1
|
Yang M, Li Y, Liang Q, Dong H, Ma Y, Andersson G, Bongcam-Rudloff E, Ahmad HI, Fu X, Han J. Identification of lncRNAs involved in the hair follicle cycle transition of cashmere goats in response to photoperiod change. BMC Genomics 2025; 26:487. [PMID: 40375123 PMCID: PMC12080124 DOI: 10.1186/s12864-025-11675-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Cashmere goats, as one of the important domesticated animal species, are known for their high-quality fiber. The growth of cashmere has seasonal variations caused by photoperiodic changes, but the molecular genetic mechanisms underlying this phenotype including the functional role of long non-coding RNAs (lncRNA) is still poorly understood. RESULTS In this study, we analyzed the RNA-seq dataset of 39 Cashmere goat skin samples including all different growth stages and identified 1591 lncRNAs. These lncRNAs exhibited growth stage-specific expression patterns. Combining shortened light and hair follicle growth cycles, we found that 68% of differentially photo-responsive lncRNAs showed similar expression trends during transition phase I (early anagen to anagen phase). This suggests that the mechanism of light-controlled induction of hair follicles from early anagen to anagen is similar to that of transition phase I. According to weighted gene co-expression network analyses (WGCNA) analysis, it was found that two gene clusters and 10 hub lncRNAs participated in the transformation of hair follicle cycle, inducing hair follicles to enter the full growth phase in advance. These hub lncRNAs may regulate the development cycle of hair follicles through cis- or trans-regulation on clock genes, SLC superfamily genes, fibroblast growth factor genes. CONCLUSIONS This study identified the key lncRNAs and target genes probably participating in the transformation of hair follicle cycle. This study will help further elucidate the role of lncRNAs in the hair follicle cycle and development.
Collapse
Affiliation(s)
- Min Yang
- College of Animal Science and Technology, Shihezi University, Shihezi, 832061, China
| | - Yingying Li
- College of Animal Science and Technology, Shihezi University, Shihezi, 832061, China
| | - Qianqian Liang
- College of Animal Science and Technology, Shihezi University, Shihezi, 832061, China
| | - Huajiao Dong
- College of Animal Science and Technology, Shihezi University, Shihezi, 832061, China
| | - Yuehui Ma
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Göran Andersson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, 75007, Uppsala, Sweden
| | - Erik Bongcam-Rudloff
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, 75007, Uppsala, Sweden
| | - Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, Faculty of Veterinary and Animal Sciences, Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Xuefeng Fu
- Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, 830011, China
| | - Jilong Han
- College of Animal Science and Technology, Shihezi University, Shihezi, 832061, China.
| |
Collapse
|
2
|
Koczkowska M, Kostecka A, Zawrzykraj M, Myszczyński K, Skoniecka A, Deptuła M, Tymińska A, Czerwiec K, Jąkalski M, Zieliński J, Crossman DK, Crowley MR, Cichorek M, Skowron PM, Pikuła M, Piotrowski A. Identifying differentiation markers between dermal fibroblasts and adipose-derived mesenchymal stromal cells (AD-MSCs) in human visceral and subcutaneous tissues using single-cell transcriptomics. Stem Cell Res Ther 2025; 16:64. [PMID: 39934849 PMCID: PMC11818286 DOI: 10.1186/s13287-025-04185-w] [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: 05/16/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (AD-MSCs) and fibroblasts are both widely used in regenerative medicine, demonstrating significant potential for personalized cell therapy. A major challenge in their use lies in their high biological similarity, encompassing morphology, differentiation capabilities, and flow cytometric markers, making their distinction difficult. METHODS In our study, we aimed to compare AD-MSCs obtained from two types of adipose tissue, subcutaneous and visceral, alongside skin fibroblasts. Notably, all tissue samples were sourced from the same donors. We analyzed the cells for surface antigens via flow cytometry and conducted single-cell RNA sequencing, followed by verification with quantitative PCR (qPCR). RESULTS Our results revealed phenotypic similarities between the isolated AD-MSCs and dermal fibroblasts, particularly in the expression of markers characteristic of AD-MSCs. However, through in-depth analyses, we identified distinct differences between these cell types. Specifically, we pinpointed 30 genes exhibiting the most significant variations in expression between AD-MSCs and fibroblasts. These genes are associated with biological processes such as tissue remodeling, cell movement, and activation in response to external stimuli. Among them, MMP1, MMP3, S100A4, CXCL1, PI16, IGFBP5, COMP were further validated using qPCR, clearly demonstrating their potential to differentiate between AD-MSCs and fibroblasts. CONCLUSIONS Our scRNA-seq analysis elucidates the transcriptional landscape of AD-MSCs and fibroblasts with unprecedented resolution, highlighting both the population-specific markers and the intrapopulation heterogeneity. Our findings underscore the importance of employing high-resolution techniques for cell identification.
Collapse
Affiliation(s)
| | - Anna Kostecka
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Kamil Myszczyński
- Centre of Biostatistics and Bioinformatics Analysis, Medical University of Gdansk, Gdansk, Poland
| | - Aneta Skoniecka
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Milena Deptuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Agata Tymińska
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Katarzyna Czerwiec
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Jąkalski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Jacek Zieliński
- Department of Surgical Oncology, Transplant Surgery and General Surgery, Medical University of Gdansk, Gdansk, Poland
| | - David K Crossman
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael R Crowley
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mirosława Cichorek
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Michał Pikuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland.
| | - Arkadiusz Piotrowski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
3
|
Muntión S, Sánchez-Luis E, Díez-Campelo M, Blanco JF, Sánchez-Guijo F, De Las Rivas J. Novel Gene Biomarkers Specific to Human Mesenchymal Stem Cells Isolated from Bone Marrow. Int J Mol Sci 2024; 25:11906. [PMID: 39595975 PMCID: PMC11593895 DOI: 10.3390/ijms252211906] [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: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
In this paper, we present a comparative analysis of the transcriptomic profile of three different human cell types: hematopoietic stem cells (HSCs), bone marrow-derived mesenchymal stem cells (MSCs) and fibroblasts (FIBs). The work aims to identify unique genes that are differentially expressed as specific markers of bone marrow-derived MSCs, and to achieve this undertakes a detailed analysis of three independent datasets that include quantification of the global gene expression profiles of three primary cell types: HSCs, MSCs and FIBs. A robust bioinformatics method, called GlobalTest, is used to assess the specific association between one or more genes expressed in a sample and the outcome variable, that is, the 'cell type' provided as a single univariate response. This outcome variable is predicted for each sample tested, based on the expression profile of the specific genes that are used as input to the test. The precision of the tests is calculated along with the statistical sensitivity and specificity for each gene in each dataset, yielding four genes that mark MSCs with high accuracy. Among these, the best performer is the protein-coding gene Transgelin (TAGLN, Gene ID: 6876) (with a Positive Predictive Value > 0.96 and FDR < 0.001), which identifies MSCs better than any of the currently used standard markers: ENG (CD105), THY1 (CD90) or NT5E (CD73). The results are validated by RT-qPCR, providing novel gene biomarkers specific for human MSCs.
Collapse
Affiliation(s)
- Sandra Muntión
- Cell Therapy Area, Department of Hematology, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain; (S.M.); (F.S.-G.)
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain
| | - Elena Sánchez-Luis
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain;
- Bioinformatics Functional Genomics CANC-14 Group, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - María Díez-Campelo
- Department of Hematology, Center for Biomedical Research in Network of Cancer (CIBERONC), Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain;
- Department of Medicine, Faculty of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain;
| | - Juan F. Blanco
- Department of Medicine, Faculty of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain;
- Department of Trauma and Orthopedic Surgery, University Hospital of Salamanca (IBSAL-HUS), 37007 Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Cell Therapy Area, Department of Hematology, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain; (S.M.); (F.S.-G.)
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain
- Department of Hematology, Center for Biomedical Research in Network of Cancer (CIBERONC), Institute of Biomedical Research of Salamanca-Hospital Universitario de Salamanca (IBSAL-HUS), 37007 Salamanca, Spain;
- Department of Medicine, Faculty of Medicine, University of Salamanca (USAL), 37007 Salamanca, Spain;
| | - Javier De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), 37007 Salamanca, Spain;
- Bioinformatics Functional Genomics CANC-14 Group, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
4
|
Damerau A, Kirchner M, Mertins P, Buttgereit F, Gaber T. A point-of-research decision in synovial tissue engineering: Mesenchymal stromal cells, tissue derived fibroblast or CTGF-mediated mesenchymal-to-fibroblast transition. Eur J Cell Biol 2024; 103:151455. [PMID: 39293131 DOI: 10.1016/j.ejcb.2024.151455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/20/2024] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are prevalent inflammatory joint diseases characterized by synovitis, cartilage, and bone destruction. Fibroblast-like synoviocytes (FLSs) of the synovial membrane are a decisive factor in arthritis, making them a target for future therapies. Developing novel strategies targeting FLSs requires advanced in vitro joint models that accurately replicate non-diseased joint tissue. This study aims to identify a cell source reflecting physiological synovial fibroblasts. Therefore, we newly compared the phenotype and metabolism of "healthy" knee-derived FLSs from patients with ligament injuries (trauma-FLSs) to mesenchymal stromal cells (MSCs), their native precursors. We differentiated MSCs into fibroblasts using connective tissue growth factor (CTGF) and compared selected protein and gene expression patterns to those obtained from trauma-FLSs and OA-FLSs. Based on these findings, we explored the potential of an MSC-derived synovial tissue model to simulate a chronic inflammatory response akin to that seen in arthritis. We have identified MSCs as a suitable cell source for synovial tissue engineering because, despite metabolic differences, they closely resemble human trauma-derived FLSs. CTGF-mediated differentiation of MSCs increased HAS2 expression, essential for hyaluronan synthesis. It showed protein expression patterns akin to OA-FLSs, including markers of ECM components and fibrosis, and enzymes leading to a shift in metabolism towards increased fatty acid oxidation. In general, cytokine stimulation of MSCs in a synovial tissue model induced pro-inflammatory and pro-angiogenic gene expression, hyperproliferation, and increased glucose consumption, reflecting cellular response in human arthritis. We conclude that MSCs can serve as a proxy to study physiological synovial processes and inflammatory responses. In addition, CTGF-mediated mesenchymal-to-fibroblast transition resembles OA-FLSs. Thus, we emphasize MSCs as a valuable cell source for tools in preclinical drug screening and their application in tissue engineering.
Collapse
Affiliation(s)
- Alexandra Damerau
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Frank Buttgereit
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Timo Gaber
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.
| |
Collapse
|
5
|
Zou Z, Li H, Yu K, Ma K, Wang Q, Tang J, Liu G, Lim K, Hooper G, Woodfield T, Cui X, Zhang W, Tian K. The potential role of synovial cells in the progression and treatment of osteoarthritis. EXPLORATION (BEIJING, CHINA) 2023; 3:20220132. [PMID: 37933282 PMCID: PMC10582617 DOI: 10.1002/exp.20220132] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/15/2023] [Indexed: 11/08/2023]
Abstract
Osteoarthritis (OA), the commonest arthritis, is characterized by the progressive destruction of cartilage, leading to disability. The Current early clinical treatment strategy for OA often centers on anti-inflammatory or analgesia medication, weight loss, improved muscular function and articular cartilage repair. Although these treatments can relieve symptoms, OA tends to be progressive, and most patients require arthroplasty at the terminal stages of OA. Recent studies have shown a close correlation between joint pain, inflammation, cartilage destruction and synovial cells. Consequently, understanding the potential mechanisms associated with the action of synovial cells in OA could be beneficial for the clinical management of OA. Therefore, this review comprehensively describes the biological functions of synovial cells, the synovium, together with the pathological changes of synovial cells in OA, and the interaction between the cartilage and synovium, which is lacking in the present literature. Additionally, therapeutic approaches based on synovial cells for OA treatment are further discussed from a clinical perspective, highlighting a new direction in the treatment of OA.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Han Li
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Kai Yu
- Department of Bone and JointCentral Hospital of Zhuang He CityDalianLiaoningChina
| | - Ke Ma
- Department of Clinical MedicineChina Medical UniversityShenyangLiaoningChina
| | - Qiguang Wang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuanChina
| | - Junnan Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Guozhen Liu
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
| | - Khoon Lim
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Tim Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Xiaolin Cui
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Weiguo Zhang
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| | - Kang Tian
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| |
Collapse
|
6
|
Ryan CN, Pugliese E, Shologu N, Gaspar D, Rooney P, Islam MN, O'Riordan A, Biggs MJ, Griffin MD, Zeugolis DI. Physicochemical cues are not potent regulators of human dermal fibroblast trans-differentiation. BIOMATERIALS AND BIOSYSTEMS 2023; 11:100079. [PMID: 37720487 PMCID: PMC10499661 DOI: 10.1016/j.bbiosy.2023.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/25/2023] [Accepted: 05/29/2023] [Indexed: 09/19/2023] Open
Abstract
Due to their inherent plasticity, dermal fibroblasts hold great promise in regenerative medicine. Although biological signals have been well-established as potent regulators of dermal fibroblast function, it is still unclear whether physiochemical cues can induce dermal fibroblast trans-differentiation. Herein, we evaluated the combined effect of surface topography, substrate rigidity, collagen type I coating and macromolecular crowding in human dermal fibroblast cultures. Our data indicate that tissue culture plastic and collagen type I coating increased cell proliferation and metabolic activity. None of the assessed in vitro microenvironment modulators affected cell viability. Anisotropic surface topography induced bidirectional cell morphology, especially on more rigid (1,000 kPa and 130 kPa) substrates. Macromolecular crowding increased various collagen types, but not fibronectin, deposition. Macromolecular crowding induced globular extracellular matrix deposition, independently of the properties of the substrate. At day 14 (longest time point assessed), macromolecular crowding downregulated tenascin C (in 9 out of the 14 groups), aggrecan (in 13 out of the 14 groups), osteonectin (in 13 out of the 14 groups), and collagen type I (in all groups). Overall, our data suggest that physicochemical cues (such surface topography, substrate rigidity, collagen coating and macromolecular crowding) are not as potent as biological signals in inducing dermal fibroblast trans-differentiation.
Collapse
Affiliation(s)
- Christina N.M. Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Naledi Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Peadar Rooney
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Md Nahidul Islam
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, University of Galway, Galway, Ireland
- Discipline of Biochemistry, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Alan O'Riordan
- Tyndall National Institute, University College Cork (UCC), Cork, Ireland
| | - Manus J. Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
7
|
Zară-Dănceanu CM, Minuti AE, Stavilă C, Lăbuscă L, Herea DD, Tiron CE, Chiriac H, Lupu N. Magnetic Nanoparticle Coating Decreases the Senescence and Increases the Targeting Potential of Fibroblasts and Adipose-Derived Mesenchymal Stem Cells. ACS OMEGA 2023; 8:23953-23963. [PMID: 37426224 PMCID: PMC10324382 DOI: 10.1021/acsomega.3c02449] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023]
Abstract
Magnetic nanoparticles (MNPs) are intensely scrutinized for applications in emerging biomedical fields. Their potential use for drug delivery, tracking, and targeting agents or for cell handling is tested for regenerative medicine and tissue engineering applications. The large majority of MNPs tested for biomedical use are coated with different lipids and natural or synthetic polymers in order to decrease their degradation process and to increase the ability to transport drugs or bioactive molecules. Our previous studies highlighted the fact that the as-prepared MNP-loaded cells can display increased resistance to culture-induced senescence as well as ability to target pathological tissues; however, this effect tends to be dependent on the cell type. Here, we assessed comparatively the effect of two types of commonly used lipid coatings, oleic acid (OA) and palmitic acid (PA), on normal human dermal fibroblasts and adipose-derived mesenchymal cells with culture-induced senescence and cell motility in vitro. OA and PA coatings improved MNPs stability and dispersibility. We found good viability for cells loaded with all types of MNPs; however, a significant increase was obtained with the as-prepared MNPs and OA-MNPs. The coating decreases iron uptake in both cell types. Fibroblasts (Fb) integrate MNPs at a slower rate compared to adipose-derived mesenchymal stem cells (ADSCs). The as-prepared MNPs induced a significant decrease in beta-galactosidase (B-Gal) activity with a nonsignificant one observed for OA-MNPs and PA-MNPs in ADSCs and Fb. The as-prepared MNPs significantly decrease senescence-associated B-Gal enzymatic activity in ADSCs but not in Fb. Remarkably, a significant increase in cell mobility could be detected in ADSCs loaded with OA-MNPscompared to controls. The OA-MNPs uptake significantly increases ADSCs mobility in a wound healing model in vitro compared to nonloaded counterparts, while these observations need to be validated in vivo. The present findings provide evidence that support applications of OA-MNPs in wound healing and cell therapy involving reparative processes as well as organ and tissue targeting.
Collapse
Affiliation(s)
- Camelia-Mihaela Zară-Dănceanu
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
| | - Anca-Emanuela Minuti
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
- Faculty
of Physics, Alexandru Ioan Cuza University, 700506 Iaşi, Romania
| | - Cristina Stavilă
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
- Faculty
of Physics, Alexandru Ioan Cuza University, 700506 Iaşi, Romania
| | - Luminiţa Lăbuscă
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
- County
Emergency Hospital Saint Spiridon, Orthopedics
and Traumatology Clinic, Bulevardul Independenţei 1, 700111 Iaşi, Romania
| | - Dumitru-Daniel Herea
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
| | - Crina Elena Tiron
- Regional
Institute of Oncology, TRANSCEND Centre General Mathias Berthelot 2-4, 700483 Iaşi, Romania
| | - Horia Chiriac
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
| | - Nicoleta Lupu
- Department
of Magnetic Materials and Devices, National
Institute of Research and Development for Technical Physics, 47 Dimitrie Mangeron Boulevard, 700050 Iaşi, Romania
| |
Collapse
|
8
|
Braga GCD, Camargo CP, Harmsen MC, Correia AT, Souza S, Seelaender M, Nunes VA, dos Santos JF, Neri EA, Valadão IC, Moreira LFP, Gemperli R. A modified hydrogel production protocol to decrease cellular content. Acta Cir Bras 2022; 37:e371005. [PMID: 36542042 PMCID: PMC9762429 DOI: 10.1590/acb371005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/17/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To analyze the cytotoxicity and cell in porcine-derived decellularized skin matrix. METHODS We analyzed the effect of multiple decellularization processes by histological analysis, DNA quantification, and flow cytometry. Subsequently, we analyzed the most appropriate hydrogel concentration to minimize cytotoxicity on fibroblast culture and to maximize cell proliferation. RESULTS After the fourth decellularization, the DNA quantification showed the lowest DNA concentration (< 50 ng/mg). Histological analysis showed no cell components in the hydrogel. Moreover, hematoxylin and eosin showed a heterogeneous structure of collagen fibers. The best hydrogel concentration ranged from 3 to 25%, and there was no significant difference between the 24 hours and seven days. CONCLUSIONS The process of hydrogel production was effective for removing cells and DNA elements. The best hydrogel concentration ranged from 3 to 25%.
Collapse
Affiliation(s)
- Gabriela Catão Diniz Braga
- Bachelor. Universidade de São Paulo – Discipline of Plastic Surgery, Microsurgery and Plastic Surgery Laboratory – School of Medicine – São Paulo (SP), Brazil
| | - Cristina Pires Camargo
- PhD. Universidade de São Paulo – Discipline of Plastic Surgery, Microsurgery and Plastic Surgery Laboratory – School of Medicine – São Paulo (SP), Brazil.,Corresponding author:
- (55 11) 30620415
| | - Martin Conrad Harmsen
- PhD. Associate professor. University Medical Center Groningen – Laboratory for Cardiovascular Regenerative Medicine – Department of Pathology and Medical Biology – Hanzeplein 1, Netherlands
| | - Aristides Tadeu Correia
- PhD. Universidade de São Paulo – Department of Cardiopneumology – Thoracic Surgery Research Laboratory – Heart Institute of School of Medicine – São Paulo (SP), Brazil
| | - Sonia Souza
- Bachelor. Universidade de São Paulo – Department of Cardiopneumology – Cardiovascular Surgery and Circulatory Physiopathology Laboratory – School of Medicine – São Paulo (SP), Brazil
| | - Marilia Seelaender
- PhD. Associate professor. Universidade de São Paulo – Department of Clinical Surgery – School of Medicine – São Paulo (SP), Brazil
| | - Viviane Araujo Nunes
- PhD. Associate professor. Universidade de São Paulo – Department of Biotechnology – School of Arts, Sciences and Humanities – São Paulo (SP), Brazil
| | - Jeniffer Farias dos Santos
- PhD. Universidade de São Paulo – Department of Biotechnology – School of Arts, Sciences and Humanities – São Paulo (SP), Brazil
| | - Elida Adalgisa Neri
- PhD. Universidade de São Paulo – Laboratory of Genetics and Molecular Cardiology – Heart Institute – School of Medicine – São Paulo (SP), Brazil
| | - Iuri Cordeiro Valadão
- PhD. Universidade de São Paulo – Laboratory of Genetics and Molecular Cardiology – Heart Institute – School of Medicine – São Paulo (SP), Brazil
| | - Luiz Felipe Pinho Moreira
- PhD. Associate professor. Universidade de São Paulo – Department of Cardiopneumology, Cardiovascular Surgery and Circulatory Physiopathology Laboratory – School of Medicine – São Paulo (SP), Brazil
| | - Rolf Gemperli
- PhD. Full professor. Universidade de São Paulo – Discipline of Plastic Surgery, Microsurgery and Plastic Surgery Laboratory – School of Medicine – São Paulo (SP) Brazil
| |
Collapse
|
9
|
Álvarez-Vásquez JL, Castañeda-Alvarado CP. Dental pulp fibroblast: A star Cell. J Endod 2022; 48:1005-1019. [DOI: 10.1016/j.joen.2022.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 12/16/2022]
|
10
|
Zanata F, Curley L, Martin E, Bowles A, Bunnell BA, Wu X, Ferreira LM, Gimble JM. Comparative Analysis of Human Adipose-Derived Stromal/Stem Cells and Dermal Fibroblasts. Stem Cells Dev 2021; 30:1171-1178. [PMID: 34486404 DOI: 10.1089/scd.2021.0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dermal fibroblasts (DFs) share several qualities with mesenchymal stem cell/multipotent stromal cells (MSCs) derived from various tissues, including adipose-derived stromal/stem cells (ASCs). ASCs and DFs are morphologically comparable and both cell types can be culture expanded through the utilization of their plastic-adherence properties. Despite these similar characteristics, numerous studies indicate that ASC and DF display distinct therapeutic benefits in clinical applications. To more accurately distinguish between these cell types, human DFs and ASCs isolated from three individual donors were analyzed for multipotency and cell surface marker expressions. The detection of cell surface markers, CD29, CD34, CD44, CD73, CD90, and CD105, were used for phenotypic characterization of the DFs and ASCs. Furthermore, both cell types underwent lineage differentiation based on histochemical staining and the expression of adipogenic related genes, CCAAT/Enhancer-Binding Protein alpha (CEBPα), Peroxisome proliferator-activated receptor gamma (PPARγ), UCP1, Leptin (LEP), and Adiponectin (ADIPOQ); and osteogenic related genes, Runt related transcription factor 2 (Runx2), Alkaline phosphatase (ALPL), Osteocalcin (OCN), and Osteopontin (OPN). Evidence provided by this study demonstrates similarities between donor-matched ASC and DF with respect to morphology, surface marker expression, differentiation potential, and gene expression, although appearance of enhanced adipogenesis in the ASC based solely on spectrophotometric analyses with no significant difference in real-time polymerase chain reaction detection of adipogenic biomarkers. Thus, there is substantial overlap between the ASC and DF phenotypes based on biochemical and differentiation metrics.
Collapse
Affiliation(s)
- Fabiana Zanata
- Plastic Surgery Division, Universidade Federal de Sao Paulo UNIFESP/EPM, Sao Paulo, Brazil
| | | | - Elizabeth Martin
- Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Annie Bowles
- Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, The University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Xiying Wu
- La Cell LLC, New Orleans, Louisiana, USA
| | - Lydia Masako Ferreira
- Plastic Surgery Division, Universidade Federal de Sao Paulo UNIFESP/EPM, Sao Paulo, Brazil
| | - Jeffrey M Gimble
- La Cell LLC, New Orleans, Louisiana, USA.,Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
11
|
Sung SE, Seo MS, Kang KK, Choi JH, Lee S, Sung M, Kim K, Lee GW, Lim JH, Yang SY, Yim SG, Kim SK, Park S, Kwon YS, Yun S. Mesenchymal Stem Cell Exosomes Derived from Feline Adipose Tissue Enhance the Effects of Anti-Inflammation Compared to Fibroblasts-Derived Exosomes. Vet Sci 2021; 8:182. [PMID: 34564576 PMCID: PMC8473240 DOI: 10.3390/vetsci8090182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/22/2021] [Accepted: 08/29/2021] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue-derived mesenchymal stem cells (AD-MSCs) release extracellular vesicles such as exosomes, apoptotic bodies, and microparticles. In particular, exosomes are formed inside cells via multivesicular bodies (MVBs), thus their protein, DNA, and RNA content are similar to those of the parent cells. Exosome research is rapidly expanding, with an increase in the number of related publications observed in recent years; therefore, the function and application of MSC-derived exosomes could emerge as cell-free therapeutics. Exosomes have been isolated from feline AD-MSCs and feline fibroblast cell culture media using ultracentrifugation. Feline exosomes have been characterized by FACS, nanoparticle tracking analysis, and transmission electron microscopy imaging. Moreover, cytokine levels were detected by sandwich enzyme-linked immunosorbent assay in exosomes and LPS-induced THP-1 macrophages. The size of the isolated exosomes was that of a typical exosome, i.e., approximately 150 nm, and they expressed tetraspanins CD9 and CD81. The anti-inflammatory factor IL-10 was increased in feline AD-MSC-derived exosomes. However, pro-inflammatory factors such as IL-1β, IL-8, IL-2, RANTES, and IFN-gamma were significantly decreased in feline AD-MSC-derived exosomes. This was the first demonstration that feline AD-MSC-derived exosomes enhance the inflammatory suppressive effects and have potential for the treatment of immune diseases or as an inflammation-inhibition therapy.
Collapse
Affiliation(s)
- Soo-Eun Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
- Department of Biomaterials Science (BK21 Four Program), Pusan National University, Miryang 50463, Korea; (S.Y.Y.); (S.-G.Y.)
| | - Min-Soo Seo
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
| | - Kyung-Ku Kang
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
| | - Joo-Hee Choi
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
| | - Sijoon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
| | - Minkyoung Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
| | - Kilsoo Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (M.-S.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.); (K.K.)
- College of Veterinary Medicine, Kyungpook National University, 80 Daehakro, Daegu 41566, Korea
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170 Hyochung-ro, Daegu 42415, Korea; (G.W.L.); (J.-H.L.)
| | - Ju-Hyeon Lim
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170 Hyochung-ro, Daegu 42415, Korea; (G.W.L.); (J.-H.L.)
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk 28160, Korea
| | - Seung Yun Yang
- Department of Biomaterials Science (BK21 Four Program), Pusan National University, Miryang 50463, Korea; (S.Y.Y.); (S.-G.Y.)
| | - Sang-Gu Yim
- Department of Biomaterials Science (BK21 Four Program), Pusan National University, Miryang 50463, Korea; (S.Y.Y.); (S.-G.Y.)
| | - Seul-Ki Kim
- Efficacy Evaluation Team, Food Science R&D Center, KolmarBNH CO., LTD., 61Heolleungro 8-gil, Seoul 06800, Korea;
| | - Sangbum Park
- Institute for Quantitative Health Science & Engineering (IQ), Michigan State University, Auditorium Road 775 Woodlot Drive, East Lansing, MI 48824, USA;
- Division of Dermatology, Department of Medicine, College of Human Medicine, Michigan State University, Auditorium Road 775 Woodlot Drive, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, 775 Woodlot Drive, East Lansing, MI 48824, USA
| | - Young-Sam Kwon
- Department of Veterinary Surgery, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| | - Sungho Yun
- Department of Veterinary Surgery, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
12
|
Single-Cell Transcriptome Analysis of Human Adipose-Derived Stromal Cells Identifies a Contractile Cell Subpopulation. Stem Cells Int 2021; 2021:5595172. [PMID: 34007285 PMCID: PMC8102097 DOI: 10.1155/2021/5595172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Accepted: 03/27/2021] [Indexed: 12/02/2022] Open
Abstract
The potential for human adipose-derived stromal cells (hASCs) to be used as a therapeutic product is being assessed in multiple clinical trials. However, much is still to be learned about these cells before they can be used with confidence in the clinical setting. An inherent characteristic of hASCs that is not well understood is their heterogeneity. The aim of this exploratory study was to characterize the heterogeneity of freshly isolated hASCs after two population doublings (P2) using single-cell transcriptome analysis. A minimum of two subpopulations were identified at P2. A major subpopulation was identified as contractile cells which, based on gene expression patterns, are likely to be pericytes and/or vascular smooth muscle cells (vSMCs).
Collapse
|
13
|
Domenici G, Eduardo R, Castillo-Ecija H, Orive G, Montero Carcaboso Á, Brito C. PDX-Derived Ewing's Sarcoma Cells Retain High Viability and Disease Phenotype in Alginate Encapsulated Spheroid Cultures. Cancers (Basel) 2021; 13:cancers13040879. [PMID: 33669730 PMCID: PMC7922076 DOI: 10.3390/cancers13040879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/29/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Ewing’s Sarcoma (ES) is the second most frequent bone tumour in children and young adults, with very aggressive behaviour and significant disease recurrence. To better study the disease and find new therapies, experimental models are needed. Recently, patient-derived xenografts (PDX), obtained by implanting patient tumour samples in immunodeficient mice, have been developed. However, when ES cells are extracted from the patient’s tumour or from PDX and placed on plasticware surfaces, they lose their original 3D configuration, cell identity and function. To overcome these issues, we implemented cultures of PDX-derived ES cells, by making them aggregate to form ES cell spheroids and then encapsulating these 3D spheroids into a hydrogel, alginate, to stabilize the culture. We show that this methodology maintained ES cell viability and intrinsic characteristics of the original ES tumour cells for at least one month and that it is suitable for study the effect of anticancer drugs. Abstract Ewing’s Sarcoma (ES) is the second most frequent malignant bone tumour in children and young adults and currently only untargeted chemotherapeutic approaches and surgery are available as treatment, although clinical trials are on-going for recently developed ES-targeted therapies. To study ES pathobiology and develop novel drugs, established cell lines and patient-derived xenografts (PDX) are the most employed experimental models. Nevertheless, the establishment of ES cell lines is difficult and the extensive use of PDX raises economic/ethical concerns. There is a growing consensus regarding the use of 3D cell culture to recapitulate physiological and pathophysiological features of human tissues, including drug sensitivity. Herein, we implemented a 3D cell culture methodology based on encapsulation of PDX-derived ES cell spheroids in alginate and maintenance in agitation-based culture systems. Under these conditions, ES cells displayed high proliferative and metabolic activity, while retaining the typical EWSR1-FLI1 chromosomal translocation. Importantly, 3D cultures presented reduced mouse PDX cell contamination compared to 2D cultures. Finally, we show that these 3D cultures can be employed in drug sensitivity assays, with results similar to those reported for the PDX of origin. In conclusion, this novel 3D cell culture method involving ES-PDX-derived cells is a suitable model to study ES pathobiology and can assist in the development of novel drugs against this disease, complementing PDX studies.
Collapse
Affiliation(s)
- Giacomo Domenici
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (G.D.); (R.E.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Rodrigo Eduardo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (G.D.); (R.E.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Helena Castillo-Ecija
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Déu, Passeig Sant Joan de Déu 2, 08950 Barcelona, Spain; (H.C.-E.); (Á.M.C.)
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain;
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Ángel Montero Carcaboso
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Institut de Recerca Sant Joan de Déu, Passeig Sant Joan de Déu 2, 08950 Barcelona, Spain; (H.C.-E.); (Á.M.C.)
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (G.D.); (R.E.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- Correspondence:
| |
Collapse
|
14
|
Comparison of similar cells: Mesenchymal stromal cells and fibroblasts. Acta Histochem 2020; 122:151634. [PMID: 33059115 PMCID: PMC7550172 DOI: 10.1016/j.acthis.2020.151634] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/03/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022]
Abstract
Almost from all organs, both mesenchymal stromal cells and fibroblasts can be isolated. Mesenchymal stromal cells (MSCs) are the most preferred cellular therapeutic agents with the regenerative potential, and fibroblasts are one of the most abundant cell types with the ability to maintain homeostasis. Because of the promising properties of MSCs, they have been well studied and their differentiation potentials, immunomodulatory potentials, gene expression profiles are identified. It has been observed that fibroblasts and mesenchymal stromal cells have similar morphology, gene expression patterns, surface markers, proliferation, differentiation, and immunomodulatory capacities. Thus, it is hard to distinguish these two cell types. Epigenetic signatures, i.e., methylation patterns of cells, are the only usable promising difference between them. Such significant similarities show that these two cells may be related to each other.
Collapse
|
15
|
Zhu W, Cao L, Song C, Pang Z, Jiang H, Guo C. Cell-derived decellularized extracellular matrix scaffolds for articular cartilage repair. Int J Artif Organs 2020; 44:269-281. [PMID: 32945220 DOI: 10.1177/0391398820953866] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Articular cartilage repair remains a great clinical challenge. Tissue engineering approaches based on decellularized extracellular matrix (dECM) scaffolds show promise for facilitating articular cartilage repair. Traditional regenerative approaches currently used in clinical practice, such as microfracture, mosaicplasty, and autologous chondrocyte implantation, can improve cartilage repair and show therapeutic effect to some degree; however, the long-term curative effect is suboptimal. As dECM prepared by proper decellularization procedures is a biodegradable material, which provides space for regeneration tissue growth, possesses low immunogenicity, and retains most of its bioactive molecules that maintain tissue homeostasis and facilitate tissue repair, dECM scaffolds may provide a biomimetic microenvironment promoting cell attachment, proliferation, and chondrogenic differentiation. Currently, cell-derived dECM scaffolds have become a research hotspot in the field of cartilage tissue engineering, as ECM derived from cells cultured in vitro has many advantages compared with native cartilage ECM. This review describes cell types used to secrete ECM, methods of inducing cells to secrete cartilage-like ECM and decellularization methods to prepare cell-derived dECM. The potential mechanism of dECM scaffolds on cartilage repair, methods for improving the mechanical strength of cell-derived dECM scaffolds, and future perspectives on cell-derived dECM scaffolds are also discussed in this review.
Collapse
Affiliation(s)
- Wenrun Zhu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Cao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunfeng Song
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiying Pang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochen Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changan Guo
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Al Madhoun A, Marafie SK, Haddad D, Melhem M, Abu-Farha M, Ali H, Sindhu S, Atari M, Al-Mulla F. Comparative Proteomic Analysis Identifies EphA2 as a Specific Cell Surface Marker for Wharton's Jelly-Derived Mesenchymal Stem Cells. Int J Mol Sci 2020; 21:6437. [PMID: 32899389 PMCID: PMC7503404 DOI: 10.3390/ijms21176437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are a valuable tool in stem cell research due to their high proliferation rate, multi-lineage differentiation potential, and immunotolerance properties. However, fibroblast impurity during WJ-MSCs isolation is unavoidable because of morphological similarities and shared surface markers. Here, a proteomic approach was employed to identify specific proteins differentially expressed by WJ-MSCs in comparison to those by neonatal foreskin and adult skin fibroblasts (NFFs and ASFs, respectively). Mass spectrometry analysis identified 454 proteins with a transmembrane domain. These proteins were then compared across the different cell-lines and categorized based on their cellular localizations, biological processes, and molecular functions. The expression patterns of a selected set of proteins were further confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blotting, and immunofluorescence assays. As anticipated, most of the studied proteins had common expression patterns. However, EphA2, SLC25A4, and SOD2 were predominantly expressed by WJ-MSCs, while CDH2 and Talin2 were specific to NFFs and ASFs, respectively. Here, EphA2 was established as a potential surface-specific marker to distinguish WJ-MSCs from fibroblasts and for prospective use to prepare pure primary cultures of WJ-MSCs. Additionally, CDH2 could be used for a negative-selection isolation/depletion method to remove neonatal fibroblasts contaminating preparations of WJ-MSCs.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait;
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (M.M.); (H.A.); (F.A.-M.)
| | - Sulaiman K. Marafie
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.M.); (M.A.-F.)
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (M.M.); (H.A.); (F.A.-M.)
| | - Motasem Melhem
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (M.M.); (H.A.); (F.A.-M.)
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.M.); (M.A.-F.)
| | - Hamad Ali
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (M.M.); (H.A.); (F.A.-M.)
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya 046302, Kuwait
| | - Sardar Sindhu
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Maher Atari
- Medical-Surgical Pathology Department, Regenerative Medicine Research Institute, Universitat Internacional de Catalunya, 08195 Barcelona, Spain;
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (M.M.); (H.A.); (F.A.-M.)
| |
Collapse
|
17
|
The Role of Pref-1 during Adipogenic Differentiation: An Overview of Suggested Mechanisms. Int J Mol Sci 2020; 21:ijms21114104. [PMID: 32526833 PMCID: PMC7312882 DOI: 10.3390/ijms21114104] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022] Open
Abstract
Obesity contributes significantly to the global health burden. A better understanding of adipogenesis, the process of fat formation, may lead to the discovery of novel treatment strategies. However, it is of concern that the regulation of adipocyte differentiation has predominantly been studied using the murine 3T3-L1 preadipocyte cell line and murine experimental animal models. Translation of these findings to the human setting requires confirmation using experimental models of human origin. The ability of mesenchymal stromal/stem cells (MSCs) to differentiate into adipocytes is an attractive model to study adipogenesis in vitro. Differences in the ability of MSCs isolated from different sources to undergo adipogenic differentiation, may be useful in investigating elements responsible for regulating adipogenic differentiation potential. Genes involved may be divided into three broad categories: early, intermediate and late-stage regulators. Preadipocyte factor-1 (Pref-1) is an early negative regulator of adipogenic differentiation. In this review, we briefly discuss the adipogenic differentiation potential of MSCs derived from two different sources, namely adipose-derived stromal/stem cells (ASCs) and Wharton’s Jelly derived stromal/stem cells (WJSCs). We then discuss the function and suggested mechanisms of action of Pref-1 in regulating adipogenesis, as well as current findings regarding Pref-1’s role in human adipogenesis.
Collapse
|
18
|
Cao Y, Wang X, Peng G. SCSA: A Cell Type Annotation Tool for Single-Cell RNA-seq Data. Front Genet 2020; 11:490. [PMID: 32477414 PMCID: PMC7235421 DOI: 10.3389/fgene.2020.00490] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/20/2020] [Indexed: 11/13/2022] Open
Abstract
Currently most methods take manual strategies to annotate cell types after clustering the single-cell RNA sequencing (scRNA-seq) data. Such methods are labor-intensive and heavily rely on user expertise, which may lead to inconsistent results. We present SCSA, an automatic tool to annotate cell types from scRNA-seq data, based on a score annotation model combining differentially expressed genes (DEGs) and confidence levels of cell markers from both known and user-defined information. Evaluation on real scRNA-seq datasets from different sources with other methods shows that SCSA is able to assign the cells into the correct types at a fully automated mode with a desirable precision.
Collapse
Affiliation(s)
| | - Xiaoyue Wang
- Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Gongxin Peng
- Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Li J, Narayanan K, Zhang Y, Hill RC, He F, Hansen KC, Pei M. Role of lineage-specific matrix in stem cell chondrogenesis. Biomaterials 2019; 231:119681. [PMID: 31864016 DOI: 10.1016/j.biomaterials.2019.119681] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Cartilage repair in clinics is a challenge owing to the limited regenerative capacities of cartilage. Synovium-derived stem cells (SDSCs) are suggested as tissue-specific stem cells for chondrogenesis. In this study, we hypothesize that decellularized extracellular matrix (dECM) deposited by SDSCs could provide a superior tissue-specific matrix microenvironment for optimal rejuvenation of adult SDSCs for cartilage regeneration. dECMs were deposited by adult stem cells with varying chondrogenic capacities; SDSCs (strong) (SECM), adipose-derived stem cells (weak) (AECM) and dermal fibroblasts (weak) (DECM), and urine-derived stem cells (none) (UECM). Plastic flasks (Plastic) were used as a control substrate. Human SDSCs were expanded on the above substrates for one passage and examined for chondrogenic capacities. We found that each dECM consisted of unique matrix proteins and exhibited varied stiffnesses, which affected cell morphology and elasticity. Human SDSCs grown on dECMs displayed a significant increase in cell proliferation and unique surface phenotypes. Under induction media, dECM expanded cells yielded pellets with a dramatically increased number of chondrogenic markers. Interestingly, SECM expanded cells had less potential for hypertrophy compared to those grown on other dECMs, indicating that a tissue-specific matrix might provide a superior microenvironment for stem cell chondrogenic differentiation.
Collapse
Affiliation(s)
- Jingting Li
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Karthikeyan Narayanan
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV, 26506, USA
| | - Ying Zhang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, Shandong, 272067, China
| | - Ryan C Hill
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Kirk C Hansen
- Department of Biochemistry & Molecular Genetics, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
20
|
Moriyama M, Sahara S, Zaiki K, Ueno A, Nakaoji K, Hamada K, Ozawa T, Tsuruta D, Hayakawa T, Moriyama H. Adipose-derived stromal/stem cells improve epidermal homeostasis. Sci Rep 2019; 9:18371. [PMID: 31797970 PMCID: PMC6892794 DOI: 10.1038/s41598-019-54797-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/19/2019] [Indexed: 12/24/2022] Open
Abstract
Wound healing is regulated by complex interactions between the keratinocytes and other cell types including fibroblasts. Recently, adipose-derived mesenchymal stromal/stem cells (ASCs) have been reported to influence wound healing positively via paracrine involvement. However, their roles in keratinocytes are still obscure. Therefore, investigation of the precise effects of ASCs on keratinocytes in an in vitro culture system is required. Our recent data indicate that the epidermal equivalents became thicker on a collagen vitrigel membrane co-cultured with human ASCs (hASCs). Co-culturing the human primary epidermal keratinocytes (HPEK) with hASCs on a collagen vitrigel membrane enhanced their abilities for cell proliferation and adhesion to the membrane but suppressed their differentiation suggesting that hASCs could maintain the undifferentiated status of HPEK. Contrarily, the effects of co-culture using polyethylene terephthalate or polycarbonate membranes for HPEK were completely opposite. These differences may depend on the protein permeability and/or structure of the membrane. Taken together, our data demonstrate that hASCs could be used as a substitute for fibroblasts in skin wound repair, aesthetic medicine, or tissue engineering. It is also important to note that a co-culture system using the collagen vitrigel membrane allows better understanding of the interactions between the keratinocytes and ASCs.
Collapse
Affiliation(s)
- Mariko Moriyama
- Pharmaceutical Research and Technology Institute, Kindai University, Higashi-Osaka, Osaka, 577-8502, Japan
| | - Shunya Sahara
- Research and Development Division, PIAS Corporation, Kobe, Hyogo, 651-2241, Japan
| | - Kaori Zaiki
- Research and Development Division, PIAS Corporation, Kobe, Hyogo, 651-2241, Japan
| | - Ayumi Ueno
- Research and Development Division, PIAS Corporation, Kobe, Hyogo, 651-2241, Japan
| | - Koichi Nakaoji
- Research and Development Division, PIAS Corporation, Kobe, Hyogo, 651-2241, Japan
| | - Kazuhiko Hamada
- Research and Development Division, PIAS Corporation, Kobe, Hyogo, 651-2241, Japan
| | - Toshiyuki Ozawa
- Department of Dermatology, Graduate School of Medicine, Osaka City University, Abeno-Ku, Osaka, 545-8585, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Graduate School of Medicine, Osaka City University, Abeno-Ku, Osaka, 545-8585, Japan
| | - Takao Hayakawa
- Pharmaceutical Research and Technology Institute, Kindai University, Higashi-Osaka, Osaka, 577-8502, Japan
| | - Hiroyuki Moriyama
- Pharmaceutical Research and Technology Institute, Kindai University, Higashi-Osaka, Osaka, 577-8502, Japan.
| |
Collapse
|
21
|
Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. FEBS J 2019; 286:4160-4175. [PMID: 31365790 PMCID: PMC6899673 DOI: 10.1111/febs.15028] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/24/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022]
Abstract
Tumor immunotherapy has achieved remarkable efficacy, with immune-checkpoint inhibitors as especially promising candidates for cancer therapy. However, some issues caused by immunotherapy have raised attention, such as limited efficacy for some patients, narrow antineoplastic spectrum, and adverse reactions, suggesting that using regulators of tumor immune response may prove to be more complicated than anticipated. Current evidence indicates that different factors collectively constituting the unique tumor microenvironment promote immune tolerance, and these include the expression of co-inhibitory molecules, the secretion of lactate, and competition for nutrients between tumor cells and immune cells. Furthermore, cancer-associated fibroblasts, the main cellular components of solid tumors, promote immunosuppression through inhibition of T cell function and extracellular matrix remodeling. Here, we summarize the research advances in tumor immunotherapy and the latest insights into the influence of microenvironment on tumor immunotherapy.
Collapse
Affiliation(s)
- Jieying Zhang
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
- Research Center for Translational MedicineEast HospitalTongJi University School of MedicineShanghaiChina
| | - Zhaopeng Shi
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Xiang Xu
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Zuoren Yu
- Research Center for Translational MedicineEast HospitalTongJi University School of MedicineShanghaiChina
| | - Jun Mi
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
- Hongqiao International Institute of MedicineTongren HospitalShanghai Jiao Tong University School of MedicineChina
| |
Collapse
|
22
|
Zerdoum AB, Fowler EW, Jia X. Induction of Fibrogenic Phenotype in Human Mesenchymal Stem Cells by Connective Tissue Growth Factor in a Hydrogel Model of Soft Connective Tissue. ACS Biomater Sci Eng 2019; 5:4531-4541. [PMID: 33178886 PMCID: PMC7654958 DOI: 10.1021/acsbiomaterials.9b00425] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Scar formation is the typical endpoint of wound healing in adult mammalian tissues. An overactive or prolonged fibrogenic response following injury leads to excessive deposition of fibrotic proteins that promote tissue contraction and scar formation. Although well-defined in the dermal tissue, the progression of fibrosis is less explored in other connective tissues, such as the vocal fold. To establish a physiologically relevant 3D model of loose connective tissue fibrosis, we have developed a synthetic extracellular matrix using hyaluronic acid (HA) and peptidic building blocks carrying complementary functional groups. The resultant network was cell adhesive and protease degradable, exhibiting viscoelastic properties similar to the human vocal fold. Human mesenchymal stem cells (hMSCs) were encapsulated in the HA matrix as single cells or multicellular aggregates and cultured in pro-fibrotic media containing connective tissue growth factor (CTGF) for up to 21 days. hMSCs treated with CTGF-supplemented media exhibited an increased expression of fibrogenic markers and ECM proteins associated with scarring. Incorporation of α-smooth muscle actin into F-actin stress fibers was also observed. Furthermore, CTGF treatment increased the migratory capacity of hMSCs as compared to the CTGF-free control groups, indicative of the development of a myofibroblast phenotype. Addition of an inhibitor of the mitogen-activated protein kinase (MAPK) pathway attenuated cellular expression of fibrotic markers and related ECM proteins. Overall, this study demonstrates that CTGF promotes the development of a fibrogenic phenotype in hMSCs encapsulated within an HA matrix and that the MAPK pathway is a potential target for future therapeutic endeavors towards limiting scar formation in loose connective tissues.
Collapse
Affiliation(s)
- Aidan B. Zerdoum
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Eric W. Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | - Xinqiao Jia
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
23
|
Li F, Tang Y, Song B, Yu M, Li Q, Zhang C, Hou J, Yang R. Nomenclature clarification: synovial fibroblasts and synovial mesenchymal stem cells. Stem Cell Res Ther 2019; 10:260. [PMID: 31426847 PMCID: PMC6701095 DOI: 10.1186/s13287-019-1359-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Synovial-derived cells, found in the synovial membrane of human joints, were obtained by digestion of the synovial membrane and were subsequently expanded in vitro. The identity of synovial-derived cells has long been a topic of debate. The terms "type B synoviocytes," "fibroblast-like synoviocytes (FLS)," "synovium-derived mesenchymal stem cells (MSCs)," and "synovial fibroblasts (SF)" appeared in different articles related to human synovial-derived cells in various disease models, yet they seemed to be describing the same cell type. However, to date, there is no clear standard to distinguish these terms; thus, the hypothesis that they represent the same cell type is currently inconclusive. Therefore, this review aims to clarify the similarities and differences between these terms and to diffuse the chaotic nomenclature of synovial-derived cells.
Collapse
Affiliation(s)
- Fangqi Li
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China
| | - Yiyong Tang
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China
| | - Bin Song
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China
| | - Menglei Yu
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China
| | - Qingyue Li
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China
| | - Congda Zhang
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China
| | - Jingyi Hou
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China.
| | - Rui Yang
- Department of Orthopedic, Sun Yat-sen Memorial Hospital Sun Yat-sen University, NO.107 Yan Jiang West Road, Guangzhou, Guangdong Province, 510120, China.
| |
Collapse
|
24
|
Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. THE FEBS JOURNAL 2019. [PMID: 31365790 DOI: 10.1111/febs.15028.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
Tumor immunotherapy has achieved remarkable efficacy, with immune-checkpoint inhibitors as especially promising candidates for cancer therapy. However, some issues caused by immunotherapy have raised attention, such as limited efficacy for some patients, narrow antineoplastic spectrum, and adverse reactions, suggesting that using regulators of tumor immune response may prove to be more complicated than anticipated. Current evidence indicates that different factors collectively constituting the unique tumor microenvironment promote immune tolerance, and these include the expression of co-inhibitory molecules, the secretion of lactate, and competition for nutrients between tumor cells and immune cells. Furthermore, cancer-associated fibroblasts, the main cellular components of solid tumors, promote immunosuppression through inhibition of T cell function and extracellular matrix remodeling. Here, we summarize the research advances in tumor immunotherapy and the latest insights into the influence of microenvironment on tumor immunotherapy.
Collapse
Affiliation(s)
- Jieying Zhang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China.,Research Center for Translational Medicine, East Hospital, TongJi University School of Medicine, Shanghai, China
| | - Zhaopeng Shi
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China
| | - Xiang Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China
| | - Zuoren Yu
- Research Center for Translational Medicine, East Hospital, TongJi University School of Medicine, Shanghai, China
| | - Jun Mi
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China.,Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
25
|
Directional Topography Influences Adipose Mesenchymal Stromal Cell Plasticity: Prospects for Tissue Engineering and Fibrosis. Stem Cells Int 2019; 2019:5387850. [PMID: 31191675 PMCID: PMC6525798 DOI: 10.1155/2019/5387850] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/24/2018] [Accepted: 02/11/2019] [Indexed: 01/17/2023] Open
Abstract
Introduction Progenitor cells cultured on biomaterials with optimal physical-topographical properties respond with alignment and differentiation. Stromal cells from connective tissue can adversely differentiate to profibrotic myofibroblasts or favorably to smooth muscle cells (SMC). We hypothesized that myogenic differentiation of adipose tissue-derived stromal cells (ASC) depends on gradient directional topographic features. Methods Polydimethylsiloxane (PDMS) samples with nanometer and micrometer directional topography gradients (wavelength (w) = 464-10, 990 nm; amplitude (a) = 49-3, 425 nm) were fabricated. ASC were cultured on patterned PDMS and stimulated with TGF-β1 to induce myogenic differentiation. Cellular alignment and adhesion were assessed by immunofluorescence microscopy after 24 h. After seven days, myogenic differentiation was examined by immunofluorescence microscopy, gene expression, and immunoblotting. Results Cell alignment occurred on topographies larger than w = 1758 nm/a = 630 nm. The number and total area of focal adhesions per cell were reduced on topographies from w = 562 nm/a = 96 nm to w = 3919 nm/a = 1430 nm. Focal adhesion alignment was increased on topographies larger than w = 731 nm/a = 146 nm. Less myogenic differentiation of ASC occurred on topographies smaller than w = 784 nm/a = 209 nm. Conclusion ASC adherence, alignment, and differentiation are directed by topographical cues. Our evidence highlights a minimal topographic environment required to facilitate the development of aligned and differentiated cell layers from ASC. These data suggest that nanotopography may be a novel tool for inhibiting fibrosis.
Collapse
|
26
|
Zomer HD, Varela GKDS, Delben PB, Heck D, Jeremias TDS, Trentin AG. In vitro comparative study of human mesenchymal stromal cells from dermis and adipose tissue for application in skin wound healing. J Tissue Eng Regen Med 2019; 13:729-741. [PMID: 30773827 DOI: 10.1002/term.2820] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 11/09/2018] [Accepted: 02/13/2019] [Indexed: 12/31/2022]
Abstract
Novel strategies combining cell therapy, tissue engineering, and regenerative medicine have been developed to treat major skin wounds. Although mesenchymal stromal cells (MSCs) from different tissues have similar stem cell features, such as self-renewing mesodermal differentiation potential and expression of immunophenotypic markers, they also have distinct characteristics. Therefore, we aimed to characterize the application of MSCs derived from the dermis and adipose tissue (DSCs and ASCs, respectively) in cutaneous wound healing by in vitro approaches. Human DSC and ASC were obtained and evaluated for their isolation efficiency, stemness, proliferative profile, and genetic stability over time in culture. The ability of wound closure was first assessed by direct cell scratch assay. The paracrine effects of DSC- and ASC-conditioned medium in dermal fibroblasts and keratinocytes and in the induction of tubule formation were also investigated. Although the ASC isolation procedures resulted in 100 times more cells than DSC, the latter had a higher proliferation rate in culture. Both presented low frequency of nuclear alterations over time in culture and showed similar characteristics of stem cells, such as expression of immunophenotypic markers and differentiation potential. DSCs showed increased healing capacity, and their conditioned media had greater paracrine effect in closing the wound of dermal fibroblasts and keratinocytes and in inducing angiogenesis. In conclusion, the therapeutic potential of MSCs is influenced by the obtainment source. Both ASCs and DSCs are applicable for skin wound healing; however, DSCs have an improved potential and should be considered for future applications in cell therapy.
Collapse
Affiliation(s)
- Helena Debiazi Zomer
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, United States of America.,Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Priscilla Barros Delben
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Diana Heck
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Talita da Silva Jeremias
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Andrea Gonçalves Trentin
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Kitada M, Murakami T, Wakao S, Li G, Dezawa M. Direct conversion of adult human skin fibroblasts into functional Schwann cells that achieve robust recovery of the severed peripheral nerve in rats. Glia 2019; 67:950-966. [PMID: 30637802 DOI: 10.1002/glia.23582] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 12/03/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022]
Abstract
Direct conversion is considered a promising approach to obtain tissue-specific cells for cell therapies; however, this strategy depends on exogenous gene expression that may cause undesired adverse effects such as tumorigenesis. By optimizing the Schwann cell induction system, which was originally developed for trans-differentiation of bone marrow mesenchymal stem cells into Schwann cells, we established a system to directly convert adult human skin fibroblasts into cells comparable to authentic human Schwann cells without gene introduction. Serial treatments with beta-mercaptoethanol, retinoic acid, and finally a cocktail of basic fibroblast growth factor, forskolin, platelet-derived growth factor-AA, and heregulin-β1 (EGF domain) converted fibroblasts into cells expressing authentic Schwann cell markers at an efficiency of approximately 75%. Genome-wide gene expression analysis suggested the conversion of fibroblasts into the Schwann cell-lineage. Transplantation of induced Schwann cells into severed peripheral nerve of rats facilitated axonal regeneration and robust functional recovery in sciatic function index comparable to those of authentic human Schwann cells. The contributions of induced Schwann cells to myelination of regenerated axons and re-formation of neuromuscular junctions were also demonstrated. Our data clearly demonstrated that cells comparable to functional Schwann cells feasible for the treatment of neural disease can be induced from adult human skin fibroblasts without gene introduction. This direct conversion system will be beneficial for clinical applications to peripheral and central nervous system injuries and demyelinating diseases.
Collapse
Affiliation(s)
- Masaaki Kitada
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Murakami
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Gen Li
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
28
|
Wolf DA, Beeson W, Rachel JD, Keller GS, Hanke CW, Waibel J, Leavitt M, Sacopulos M. Mesothelial Stem Cells and Stromal Vascular Fraction for Skin Rejuvenation. Facial Plast Surg Clin North Am 2018; 26:513-532. [PMID: 30213431 DOI: 10.1016/j.fsc.2018.06.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The use of stem cells in regenerative medicine and specifically facial rejuvenation is thought provoking and controversial. Today there is increased emphasis on tissue engineering and regenerative medicine, which translates into a need for a reliable source of stem cells in addition to biomaterial scaffolds and cytokine growth factors. Adipose tissue is currently recognized as an accessible and abundant source for adult stem cells. Cellular therapies and tissue engineering are still in their infancy, and additional basic science and preclinical studies are needed before cosmetic and reconstructive surgical applications can be routinely undertaken and satisfactory levels of patient safety achieved.
Collapse
Affiliation(s)
- David A Wolf
- Johnson Space Center, Houston, TX, USA; EarthTomorrow, Inc, 1714 Neptune Lane, Houston, TX 77062, USA; Purdue University, West Lafayette, IN, USA
| | - William Beeson
- Facial Plastics, Indianapolis, IN, USA; Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | | - Gregory S Keller
- Facial Plastics, Santa Barbara, CA, USA; Facial Plastics, Los Angeles, CA, USA
| | - C William Hanke
- Dermatology, Indianapolis, IN, USA; Laser and Skin Center of Indiana, 13400 North Meridian Street, Suite 290, Carmel, IN 46032, USA; ACGME Micrographic Surgery, Dermatologic Oncology Fellowship Training Program, St. Vincent Hospital, Indianapolis, IN, USA; University of Iowa-Carver College of Medicine, Iowa City, IA, USA; University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jill Waibel
- Dermatology, Miami Dermatology and Laser Institute, 7800 Southwest 87th Avenue, Suite B200, Miami, FL 33173, USA; Baptist Hospital of Miami, Miami, FL, USA; Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Matt Leavitt
- Dermatology, Orlando, FL, USA; Advanced Dermatology and Cosmetic Surgery, The Hair Foundation, 260 Lookout Place Suite 103, Maitland, FL 32751, USA; University of Central Florida, 6850 Lake Nona Boulevard, Orlando, FL 32827, USA; Nova Southeastern University, 4850 Millenium Boulevard, Orlando, FL 32839, USA
| | - Michael Sacopulos
- Medical Risk Management, Medical Risk Institute, 676 Ohio Street, Terre Haute, IN 47807, USA
| |
Collapse
|
29
|
Differential Proteomic Analysis Predicts Appropriate Applications for the Secretome of Adipose-Derived Mesenchymal Stem/Stromal Cells and Dermal Fibroblasts. Stem Cells Int 2018; 2018:7309031. [PMID: 30158987 PMCID: PMC6109467 DOI: 10.1155/2018/7309031] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/03/2018] [Indexed: 12/12/2022] Open
Abstract
The adult stem cell secretome is currently under investigation as an alternative to cell-based therapy in regenerative medicine, thanks to the remarkable translational opportunity and the advantages in terms of handling and safety. In this perspective, we recently demonstrated the efficient performance of the adipose-derived mesenchymal stem/stromal cell (ASC) secretome in contrasting neuroinflammation in a murine model of diabetic neuropathy, where the administration of factors released by dermal fibroblasts (DFs) did not exert any effect. Up to now, the complex mixture of the constituents of the conditioned medium from ASCs has not been fully deepened, although its appropriate characterization is required in the perspective of a clinical use. Herein, we propose the differential proteomic approach for the identification of the players accounting for the functional effects of the cell secretome with the aim to unravel its appropriate applications. Out of 967 quantified proteins, 34 and 62 factors were found preponderantly or exclusively secreted by ASCs and DFs, respectively. This approach led to the recognition of distinct functions related to the conditioned medium of ASCs and DFs, with the former being involved in the regulation of neuronal death and apoptosis and the latter in bone metabolism and ossification. The proosteogenic effect of DF secretome was validated in vitro on human primary osteoblasts, providing a proof of concept of its osteoinductive potential. Besides discovering new applications of the cell type-specific secretome, the proposed strategy could allow the recognition of the cocktail of bioactive factors which might be responsible for the effects of conditioned media, thus providing a solid rationale to the implementation of a cell-free approach in several clinical scenarios involving tissue regeneration.
Collapse
|
30
|
Soundararajan M, Kannan S. Fibroblasts and mesenchymal stem cells: Two sides of the same coin? J Cell Physiol 2018; 233:9099-9109. [DOI: 10.1002/jcp.26860] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Suresh Kannan
- Department of Biomedical Sciences Sri Ramachandra University Chennai Tamil Nadu India
| |
Collapse
|
31
|
Ito N, Katoh K, Kushige H, Saito Y, Umemoto T, Matsuzaki Y, Kiyonari H, Kobayashi D, Soga M, Era T, Araki N, Furuta Y, Suda T, Kida Y, Ohta K. Ribosome Incorporation into Somatic Cells Promotes Lineage Transdifferentiation towards Multipotency. Sci Rep 2018; 8:1634. [PMID: 29374279 PMCID: PMC5786109 DOI: 10.1038/s41598-018-20057-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 01/12/2018] [Indexed: 01/10/2023] Open
Abstract
Recently, we reported that bacterial incorporation induces cellular transdifferentiation of human fibroblasts. However, the bacterium-intrinsic cellular- transdifferentiation factor remained unknown. Here, we found that cellular transdifferentiation is caused by ribosomes. Ribosomes, isolated from both prokaryotic and eukaryotic cells, induce the formation of embryoid body-like cell clusters. Numerous ribosomes are incorporated into both the cytoplasm and nucleus through trypsin-activated endocytosis, which leads to cell-cluster formation. Although ribosome-induced cell clusters (RICs) express several stemness markers and differentiate into derivatives of all three germ layers in heterogeneous cell populations, RICs fail to proliferate, alter the methylation states of pluripotent genes, or contribute to teratoma or chimera formation. However, RICs express markers of epithelial-mesenchymal transition without altering the cell cycle, despite their proliferation obstruction. These findings demonstrate that incorporation of ribosomes into host cells induces cell transdifferentiation and alters cellular plasticity.
Collapse
Affiliation(s)
- Naofumi Ito
- Department of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.,Program for Leading Graduate Schools "HIGO Program", Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Hiroko Kushige
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Yutaka Saito
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), 2-4-7 Aomi, Koto-ku, Tokyo, 135-0064, Japan
| | - Terumasa Umemoto
- International Research Center for Medical Science, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City, 860-0811, Japan
| | - Yu Matsuzaki
- International Research Center for Medical Science, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City, 860-0811, Japan
| | - Hiroshi Kiyonari
- Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Daiki Kobayashi
- Department of Tumor Genetics and Biology, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Minami Soga
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Norie Araki
- Department of Tumor Genetics and Biology, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, RIKEN Center for Life Science Technologies, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Toshio Suda
- International Research Center for Medical Science, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City, 860-0811, Japan.,Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore, Singapore
| | - Yasuyuki Kida
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Kunimasa Ohta
- Department of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan. .,Program for Leading Graduate Schools "HIGO Program", Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan. .,International Research Core for Stem Cell-based Developmental Medicine, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 860-8556, Japan. .,Japan Agency for Medical Research and Development (AMED), Tokyo, 100-0004, Japan.
| |
Collapse
|
32
|
Pageon H, Zucchi H, Rousset F, Girardeau-Hubert S, Tancrede E, Asselineau D. Glycation stimulates cutaneous monocyte differentiation in reconstructed skin in vitro. Mech Ageing Dev 2017; 162:18-26. [PMID: 28163107 DOI: 10.1016/j.mad.2017.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/07/2016] [Indexed: 12/12/2022]
Abstract
Glycation reaction is a recognized mechanism related to chronological aging. Previous investigations in cutaneous biology have considered the effect of glycation on the dermal matrix molecules, involved in tissue stiffening during skin aging. However, little is known about a possible direct effect of glycation upon cell differentiation. To address such issue, the effect of glycation has been re-investigated in a reconstructed skin model integrating monocytes that are cells capable of differentiating according to different pathways. The results showed that, in the absence of glycation, a small number of these CD45+ cells could differentiate either into dendritic-like cells (DC-SIGN+, BDC1a+, DC-LAMP+) or macrophage- like cells (CD14+, CD68+, CD163+) whereas, with glycation, the number of monocytes, dendritic cells, macrophage-like cells were found surprisingly increased. In-vivo our results showed also that dendritic and macrophage-like cells were increased and suggest a possible link with the age-dependent glycation level in the skin. In addition, we found that, unlike fibroblasts incorporated in the reconstructed skin, these cells expressed specific receptors for AGEs (RAGE and SRA). Taken altogether, our data show that cells of the monocyte lineage, in the presence of AGEs, can differentiate into dendritic or macrophage-like cells and could lead to a micro inflammatory environment.
Collapse
Affiliation(s)
- H Pageon
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France.
| | - H Zucchi
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - F Rousset
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - S Girardeau-Hubert
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - E Tancrede
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| | - D Asselineau
- L'Oréal Research & Innovation, 1 Avenue Eugène Schueller, 93600 Aulnay-sous-Bois, France
| |
Collapse
|
33
|
Fang F, Ni K, Cai Y, Ye Z, Shang J, Shen S, Xiong C. Biological characters of human dermal fibroblasts derived from foreskin of male infertile patients. Tissue Cell 2017; 49:56-63. [DOI: 10.1016/j.tice.2016.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 11/22/2016] [Accepted: 12/07/2016] [Indexed: 12/15/2022]
|
34
|
Foglietta F, Duchi S, Canaparo R, Varchi G, Lucarelli E, Dozza B, Serpe L. Selective sensitiveness of mesenchymal stem cells to shock waves leads to anticancer effect in human cancer cell co-cultures. Life Sci 2017; 173:28-35. [PMID: 28131762 DOI: 10.1016/j.lfs.2017.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 12/13/2022]
Abstract
AIM Mesenchymal stem cells (MSC) possess the distinctive feature of homing in on and engrafting into the tumor stroma making their therapeutic applications in cancer treatment very promising. Research into new effectors and external stimuli, which can selectively trigger the release of cytotoxic species from MSC toward the cancer cells, significantly raises their potential. MAIN METHODS Shock waves (SW) have recently gained recognition for their ability to induce specific biological effects, such as the local generation of cytotoxic reactive oxygen species (ROS) in a non-invasive and tunable manner. We thus investigate whether MSC are able to generate ROS and, in turn, affect cancer cell growth when in co-culture with human glioblastoma (U87) or osteosarcoma (U2OS) cells and exposed to SW. KEY FINDINGS MSC were found to be the cell line that was most sensitive to SW treatment as shown by SW-induced ROS production and cytotoxicity. Notably, U87 and U2OS cancer cell growth was unaffected by SW exposure. However, significant decreases in cancer cell growth, 1.8 fold for U87 and 2.3 fold for U2OS, were observed 24h after the SW treatment of MSC co-cultures with cancer cells. The ROS production induced in MSC by SW exposure was then responsible for lipid peroxidation and cell death in U87 and U2OS cells co-cultured with MSC. SIGNIFICANCE This experiment highlights the unique ability of MSC to generate ROS upon SW treatment and induce the cell death of co-cultured cancer cells. SW might therefore be proposed as an innovative tool for MSC-mediated cancer treatment.
Collapse
Affiliation(s)
| | - Serena Duchi
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Roberto Canaparo
- Department of Drug Science and Technology, University of Torino, Italy.
| | - Greta Varchi
- National Research Council, Institute for the Organic Synthesis and Photoreactivity, Bologna, Italy
| | - Enrico Lucarelli
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Barbara Dozza
- Osteoarticular Regeneration Laboratory, Rizzoli Orthopaedic Institute, Bologna, Italy; Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Italy
| | - Loredana Serpe
- Department of Drug Science and Technology, University of Torino, Italy
| |
Collapse
|
35
|
Mesenchymal Stem Cells from Adipose Tissue in Clinical Applications for Dermatological Indications and Skin Aging. Int J Mol Sci 2017; 18:ijms18010208. [PMID: 28117680 PMCID: PMC5297838 DOI: 10.3390/ijms18010208] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
Operating at multiple levels of control, mesenchymal stem cells from adipose tissue (ADSCs) communicate with organ systems to adjust immune response, provide signals for differentiation, migration, enzymatic reactions, and to equilibrate the regenerative demands of balanced tissue homeostasis. The identification of the mechanisms by which ADSCs accomplish these functions for dermatological rejuvenation and wound healing has great potential to identify novel targets for the treatment of disorders and combat aging. Herein, we review new insights into the role of adipose-derived stem cells in the maintenance of dermal and epidermal homeostasis, and recent advances in clinical applications of ADSCs related to dermatology.
Collapse
|
36
|
Jacków J, Titeux M, Portier S, Charbonnier S, Ganier C, Gaucher S, Hovnanian A. Gene-Corrected Fibroblast Therapy for Recessive Dystrophic Epidermolysis Bullosa using a Self-Inactivating COL7A1 Retroviral Vector. J Invest Dermatol 2016; 136:1346-1354. [DOI: 10.1016/j.jid.2016.02.811] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/12/2016] [Accepted: 02/26/2016] [Indexed: 12/16/2022]
|
37
|
Denu RA, Nemcek S, Bloom DD, Goodrich AD, Kim J, Mosher DF, Hematti P. Fibroblasts and Mesenchymal Stromal/Stem Cells Are Phenotypically Indistinguishable. Acta Haematol 2016; 136:85-97. [PMID: 27188909 DOI: 10.1159/000445096] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 03/01/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIMS Human mesenchymal stromal/stem cells (MSCs), derived from many different tissues, are characterized by a fibroblast-like morphology, the expression of certain cell surface markers and their ability to differentiate into adipocytes, chondrocytes and osteoblasts. A number of studies have shown that MSCs share many characteristics with fibroblasts; however, there is no well-defined set of phenotypic characteristics that could distinguish between these 2 types of cells. METHODS We used 4 well-established human fibroblast strains from 3 different tissue sources and several human MSC strains from 2 different tissue sources to compare the phenotypic and immunological characteristics of these cells. RESULTS Fibroblast strains had a similar morphology to MSCs, expressed the same cell surface markers as MSCs and could also differentiate into adipocytes, chondrocytes and osteoblasts. Also, similar to MSCs, these fibroblasts were capable of suppressing T cell proliferation and modulating the immunophenotype of macrophages. We also show that MSCs deposit extracellular matrices of collagen type I and fibronectin, and express FSP1 in patterns similar to fibroblasts. CONCLUSIONS Based on currently accepted definitions for cultured human MSCs and fibroblasts, we could not find any immunophenotypic property that could make a characteristic distinction between MSCs and fibroblasts.
Collapse
Affiliation(s)
- Ryan A Denu
- Department of Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wis., USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Gosnell ME, Anwer AG, Mahbub SB, Menon Perinchery S, Inglis DW, Adhikary PP, Jazayeri JA, Cahill MA, Saad S, Pollock CA, Sutton-McDowall ML, Thompson JG, Goldys EM. Quantitative non-invasive cell characterisation and discrimination based on multispectral autofluorescence features. Sci Rep 2016; 6:23453. [PMID: 27029742 PMCID: PMC4814840 DOI: 10.1038/srep23453] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/07/2016] [Indexed: 02/08/2023] Open
Abstract
Automated and unbiased methods of non-invasive cell monitoring able to deal with complex biological heterogeneity are fundamentally important for biology and medicine. Label-free cell imaging provides information about endogenous autofluorescent metabolites, enzymes and cofactors in cells. However extracting high content information from autofluorescence imaging has been hitherto impossible. Here, we quantitatively characterise cell populations in different tissue types, live or fixed, by using novel image processing and a simple multispectral upgrade of a wide-field fluorescence microscope. Our optimal discrimination approach enables statistical hypothesis testing and intuitive visualisations where previously undetectable differences become clearly apparent. Label-free classifications are validated by the analysis of Classification Determinant (CD) antigen expression. The versatility of our method is illustrated by detecting genetic mutations in cancer, non-invasive monitoring of CD90 expression, label-free tracking of stem cell differentiation, identifying stem cell subpopulations with varying functional characteristics, tissue diagnostics in diabetes, and assessing the condition of preimplantation embryos.
Collapse
Affiliation(s)
- Martin E. Gosnell
- Quantitative Pty Ltd ABN 17165684186, Beaumont Hills NSW 2155, Australia.
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, North Ryde 2109, NSW Australia
| | - Ayad G. Anwer
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, North Ryde 2109, NSW Australia
| | - Saabah B. Mahbub
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, North Ryde 2109, NSW Australia
| | - Sandeep Menon Perinchery
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, North Ryde 2109, NSW Australia
| | - David W. Inglis
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, North Ryde 2109, NSW Australia
| | - Partho P. Adhikary
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Jalal A. Jazayeri
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Michael A. Cahill
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2678, Australia
| | - Sonia Saad
- Kolling Institute of Medical Research, Royal North Shore Hospital/Northern Clinical School, University of Sydney, Pacific Hwy, St Leonards NSW 2065, Australia
| | - Carol A. Pollock
- Kolling Institute of Medical Research, Royal North Shore Hospital/Northern Clinical School, University of Sydney, Pacific Hwy, St Leonards NSW 2065, Australia
| | - Melanie L. Sutton-McDowall
- Robinson Research Institute, School of Paediatrics and Reproductive Health, The University of Adelaide, Medical School, Frome Road, Adelaide, South Australia, 5005, Australia
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics and Institute for Photonics and Advanced Sensing, The University of Adelaide, North Terrace, Adelaide, South Australia, 5005, Australia
| | - Jeremy G. Thompson
- Robinson Research Institute, School of Paediatrics and Reproductive Health, The University of Adelaide, Medical School, Frome Road, Adelaide, South Australia, 5005, Australia
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics and Institute for Photonics and Advanced Sensing, The University of Adelaide, North Terrace, Adelaide, South Australia, 5005, Australia
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale Biophotonics, Macquarie University, North Ryde 2109, NSW Australia
| |
Collapse
|
39
|
Lee SE, Moon JJM, Kim EY, Park SP. Stem Cell-Derived Bioactive Materials Accelerate Development of Porcine In Vitro-Fertilized Embryos. Cell Reprogram 2016; 17:181-90. [PMID: 26053518 DOI: 10.1089/cell.2014.0110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stem cells show the capability to proliferate in an undifferentiated state with long-term self-renewal, which gives the cells advantages for use as bioactive material (BM) for embryo culture in vitro. The objective of this experiment was to investigate the effect of two BMs-human adipose tissue-derived mesenchymal stem cell BM (hAT-MSC-BM) and human embryonic stem cell-derived BM (hESC-BM)-on porcine embryo development compared to commonly used bovine serum albumin (BSA) or serum treatment groups. In vitro-fertilized (IVF) embryos were cultured in PZM-5 with 4 mg/mL BSA until day 4 and equally divided into four groups. Starting from day 4 (until day 6), each group was treated with the following protein additives: 4 mg/mL BSA (control), 10% fetal bovine serum (FBS), 10% hAT-MSC-BM, or 10% hESC-BM. Our results show FBS- and two other BM-treated groups showed significant increases in blastocyst formation rate, hatching rate, and total cell number compared with the control group (p<0.05). The hAT-MSC-BM and hESC-BM treatment groups presented better-quality embryo development, especially from the middle expanding stage to hatching. In particular, the hAT-MSC-BM-treated group showed the highest developmental potential of all groups and formed the most expanding-stage blastocysts. The relative expression of reprogramming-related transcription factor (POU5F1, SOX2, DPPA5, and CDH1), antioxidant (PRDX5), and apoptosis (BCL2L1 and BIRC5) genes also increased in two types of BMs compared to the control. In addition, we investigated the protein synthesis of the tight junction- and gap junction-related genes, connexin 43 and zonula occludens-1 (ZO-1); these increased more than in the control. These results demonstrate that stem cell-derived BMs accelerate porcine preimplantation embryo development and that the BMs would be helpful in the development of preimplantation embryos.
Collapse
Affiliation(s)
- Seung-Eun Lee
- 1 Stem Cell Research Center, Jeju National University , Jeju Special Self-Governing Province 690-756, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea
| | - Jeremiah Ji-Man Moon
- 2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea
| | - Eun-Young Kim
- 1 Stem Cell Research Center, Jeju National University , Jeju Special Self-Governing Province 690-756, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea.,3 Mirae Cell Bio , Seoul 143-854, Korea
| | - Se-Pill Park
- 1 Stem Cell Research Center, Jeju National University , Jeju Special Self-Governing Province 690-756, Korea.,2 Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University , Jeju Special Self-Governing Province 690-781, Korea.,3 Mirae Cell Bio , Seoul 143-854, Korea
| |
Collapse
|
40
|
Freiman A, Shandalov Y, Rozenfeld D, Shor E, Segal S, Ben-David D, Meretzki S, Egozi D, Levenberg S. Adipose-derived endothelial and mesenchymal stem cells enhance vascular network formation on three-dimensional constructs in vitro. Stem Cell Res Ther 2016; 7:5. [PMID: 26753517 PMCID: PMC4709933 DOI: 10.1186/s13287-015-0251-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/23/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Background Adipose-derived mesenchymal stem cells (MSCs) have been gaining fame mainly due to their vast clinical potential, simple isolation methods and minimal donor site morbidity. Adipose-derived MSCs and microvascular endothelial cells have been shown to bear angiogenic and vasculogenic capabilities. We hypothesized that co-culture of human adipose-derived MSCs with human adipose-derived microvascular endothelial cells (HAMECs) will serve as an effective cell pair to induce angiogenesis and vessel-like network formation in three-dimensional scaffolds in vitro. Methods HAMECs or human umbilical vein endothelial cells (HUVECs) were co-cultured on scaffolds with either MSCs or human neonatal dermal fibroblasts. Cells were immunofluorescently stained within the scaffolds at different time points post-seeding. Various analyses were performed to determine vessel length, complexity and degree of maturity. Results The HAMEC:MSC combination yielded the most organized and complex vascular elements within scaffolds, and in the shortest period of time, when compared to the other tested cell combinations. These differences were manifested by higher network complexity, more tube alignment and higher α-smooth muscle actin expression. Moreover, these generated microvessels further matured and developed during the 14-day incubation period within the three-dimensional microenvironment. Conclusions These data demonstrate optimal vascular network formation upon co-culture of microvascular endothelial cells and adipose-derived MSCs in vitro and constitute a significant step in appreciation of the potential of microvascular endothelial cells and MSCs in different tissue engineering applications that can also be advantageous in in vivo studies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0251-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alina Freiman
- Inter-departmental Program in Biotechnology, Technion-Israel Institute of Technology, Haifa, 32000, Israel. .,Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Yulia Shandalov
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Dekel Rozenfeld
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Erez Shor
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Sofia Segal
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | | | | | - Dana Egozi
- Department of Plastic and Reconstructive Surgery, Kaplan Hospital, Rehovot, Israel.
| | - Shulamit Levenberg
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
41
|
Alkaline phosphatase expression/activity and multilineage differentiation potential are the differences between fibroblasts and orbital fat-derived stem cells--a study in animal serum-free culture conditions. Stem Cell Rev Rep 2015; 10:697-711. [PMID: 24913281 DOI: 10.1007/s12015-014-9529-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human orbital fat tissues are a potential source to isolate stem cells for the development of regenerative medicine therapies. For future safe clinical application of these cells, it is critical to establish animal component-free culture conditions as well as to clearly define the stem cell population characteristics differentiating them from other cell types, such as fibroblasts. Therefore, the present study aimed to compare phenotypic and functional characteristics of orbital fat-derived stem cells (OFSCs) and fibroblasts resident in the eyelid skin in donor-matched samples grown in culture medium supplemented with pooled allogeneic human serum (HS) replacing fetal bovine serum (FBS). We first investigated the proliferative effects of OFSCs on HS, and then we compared the alkaline phosphatase (AP) expression and activity, immunophenotypic profile, and in vitro multilineage differentiation potential of OFSCs side-by-side with fibroblasts. The results showed that HS enhanced OFSCs proliferation without compromising their immunophenotype, AP activity, and osteogenic, adipogenic, and chondrogenic differentiation capacities. In contrast to OFSCs, the fibroblasts did not exhibit AP expression and activity and did not have multilineage differentiation potential. The results enabled us to successfully distinguish OFSCs from fibroblasts populations, suggesting that AP expression/activity and multilineage differentiation assays can be used reliably to discriminate mesenchymal stem cells from fibroblasts. Our findings also support the feasibility of pooled allogeneic HS as a safer and more effective alternative to FBS for clinical applications.
Collapse
|
42
|
Zych J, Spangenberg L, Stimamiglio MA, Abud APR, Shigunov P, Marchini FK, Kuligovski C, Cofré AR, Schittini AV, Aguiar AM, Senegaglia A, Brofman PRS, Goldenberg S, Dallagiovanna B, Naya H, Correa A. Polysome profiling shows the identity of human adipose-derived stromal/stem cells in detail and clearly distinguishes them from dermal fibroblasts. Stem Cells Dev 2014; 23:2791-802. [PMID: 25068904 DOI: 10.1089/scd.2013.0496] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Although fibroblasts and multipotent stromal/stem cells, including adipose-derived stromal cells (ADSCs), have been extensively studied, they cannot be clearly distinguished from each other. We, therefore, investigated the cellular and molecular characteristics of ADSCs and fibroblasts. ADSCs and fibroblasts share several morphological similarities and surface markers, but were clearly found to be different types of cells. Contrary to previous reports, fibroblasts were not able to differentiate into adipocytes, osteoblasts, or chondrocytes. Polysome-bound mRNA profiling revealed that ∼ 1,547 genes were differentially expressed (DE) in the two cell types; the genes were related to cell adhesion, the extracellular matrix, differentiation, and proliferation. These findings were confirmed by functional analyses showing that ADSCs had a greater adhesion capacity than fibroblasts; the proliferation rate of fibroblasts was also higher than that of ADSCs. Importantly, 185 DE genes were integral to the plasma membrane and, thus, candidate markers for ADSC isolation and manipulation. We also observed that an established marker of fibroblasts and ADSCs, CD105, was overexpressed in ADSCs at both mRNA and protein levels. CD105 expression seemed to be related to differentiation capacity, at least for adipogenesis. This study shows that ADSCs and fibroblasts are distinct cell types. These findings should be taken into account when using these two cell types in basic and therapeutic studies.
Collapse
Affiliation(s)
- Jaiesa Zych
- 1 Instituto Carlos Chagas , Fiocruz-Paraná, Curitiba, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|