
- Bone
- DEG, differentially expressed genes
- FDR, false-discovery rate
- HA, hydroxyapatite
- HA/TCP, hydroxyapatite/tricalcium phosphate
- MRI, magnetic resonance imaging
- MSC, mesenchymal stem cells
- ONFH, osteonecrosis of the femoral head
- PCL, Poly (ϵ-caprolactone)
- PEG, polyethylene glycol
- PLGA, polylactic-coglycolic acid
- TCP, beta tri-calcium phosphate
- USSC, unrestricted somatic stem cell
- scaffolds
- stem cells
- swine
- tissue engineering
Collapse
Affiliation(s)
- Marcello Rubessa
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Kathryn Polkoff
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | | | - Elisa Monaco
- b Oregon State University , Corvallis , Oregon , USA
| | - Derek J Milner
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | | | - Michael S Goldwasser
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA.,d New Hanover Regional Medical Center , Wilmington , North Carolina , USA
| | - Matthew B Wheeler
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| |
Collapse
|
35
|
Hu Y, Xiong LL, Zhang P, Wang TH. Microarray expression profiles of genes in lung tissues of rats subjected to focal cerebral ischemia-induced lung injury following bone marrow-derived mesenchymal stem cell transplantation. Int J Mol Med 2016; 39:57-70. [PMID: 27922691 PMCID: PMC5179184 DOI: 10.3892/ijmm.2016.2819] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 11/21/2016] [Indexed: 02/05/2023] Open
Abstract
Ischemia-induced stroke is the most common disease of the nervous system and is associated with a high mortality rate worldwide. Cerebral ischemia may lead to remote organ dysfunction, particular in the lungs, resulting in lung injury. Nowadays, bone marrow-derived mesenchymal stem cells (BMSCs) are widely studied in clinical trials as they may provide an effective solution to the treatment of neurological and cardiac diseases; however, the underlying molecular mechanisms remain unknown. In this study, a model of permanent focal cerebral ischemia-induced lung injury was successfully established and confirmed by neurological evaluation and lung injury scores. We demonstrated that the transplantation of BMSCs (passage 3) via the tail vein into the lung tissues attenuated lung injury. In order to elucidate the underlying molecular mechanisms, we analyzed the gene expression profiles in lung tissues from the rats with focal cerebral ischemia and transplanted with BMSCs using a Gene microarray. Moreover, the Gene Ontology database was employed to determine gene function. We found that the phosphoinositide 3-kinase (PI3K)-AKT signaling pathway, transforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF) were downregulated in the BMSC transplantation groups, compared with the control group. These results suggested that BMSC transplantation may attenuate lung injury following focal cerebral ischemia and that this effect is associated with the downregulation of TGF-β, PDGF and the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Liu-Lin Xiong
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Piao Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Ting-Hua Wang
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
36
|
de Souza LEB, Malta TM, Kashima Haddad S, Covas DT. Mesenchymal Stem Cells and Pericytes: To What Extent Are They Related? Stem Cells Dev 2016; 25:1843-1852. [PMID: 27702398 DOI: 10.1089/scd.2016.0109] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) were initially identified as progenitors of skeletal tissues within mammalian bone marrow and cells with similar properties were also obtained from other tissues such as adipose and dental pulp. Although MSCs have been extensively investigated, their native behavior and in vivo identity remain poorly defined. Uncovering the in vivo identity of MSCs has been challenging due to the lack of exclusive cell markers, cellular alterations caused by culture methods, and extensive focus on in vitro properties for characterization. Although MSC site of origin influences their functional properties, these mesenchymal progenitors can be found in the perivascular space in virtually all organs from where they were obtained. However, the precise identity of MSCs within the vascular wall is highly controversial. The recurrent concept that MSCs correspond to pericytes in vivo has been supported mainly by their perivascular localization and expression of some molecular markers. However, this view has been a subject of controversy, in part, due to the application of loose criteria to define pericytes and due to the lack of a marker able to unequivocally identify these cells. Furthermore, recent evidences indicate that subpopulations of MSCs can be found at extravascular sites such as the endosteum. In this opinion review, we bring together the advances and pitfalls on the search for the in vivo identity of MSCs and highlight the recent evidences that suggest that perivascular MSCs are adventitial cells, acting as precursors of pericytes and other stromal cells during tissue homeostasis.
Collapse
Affiliation(s)
- Lucas Eduardo Botelho de Souza
- 1 Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, Brazil .,2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil
| | - Tathiane Maistro Malta
- 2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil .,3 Department of Genetics, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, Brazil
| | - Simone Kashima Haddad
- 2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- 1 Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, Brazil .,2 National Institute of Science and Technology for Stem Cell and Cell Therapy , Ribeirão Preto, Brazil
| |
Collapse
|
37
|
Murgia A, Veronesi E, Candini O, Caselli A, D’souza N, Rasini V, Giorgini A, Catani F, Iughetti L, Dominici M, Burns JS. Potency Biomarker Signature Genes from Multiparametric Osteogenesis Assays: Will cGMP Human Bone Marrow Mesenchymal Stromal Cells Make Bone? PLoS One 2016; 11:e0163629. [PMID: 27711115 PMCID: PMC5053614 DOI: 10.1371/journal.pone.0163629] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
In skeletal regeneration approaches using human bone marrow derived mesenchymal stromal cells (hBM-MSC), functional evaluation before implantation has traditionally used biomarkers identified using fetal bovine serum-based osteogenic induction media and time courses of at least two weeks. However, emerging pre-clinical evidence indicates donor-dependent discrepancies between these ex vivo measurements and the ability to form bone, calling for improved tests. Therefore, we adopted a multiparametric approach aiming to generate an osteogenic potency assay with improved correlation. hBM-MSC populations from six donors, each expanded under clinical-grade (cGMP) conditions, showed heterogeneity for ex vivo growth response, mineralization and bone-forming ability in a murine xenograft assay. A subset of literature-based biomarker genes was reproducibly upregulated to a significant extent across all populations as cells responded to two different osteogenic induction media. These 12 biomarkers were also measurable in a one-week assay, befitting clinical cell expansion time frames and cGMP growth conditions. They were selected for further challenge using a combinatorial approach aimed at determining ex vivo and in vivo consistency. We identified five globally relevant osteogenic signature genes, notably TGF-ß1 pathway interactors; ALPL, COL1A2, DCN, ELN and RUNX2. Used in agglomerative cluster analysis, they correctly grouped the bone-forming cell populations as distinct. Although donor #6 cells were correlation slope outliers, they contrastingly formed bone without showing ex vivo mineralization. Mathematical expression level normalization of the most discrepantly upregulated signature gene COL1A2, sufficed to cluster donor #6 with the bone-forming classification. Moreover, attenuating factors causing genuine COL1A2 gene down-regulation, restored ex vivo mineralization. This suggested that the signature gene had an osteogenically influential role; nonetheless no single biomarker was fully deterministic whereas all five signature genes together led to accurate cluster analysis. We show proof of principle for an osteogenic potency assay providing early characterization of primary cGMP-hBM-MSC cultures according to their donor-specific bone-forming potential.
Collapse
Affiliation(s)
- Alba Murgia
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Elena Veronesi
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
| | - Olivia Candini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Anna Caselli
- CVBF - Consorzio per le Valutazioni Biologiche e Farmacologiche, Ospedale Pediatrico Giovanni XXIII, Bari, Italia
| | - Naomi D’souza
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Valeria Rasini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Andrea Giorgini
- Department of Orthopedic Surgery, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Fabio Catani
- Department of Orthopedic Surgery, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Lorenzo Iughetti
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
- * E-mail: (MD); (JSB)
| | - Jorge S. Burns
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italia
- TPM, Science & Technology Park for Medicine, Mirandola, Modena, Italia
- * E-mail: (MD); (JSB)
| |
Collapse
|
38
|
Arai Y, Park S, Choi B, Ko KW, Choi WC, Lee JM, Han DW, Park HK, Han I, Lee JH, Lee SH. Enhancement of Matrix Metalloproteinase-2 (MMP-2) as a Potential Chondrogenic Marker during Chondrogenic Differentiation of Human Adipose-Derived Stem Cells. Int J Mol Sci 2016; 17:ijms17060963. [PMID: 27322256 PMCID: PMC4926495 DOI: 10.3390/ijms17060963] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 01/09/2023] Open
Abstract
Human adipose-derived stem cells (hASCs) have a capacity to undergo adipogenic, chondrogenic, and osteogenic differentiation. Recently, hASCs were applied to various fields including cell therapy for tissue regeneration. However, it is hard to predict the direction of differentiation of hASCs in real-time. Matrix metalloproteinases (MMPs) are one family of proteolytic enzymes that plays a pivotal role in regulating the biology of stem cells. MMPs secreted by hASCs are expected to show different expression patterns depending on the differentiation state of hASCs because biological functions exhibit different patterns during the differentiation of stem cells. Here, we investigated proteolytic enzyme activity, especially MMP-2 activity, in hASCs during their differentiation. The activities of proteolytic enzymes and MMP-2 were higher during chondrogenic differentiation than during adipogenic and osteogenic differentiation. During chondrogenic differentiation, mRNA expression of MMP-2 and the level of the active form of MMP-2 were increased, which also correlated with Col II. It is concluded that proteolytic enzyme activity and the level of the active form of MMP-2 were increased during chondrogenic differentiation, which was accelerated in the presence of Col II protein. According to our findings, MMP-2 could be a candidate maker for real-time detection of chondrogenic differentiation of hASCs.
Collapse
Affiliation(s)
- Yoshie Arai
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Sunghyun Park
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Bogyu Choi
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Kyoung-Won Ko
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Won Chul Choi
- Department of Orthopedic Surgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Joong-Myung Lee
- Department of Orthopedic Surgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Dong-Wook Han
- Department of Optics and Mechatronics Engineering, BK21+ Nano-Integrated Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan 619-961, Korea.
| | - Hun-Kuk Park
- Department of Biomedical Engineering, Collage of Medicine, Kyung Hee University, Seoul 151-742, Korea.
| | - Inbo Han
- Department of Neurosurgery, Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| | - Jong Hun Lee
- Department of Food Science and Biotechnology, College of Life Science, CHA University, Gyeonggi-do 443-742, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do 443-742, Korea.
| |
Collapse
|
39
|
Montemurro T, Viganò M, Ragni E, Barilani M, Parazzi V, Boldrin V, Lavazza C, Montelatici E, Banfi F, Lauri E, Giovanelli S, Baccarin M, Guerneri S, Giordano R, Lazzari L. Angiogenic and anti-inflammatory properties of mesenchymal stem cells from cord blood: soluble factors and extracellular vesicles for cell regeneration. Eur J Cell Biol 2016; 95:228-38. [PMID: 27139721 DOI: 10.1016/j.ejcb.2016.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 02/25/2016] [Accepted: 04/06/2016] [Indexed: 12/18/2022] Open
Abstract
In a recent work, our group showed the existence of two distinct mesenchymal stem cell (MSC) subsets within human umbilical cord blood. One less proliferative and short-living (SL-CBMSC), the other with higher growth rate and long-living (LL-CBMSC), and therefore better suited for regenerative medicine applications. We examined whether LL-CBMSC possess peculiar paracrine properties able to affect angiogenesis or inflammatory processes. It was shown for the first time that pro-angiogenic, proliferation-stimulating and tissue repairing factors were released at high level not only as soluble cytokines, but also as mRNA precursors embedded in membrane vesicles. The combination of this primary (proteic factors interacting with surface receptors) and delayed (mRNA transferred and translated via vesicle fusion and cargo release) interaction in endothelial target cells resulted in strong blood vessel induction with the development of capillary-like structures. In addition, LL-CBMSC dynamically modulated their release of pro-angiogenic and anti-inflammatory factors in an in vitro model of damage. In conclusion, LL-CBMSC synthesize and secrete multiple factors that may be attuned in response to the status of the target cell, a crucial requisite when paracrine mechanisms are needed at onset of tissue regeneration.
Collapse
Affiliation(s)
- Tiziana Montemurro
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Mariele Viganò
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Enrico Ragni
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Mario Barilani
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valentina Parazzi
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Valentina Boldrin
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Cristiana Lavazza
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Elisa Montelatici
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Federica Banfi
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Eleonora Lauri
- Anatomia Patologica, Ospedale Sacco, Università degli Studi di Milano, Milano, Italy
| | - Silvia Giovanelli
- Milano Cord Blood Bank, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Marco Baccarin
- Laboratorio Citogenetica e Genetica molecolare, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Silvana Guerneri
- Laboratorio Citogenetica e Genetica molecolare, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Rosaria Giordano
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Lorenza Lazzari
- Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
| |
Collapse
|
40
|
Mitsialis SA, Kourembanas S. Stem cell-based therapies for the newborn lung and brain: Possibilities and challenges. Semin Perinatol 2016; 40:138-51. [PMID: 26778234 PMCID: PMC4808378 DOI: 10.1053/j.semperi.2015.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There have been substantial advances in neonatal medical care over the past 2 decades that have resulted in the increased survival of very low birth weight infants, survival that in some centers extends to 22 weeks gestational age. Despite these advances, there continues to be significant morbidity associated with extreme preterm birth that includes both short-term and long-term pulmonary and neurologic consequences. No single therapy has proven to be effective in preventing or treating either developmental lung and brain injuries in preterm infants or the hypoxic-ischemic injury that can be inflicted on the full-term brain as a result of in utero or perinatal complications. Stem cell-based therapies are emerging as a potential paradigm-shifting approach for such complex diseases with multifactorial etiologies, but a great deal of work is still required to understand the role of stem/progenitor cells in normal development and in the repair of injured tissue. This review will summarize the biology of the various stem/progenitor cells, their effects on tissue repair in experimental models of lung and brain injury, the recent advances in our understanding of their mechanism of action, and the challenges that remain to be addressed before their eventual application to clinical care.
Collapse
|
41
|
Maeda K, Enomoto A, Hara A, Asai N, Kobayashi T, Horinouchi A, Maruyama S, Ishikawa Y, Nishiyama T, Kiyoi H, Kato T, Ando K, Weng L, Mii S, Asai M, Mizutani Y, Watanabe O, Hirooka Y, Goto H, Takahashi M. Identification of Meflin as a Potential Marker for Mesenchymal Stromal Cells. Sci Rep 2016; 6:22288. [PMID: 26924503 PMCID: PMC4770287 DOI: 10.1038/srep22288] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 02/11/2016] [Indexed: 01/14/2023] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) in culture are derived from BM stromal cells or skeletal stem cells. Whereas MSCs have been exploited in clinical medicine, the identification of MSC-specific markers has been limited. Here, we report that a cell surface and secreted protein, Meflin, is expressed in cultured MSCs, fibroblasts and pericytes, but not other types of cells including epithelial, endothelial and smooth muscle cells. In vivo, Meflin is expressed by immature osteoblasts and chondroblasts. In addition, Meflin is found on stromal cells distributed throughout the BM, and on pericytes and perivascular cells in multiple organs. Meflin maintains the undifferentiated state of cultured MSCs and is downregulated upon their differentiation, consistent with the observation that Meflin-deficient mice exhibit increased number of osteoblasts and accelerated bone development. In the bone and BM, Meflin is more highly expressed in primitive stromal cells that express platelet-derived growth factor receptor α and Sca-1 than the Sca-1-negative adipo-osteogenic progenitors, which create a niche for hematopoiesis. Those results are consistent with a decrease in the number of clonogenic colony-forming unit-fibroblasts within the BM of Meflin-deficient mice. These preliminary data suggest that Meflin is a potential marker for cultured MSCs and their source cells in vivo.
Collapse
Affiliation(s)
- Keiko Maeda
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Akitoshi Hara
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Naoya Asai
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takeshi Kobayashi
- Department of Physiology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Asuka Horinouchi
- Department of Nephrology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shoichi Maruyama
- Department of Nephrology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takahiro Nishiyama
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, , 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takuya Kato
- Tumour Cell Biology Laboratory, The Francis-Crick Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, United Kingdom
| | - Kenju Ando
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Liang Weng
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinji Mii
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masato Asai
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.,Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Osamu Watanabe
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidemi Goto
- Department of Gastroenterology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masahide Takahashi
- Department of Pathology, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
42
|
Najar M, Raicevic G, Fayyad-Kazan H, De Bruyn C, Bron D, Toungouz M, Lagneaux L. Bone Marrow Mesenchymal Stromal Cells Induce Proliferative, Cytokinic and Molecular Changes During the T Cell Response: The Importance of the IL-10/CD210 Axis. Stem Cell Rev Rep 2016; 11:442-52. [PMID: 25326368 DOI: 10.1007/s12015-014-9567-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bone marrow mesenchymal stromal cells (BM-MSCs) display immunomodulatory features, representing a promising tool for cell-based therapies. However, the mechanisms used by MSCs to regulate T cell fate remain unclear. AIMS We investigated the potential of BM-MSCs to modulate T cell activation, proliferation, cytokine secretion and immunophenotype. MATERIALS AND METHODS T cells were co-cultured with BM-MSCs to assess their immunomodulatory impact. T cell characterization was performed using cell tracing, ELISA, intracellular and surface staining, flow cytometry analysis and qPCR. RESULTS The activation and proliferation of T cells were downregulated during coculture with BM-MSCs. We also observed that BM-MSCs upregulated IL-10 secretion as well as the expression of its receptor CD210 on T cells, thus creating a loop favoring the expansion of IL-10-producing T cells. IL-10 neutralization restored T cell proliferation, demonstrating that IL-10 is functionally relevant during immunomodulation. Moreover, BM-MSCs differently modulated CD4 and CD8 T-cell immunophenotype by inducing broad changes in their molecular pattern. CONCLUSIONS We provide a comprehensive functional and molecular characterization of T cells that are immunomodulated by BM-MSCs. Indeed, a better understanding of the immunological interplay between T cells and MSCs will facilitate the development of new efficient approaches to improve cell-based immune therapies.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institute Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Bâtiment de Transfusion (Level +1), Route de Lennik n° 808, 1070, Brussels, Belgium,
| | | | | | | | | | | | | |
Collapse
|
43
|
Huang B, Cheng X, Wang H, Huang W, la Ga Hu Z, Wang D, Zhang K, Zhang H, Xue Z, Da Y, Zhang N, Hu Y, Yao Z, Qiao L, Gao F, Zhang R. Mesenchymal stem cells and their secreted molecules predominantly ameliorate fulminant hepatic failure and chronic liver fibrosis in mice respectively. J Transl Med 2016; 14:45. [PMID: 26861623 PMCID: PMC4746907 DOI: 10.1186/s12967-016-0792-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/20/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Orthotopic liver transplantation is the only effective treatment for liver failure but limited with shortage of available donor organs. Recent studies show promising results of mesenchymal stem cells (MSCs)-based therapies. METHODS We systematically investigate the therapeutic effects of MSCs or MSC-conditioned medium (MSC-CM) in ameliorating fulminant hepatic failure (FHF) and chronic liver fibrosis in mice. In addition, extensive flow cytometry analysis of spleens from vehicle and MSC- and MSC-CM-treated mice was applied to reveal the alteration of inflammatory state. RESULTS In FHF model, MSCs treatment reduced remarkably the death incidents; the analysis of gross histopathology showed that control livers were soft and shrunken with extensive extravasated blood, which was gradually reduced at later time points, while MSC-treated livers showed gross pathological changes, even 24 h after MSC infusion, and hematoxylin and eosin staining revealed dramatical hepatocellular death with cytoplasmic vacuolization suppressed by MSCs treatment; flow cytometry analysis of total lymphocytes showed that macrophages (F4/80) infiltrated into control livers more than MSC-treated livers; by contrast, MSC-CM partially ameliorates FHF. In chronic liver injury model, MSC and MSC-CM both suppressed fibrogenesis and necroinflammatory, and the later was better; activation of hepatic stellate cells (α-SMA) was inhibited; glycogen synthesis and storage (indicated by periodic acid-Schiff -staining) was improved; liver regeneration (Ki67) was promoted while liver apoptosis (TUNEL) was reduced. In the in vitro, MSCs promote macrophage line RAW264.7 apoptosis and MSC-CM promotes apoptosis and inhibits proliferation of HSC line LX-2. We also found that MSCs and MSC-CM could improve spleen; MSC-CM increased levels of Th2 and Treg cells, and reduced levels of Th17 cells, whereas levels of Th1 cells were unchanged; comparatively, MSC treatment did not affect Th17 and Treg cells and only slightly alters inflammatory state; MSC and MSC-CM treatment both substantially down-regulated macrophages in the spleens. CONCLUSION Both MSCs and MSC-CM exert therapeutic effects by acting on various key cells during the pathogenesis of FHF and chronic fibrosis, stimulating hepatocyte proliferation and suppressing apoptosis, down-regulating infiltrating macrophages, converting CD4(+) T lymphocyte system into an anti-inflammatory state, and facilitating hepatic stellate cell death.
Collapse
Affiliation(s)
- Biao Huang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Xixi Cheng
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Huafeng Wang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China. .,School of Life Science, Shanxi Normal University, Linfen, Shanxi Province, China.
| | - Wenjing Huang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Zha la Ga Hu
- Department of Cell Biology, Logistic College of CAPF, Tianjin, China.
| | - Dan Wang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Kai Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Huan Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Zhenyi Xue
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Yurong Da
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Ning Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Yongcheng Hu
- Department of Orthopaedic Oncology, Tianjin Hospital, Tianjin, China.
| | - Zhi Yao
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Liang Qiao
- Storr Liver Unit, Westmead Millennium Institute, The Western Clinical School of the University of Sydney, Westmead, NSW, Australia.
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Rongxin Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| |
Collapse
|
44
|
Rhee KJ, Lee JI, Eom YW. Mesenchymal Stem Cell-Mediated Effects of Tumor Support or Suppression. Int J Mol Sci 2015; 16:30015-33. [PMID: 26694366 PMCID: PMC4691158 DOI: 10.3390/ijms161226215] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can exhibit a marked tropism towards site of tumors. Many studies have reported that tumor progression and metastasis increase by MSCs. In contrast, other studies have shown that MSCs suppress growth of tumors. MSCs contribute to tumor growth promotion by several mechanisms: (1) transition to tumor-associated fibroblasts; (2) suppression of immune response; (3) promotion of angiogenesis; (4) stimulation of epithelial-mesenchymal transition (EMT); (5) contribution to the tumor microenvironment; (6) inhibition of tumor cell apoptosis; and (7) promotion of tumor metastasis. In contrast to the tumor-promoting properties, MSCs inhibit tumor growth by increasing inflammatory infiltration, inhibiting angiogenesis, suppressing Wnt signaling and AKT signaling, and inducing cell cycle arrest and apoptosis. In this review, we will discuss potential mechanisms by which MSC mediates tumor support or suppression and then the possible tumor-specific therapeutic strategies using MSCs as delivery vehicles, based on their homing potential to tumors.
Collapse
Affiliation(s)
- Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Health Sciences, Yonsei University, 1 Yonseidae-gil, Wonju 26493, Korea.
| | - Jong In Lee
- Department of Hematology-Oncology, Wonju College of Medicine, Yonsei University, 20 Ilsan-ro, Wonju 26426, Korea.
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei University, 20 Ilsan-ro, Wonju 26426, Korea.
| |
Collapse
|
45
|
Pizzute T, Lynch K, Pei M. Impact of tissue-specific stem cells on lineage-specific differentiation: a focus on the musculoskeletal system. Stem Cell Rev Rep 2015; 11:119-32. [PMID: 25113801 DOI: 10.1007/s12015-014-9546-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tissue-specific stem cells are found throughout the body and, with proper intervention and environmental cues, these stem cells exercise their capabilities for differentiation into several lineages to form cartilage, bone, muscle, and adipose tissue in vitro and in vivo. Interestingly, it has been widely demonstrated that they do not differentiate with the same efficacy during lineage-specific differentiation studies, as the tissue-specific stem cells are generally more effective when differentiating toward the tissues from which they were derived. This review focuses on four mesodermal lineages for tissue-specific stem cell differentiation: adipogenesis, chondrogenesis, myogenesis, and osteogenesis. It is intended to give insight into current multilineage differentiation and comparative research, highlight and contrast known trends regarding differentiation, and introduce supporting evidence which demonstrates particular tissue-specific stem cells' superiority in lineage-specific differentiation, along with their resident tissue origins and natural roles. In addition, some epigenetic and transcriptomic differences between stem cells which may explain the observed trends are discussed.
Collapse
Affiliation(s)
- Tyler Pizzute
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | | | | |
Collapse
|
46
|
Jeong SY, Ha J, Lee M, Jin HJ, Kim DH, Choi SJ, Oh W, Yang YS, Kim JS, Kim BG, Chang JH, Cho DH, Jeon HB. Autocrine Action of Thrombospondin-2 Determines the Chondrogenic Differentiation Potential and Suppresses Hypertrophic Maturation of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Stem Cells 2015; 33:3291-303. [PMID: 26235673 DOI: 10.1002/stem.2120] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/30/2015] [Indexed: 12/13/2022]
Abstract
Previous studies have shown that mesenchymal stem cell (MSC)-based therapies have varying efficacies for the treatment of various diseases, including cartilage defects. In this study, we demonstrated that the chondrogenic differentiation potential of human umbilical cord blood-derived MSCs (hUCB-MSCs) obtained from different individual donors varies, and we investigated the molecular basis for this variation. Microarray gene expression analysis identified thrombospondin-2 (TSP2) as a candidate gene underlying the interindividual variation in the chondrogenic differentiation potential of hUCB-MSCs. To assess the association between TSP-2 and the differentiation potential, we evaluated chondrogenic differentiation of hUCB-MSCs treated with TSP2 siRNA. In addition, we studied the effect of supplementing exogenous recombinant TSP-2 on TSP2 siRNA-treated hUCB-MSCs. We found that TSP-2 autocrinally promoted chondrogenic differentiation of hUCB-MSCs via the Notch signaling pathway, which was confirmed in MSCs from other sources such as bone marrow and adipose tissue. Interestingly, we observed that TSP-2 attenuated hypertrophy, which inevitably occurs during chondrogenic differentiation of hUCB-MSCs. Our findings indicated that the variable chondrogenic differentiation potential of MSCs obtained from different donors is influenced by the TSP-2 level in the differentiating cells. Thus, the TSP-2 level can be used as a marker to select MSCs with superior chondrogenic differentiation potential for use in cartilage regeneration therapy.
Collapse
Affiliation(s)
- Sang Young Jeong
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Jueun Ha
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Miyoung Lee
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Hye Jin Jin
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Dong Hyun Kim
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Soo Jin Choi
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Yoon Sun Yang
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| | - Jae-Sung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Byung-Gyu Kim
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, School of Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jeong Ho Chang
- Department of Biology, Teachers College, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Hyung Cho
- Graduate School of East-West Medical Science, Kyung Hee University, Seoul, Republic of Korea
| | - Hong Bae Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Gyeonggi-do, Republic of Korea
| |
Collapse
|
47
|
Schira J, Falkenberg H, Hendricks M, Waldera-Lupa DM, Kögler G, Meyer HE, Müller HW, Stühler K. Characterization of Regenerative Phenotype of Unrestricted Somatic Stem Cells (USSC) from Human Umbilical Cord Blood (hUCB) by Functional Secretome Analysis. Mol Cell Proteomics 2015; 14:2630-43. [PMID: 26183719 DOI: 10.1074/mcp.m115.049312] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 12/13/2022] Open
Abstract
Stem cell transplantation is a promising therapeutic strategy to enhance axonal regeneration after spinal cord injury. Unrestricted somatic stem cells (USSC) isolated from human umbilical cord blood is an attractive stem cell population available at GMP grade without any ethical concerns. It has been shown that USSC transplantation into acute injured rat spinal cords leads to axonal regrowth and significant locomotor recovery, yet lacking cell replacement. Instead, USSC secrete trophic factors enhancing neurite growth of primary cortical neurons in vitro. Here, we applied a functional secretome approach characterizing proteins secreted by USSC for the first time and validated candidate neurite growth promoting factors using primary cortical neurons in vitro. By mass spectrometric analysis and exhaustive bioinformatic interrogation we identified 1156 proteins representing the secretome of USSC. Using Gene Ontology we revealed that USSC secretome contains proteins involved in a number of relevant biological processes of nerve regeneration such as cell adhesion, cell motion, blood vessel formation, cytoskeleton organization and extracellular matrix organization. We found for instance that 31 well-known neurite growth promoting factors like, e.g. neuronal growth regulator 1, NDNF, SPARC, and PEDF span the whole abundance range of USSC secretome. By the means of primary cortical neurons in vitro assays we verified SPARC and PEDF as significantly involved in USSC mediated neurite growth and therewith underline their role in improved locomotor recovery after transplantation. From our data we are convinced that USSC are a valuable tool in regenerative medicine as USSC's secretome contains a comprehensive network of trophic factors supporting nerve regeneration not only by a single process but also maintained its regenerative phenotype by a multitude of relevant biological processes.
Collapse
Affiliation(s)
- Jessica Schira
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany; §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Heiner Falkenberg
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Marion Hendricks
- §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Daniel M Waldera-Lupa
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Gesine Kögler
- ¶Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Medical Center, Düsseldorf, Germany
| | - Helmut E Meyer
- ‖Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Bunsen-Kirchhoff-Str. 11, Dortmund, Germany
| | - Hans Werner Müller
- §Molecular Neurobiology Laboratory, Department of Neurology, Heinrich Heine University Medical Centre Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; **Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Kai Stühler
- From the ‡Molecular Proteomics Laboratory (MPL), Institute for Molecular Medicine, Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany; **Biologisch-Medizinisches Forschungszentrum (BMFZ), Heinrich Heine University, Universitätsstr. 1, 40225 Düsseldorf, Germany
| |
Collapse
|
48
|
Georgi N, Taipaleenmaki H, Raiss CC, Groen N, Portalska KJ, van Blitterswijk C, de Boer J, Post JN, van Wijnen AJ, Karperien M. MicroRNA Levels as Prognostic Markers for the Differentiation Potential of Human Mesenchymal Stromal Cell Donors. Stem Cells Dev 2015; 24:1946-55. [PMID: 25915705 DOI: 10.1089/scd.2014.0534] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The ability of human mesenchymal stromal/stem cells (hMSCs) to differentiate into various mesenchymal cell lineages makes them a promising cell source for the use in tissue repair strategies. Since the differentiation potential of hMSCs differs between donors, it is necessary to establish biomarkers for the identification of donors with high differentiation potential. In this study, we show that microRNA (miRNA) expression levels are effective for distinguishing donors with high differentiation potential from low differentiation potential. Twenty hMSC donors were initially tested for marker expression and differentiation potential. In particular, the chondrogenic differentiation potential was evaluated on the basis of histological matrix formation, mRNA expression levels of chondrogenic marker genes, and quantitative glycosaminoglycan deposition. Three donors out of twenty were identified as donors with high chondrogenic potential, whereas nine showed moderate and eight showed low chondrogenic potential. Expression profiles of miRNAs involved in chondrogenesis and cartilage homeostasis were used for the distinction between high-performance hMSCs and low-performance hMSCs. Global mRNA expression profiles of the donors before the onset of chondrogenic differentiation revealed minor differences in gene expression between low and high chondrogenic performers. However, analysis of miRNA expression during a 7-day differentiation period identified miR-210 and miR-630 as positive regulators of chondrogenesis. In contrast, miR-181 and miR-34a, which are negative regulators of chondrogenesis, were upregulated during differentiation in low-performing donors. In conclusion, profiling of hMSC donors for a specific panel of miRNAs may have a prognostic value for selecting donors with high differentiation potential to improve hMSC-based strategies for tissue regeneration.
Collapse
Affiliation(s)
- Nicole Georgi
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Hanna Taipaleenmaki
- 2 Heisenberg-Group for Molecular Skeletal Biology, Department of Trauma-, Hand- and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf , Hamburg, Germany
| | - Christian C Raiss
- 3 Nanobiophysics Group, Faculty of Science and Technology, MESA+Institute for Nanotechnology, University of Twente , Enschede, the Netherlands
| | - Nathalie Groen
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Karolina Janaeczek Portalska
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Clemens van Blitterswijk
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Jan de Boer
- 4 Department of Tissue Regeneration, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Janine N Post
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| | - Andre J van Wijnen
- 5 Departments of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, Faculty of Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, the Netherlands
| |
Collapse
|
49
|
Hendijani F. Human mesenchymal stromal cell therapy for prevention and recovery of chemo/radiotherapy adverse reactions. Cytotherapy 2015; 17:509-25. [DOI: 10.1016/j.jcyt.2014.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/07/2014] [Accepted: 10/27/2014] [Indexed: 12/21/2022]
|
50
|
Popov B, Petrov N. pRb-E2F signaling in life of mesenchymal stem cells: Cell cycle, cell fate, and cell differentiation. Genes Dis 2014; 1:174-187. [PMID: 30258863 PMCID: PMC6150080 DOI: 10.1016/j.gendis.2014.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/14/2014] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various mesodermal lines forming fat, muscle, bone, and other lineages of connective tissue. MSCs possess plasticity and under special metabolic conditions may transform into cells of unusual phenotypes originating from ecto- and endoderm. After transplantation, MSCs release the humoral factors promoting regeneration of the damaged tissue. During last five years, the numbers of registered clinical trials of MSCs have increased about 10 folds. This gives evidence that MSCs present a new promising resource for cell therapy of the most dangerous diseases. The efficacy of the MSCs therapy is limited by low possibilities to regulate their conversion into cells of damaged tissues that is implemented by the pRb-E2F signaling. The widely accepted viewpoint addresses pRb as ubiquitous regulator of cell cycle and tumor suppressor. However, current publications suggest that basic function of the pRb-E2F signaling in development is to regulate cell fate and differentiation. Through facultative and constitutive chromatin modifications, pRb-E2F signaling promotes transient and stable cells quiescence, cell fate choice to differentiate, to senesce, or to die. Loss of pRb is associated with cancer cell fate. pRb regulates cell fate by retaining quiescence of one cell population in favor of commitment of another or by suppression of genes of different cell phenotype. pRb is the founder member of the "pocket protein" family possessing functional redundancy. Critical increase in the efficacy of the MSCs based cell therapy will depend on precise understanding of various aspects of the pRb-E2F signaling.
Collapse
Affiliation(s)
- Boris Popov
- Institute of Cytology, Russian Academy of Sciences, St.Petersburg, 4, Tikhoretsky Av., 194064, Russia
| | | |
Collapse
|