1
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
2
|
Kaur H, Kaur G, Ali SA. Postbiotics Implication in the Microbiota-Host Intestinal Epithelial Cells Mutualism. Probiotics Antimicrob Proteins 2024; 16:443-458. [PMID: 36933160 DOI: 10.1007/s12602-023-10062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2023] [Indexed: 03/19/2023]
Abstract
To sustain host health and provide the microbial community with a nutrient-rich environment, the host and gut microbiota must interact with one another. These interactions between commensal bacterial and intestinal epithelial cells (IECs) serve as the first line of defense against gut microbiota in preserving intestinal homeostasis. In this microenvironment, the post-biotics and similar molecules such as p40 exert several beneficial effects through regulation of IECs. Importantly, post-biotics were discovered to be transactivators of the EGF receptor (EGFR) in IECs, inducing protective cellular responses and alleviating colitis. The transient exposure to post-biotics such as p40 during the neonatal period reprograms IECs by upregulation of a methyltransferase, Setd1β, leading to a sustained increase in TGF- β release for the expansion of regulatory T cells (Tregs) in the intestinal lamina propria and durable protection against colitis in adulthood. This crosstalk between the IECs and post-biotic secreted factors was not reviewed previously. Therefore, this review describes the role of probiotic-derived factors in the sustainability of intestinal health and improving gut homeostasis via certain signaling pathways. In the era of precision medicine and targeted therapies, more basic, preclinical, and clinical evidence is needed to clarify the efficacy of probiotics released as functional factors in maintaining intestinal health and preventing and treating disease.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, Karnal, 132001, India
| | - Gurjeet Kaur
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
- Mark Wainwright Analytical Centre, Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal, 132001, India.
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, 69120, Germany.
| |
Collapse
|
3
|
Ku H, Chen JJY, Chen W, Tien PT, Lin HJ, Wan L, Xu G. The role of transforming growth factor beta in myopia development. Mol Immunol 2024; 167:34-42. [PMID: 38340674 DOI: 10.1016/j.molimm.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/28/2023] [Accepted: 01/18/2024] [Indexed: 02/12/2024]
Abstract
Myopia is widely recognized as an epidemic. Studies have found a link between Transforming Growth Factor-beta (TGF-β) and myopia, but the specific molecular mechanisms are not fully understood. In this study, a monocular model in tree shrews (Tupaia belangeri) was established to verify the molecular mechanism of TGF-β in myopia. The results indicated that there were significant changes in TGF-βs during the treatment of myopia, which could enhance the refractive ability and axial length of the eye. Immunohistochemical staining, real-time fluorescent quantitative PCR, and immunoblotting results showed a significant upregulation of MMP2 and NF-κB levels, and a significant downregulation of COL-I expression in the TGF-β treated eyes, suggesting that NF-κB and MMP2 are involved in the signaling pathways of TGF-βs induced myopia and axial elongation. Moreover, the expression levels of IL-6, IL-8, MCP-1, IL-1β, TNF-α, TAK1, and NF-κB in the retina were all significantly elevated. This indicates that TGF-β stimulates the inflammatory response of retinal pigment epithelial cells through the TAK1-NF-κB signaling pathway. In conclusion, this study suggests that TGF-β promotes the progression of myopia by enhancing intraocular inflammation.
Collapse
Affiliation(s)
- Hsiangyu Ku
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031 China; Department of Pediatric Ophthalmology, Affiliated Hospital of Yunnan University, China
| | | | - Wei Chen
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031 China
| | - Peng-Tai Tien
- Eye Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hui-Ju Lin
- Eye Center, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lei Wan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan; Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan.
| | - Gezhi Xu
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031 China.
| |
Collapse
|
4
|
Golmohammadi M, Zamanian MY, Jalal SM, Noraldeen SAM, Ramírez‐Coronel AA, Oudaha KH, Obaid RF, Almulla AF, Bazmandegan G, Kamiab Z. A comprehensive review on Ellagic acid in breast cancer treatment: From cellular effects to molecular mechanisms of action. Food Sci Nutr 2023; 11:7458-7468. [PMID: 38107139 PMCID: PMC10724635 DOI: 10.1002/fsn3.3699] [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: 05/17/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 10/16/2023] Open
Abstract
Globally, breast cancer (BC) is the leading cause of cancer-related deaths in women. Hence, developing a therapeutic plan to overcome the disease is crucial. Numerous factors such as endogenous hormones and environmental factors may play a role in the pathophysiology of BC. Regarding the multi-modality treatment of BC, natural compounds like ellagic acid (EA) received has received increased interest in antitumor efficacy with lower adverse effects. Based on the results of this comprehensive review, EA has multiple effects on BC cells including (1) suppresses the growth of BC cells by arresting the cell cycle in the G0/G1 phase, (2) suppresses migration, invasion, and metastatic, (3) stimulates apoptosis in MCF-7 cells via TGF-β/Smad3 signaling axis, (4) inhibits CDK6 that is important in cell cycle regulation, (5) binds to ACTN4 and induces its degradation via the ubiquitin-proteasome pathway, inducing decreased cell motility and invasion in BC cells, (6) inhibits the PI3K/AKT pathway, and (7) inhibits angiogenesis-associated activities including proliferation (reduces VEGFR-2 tyrosine kinase activity). In conclusion, EA exhibits anticancer activity through various molecular mechanisms that influence key cellular processes like apoptosis, cell cycle, angiogenesis, and metastasis in BC. However, further researches are essential to fully elucidate its molecular targets and implications for clinical applications.
Collapse
Affiliation(s)
| | - Mohammad Yasin Zamanian
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | | | | | - Andrés Alexis Ramírez‐Coronel
- Research Group in Educational StatisticsNational University of Education (UNAE)AzoguesEcuador
- Epidemiology and Biostatistics Research GroupCES UniversityMedellínColombia
| | - Khulood H. Oudaha
- Pharmaceutical Chemistry Department, College of PharmacyAl‐Ayen UniversityThi‐OarIraq
| | - Rasha Fadhel Obaid
- Department of Biomedical EngineeringAl‐Mustaqbal University CollegeBabylonIraq
| | - Abbas F. Almulla
- Department of Medical Laboratory Technology, College of Medical TechnologyIslamic UniversityNajafIraq
| | - Gholamreza Bazmandegan
- Physiology‐Pharmacology Research Center, Research Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
- Department of Physiology and Pharmacology, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| | - Zahra Kamiab
- Clinical Research Development Unit, Ali‐Ibn Abi‐Talib HospitalRafsanjan University of Medical SciencesRafsanjanIran
- Department of Community Medicine, School of MedicineRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
5
|
Nasir NJN, Arifin N, Noordin KBA, Yusop N. Bone repair and key signalling pathways for cell-based bone regenerative therapy: A review. J Taibah Univ Med Sci 2023; 18:1350-1363. [PMID: 37305024 PMCID: PMC10248876 DOI: 10.1016/j.jtumed.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Advances in cell-based regenerative therapy create new opportunities for the treatment of bone-related disorders and injuries, by improving the reparative phase of bone healing. Apart from the classical approach of bone grafting, the application of cell-based therapies, particularly stem cells (SCs), has gained a lot of attention in recent years. SCs play an important role in regenerative therapy due to their excellent ability to differentiate into bone-forming cells. Regeneration of new bone is regulated by a wide variety of signalling molecules and intracellular networks, which are responsible for coordinating cellular processes. The activated signalling cascade is significantly involved in cell survival, proliferation, apoptosis, and interaction with the microenvironment and other types of cells within the healing site. Despite the increasing evidence from studies conducted on signalling pathways associated with bone formation, the exact mechanism involved in controlling the differentiation stage of transplanted cells is not well understood. Identifying the key activated pathways involved in bone regeneration may allow for precise manipulation of the relevant signalling molecules within the progenitor cell population to accelerate the healing process. The in-depth knowledge of molecular mechanisms would be advantageous in improving the efficiency of personalised medicine and targeted therapy in regenerative medicine. In this review, we briefly introduce the theory of bone repair mechanism and bone tissue engineering followed by an overview of relevant signalling pathways that have been identified to play an important role in cell-based bone regenerative therapy.
Collapse
Affiliation(s)
- Nur Julia N. Nasir
- Basic and Medical Sciences Department, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Norsyahida Arifin
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang, Malaysia
| | - Khairul Bariah A.A. Noordin
- Basic and Medical Sciences Department, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Norhayati Yusop
- Basic and Medical Sciences Department, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
6
|
Liu GB, Cheng YX, Li HM, Liu Y, Sun LX, Wu Q, Guo SF, Li TT, Dong CL, Sun G. Ghrelin promotes cardiomyocyte differentiation of adipose tissue‑derived mesenchymal stem cells by DDX17‑mediated regulation of the SFRP4/Wnt/β‑catenin axis. Mol Med Rep 2023; 28:164. [PMID: 37449526 PMCID: PMC10407612 DOI: 10.3892/mmr.2023.13050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Adipose tissue‑derived mesenchymal stem cells (ADMSCs) differentiate into cardiomyocytes and may be an ideal cell source for myocardial regenerative medicine. Ghrelin is a gastric‑secreted peptide hormone involved in the multilineage differentiation of MSCs. To the best of our knowledge, however, the role and potential downstream regulatory mechanism of ghrelin in cardiomyocyte differentiation of ADMSCs is still unknown. The mRNA and protein levels were measured by reverse transcription‑quantitative PCR and western blotting. Immunofluorescence staining was used to show the expression and cellular localization of cardiomyocyte markers and β‑catenin. RNA sequencing was used to explore the differentially expressed genes (DEGs) that regulated by ghrelin. The present study found that ghrelin promoted cardiomyocyte differentiation of ADMSCs in a concentration‑dependent manner, as shown by increased levels of cardiomyocyte markers GATA binding protein 4, α‑myosin heavy chain (α‑MHC), ISL LIM homeobox 1, NK2 homeobox 5 and troponin T2, cardiac type. Ghrelin increased β‑catenin accumulation in nucleus and decreased the protein expression of secreted frizzled‑related protein 4 (SFRP4), an inhibitor of Wnt signaling. RNA sequencing was used to determine the DEGs regulated by ghrelin. Functional enrichment showed that DEGs were more enriched in cardiomyocyte differentiation‑associated terms and Wnt pathways. Dead‑box helicase 17 (DDX17), an upregulated DEG, showed enhanced mRNA and protein expression levels following ghrelin addition. Overexpression of DDX17 promoted protein expression of cardiac‑specific markers and β‑catenin and enhanced the fluorescence intensity of α‑MHC and β‑catenin. DDX17 upregulation inhibited protein expression of SFRP4. Rescue assay confirmed that the addition of SFRP4 partially reversed ghrelin‑enhanced protein levels of cardiac‑specific markers and the fluorescence intensity of α‑MHC. In conclusion, ghrelin promoted cardiomyocyte differentiation of ADMSCs by DDX17‑mediated regulation of the SFRP4/Wnt/β‑catenin axis.
Collapse
Affiliation(s)
- Gui-Bo Liu
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Yong-Xia Cheng
- Department of Pathology, The First Clinical Medical School of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Hua-Min Li
- Department of Pathology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, Heilongjiang 157000, P.R. China
| | - Yong Liu
- Department of Research Platform, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Li-Xin Sun
- Office of Educational Administration, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Qi Wu
- Department of Pathology, The First Clinical Medical School of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Shang-Fu Guo
- Department of Asset Management, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Ting-Ting Li
- Department of Pathology, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Chuan-Ling Dong
- Department of Anatomy, School of Basic Medical Sciences, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, P.R. China
| | - Ge Sun
- Department of Orthopedics, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157010, P.R. China
| |
Collapse
|
7
|
Bianconi S, Oliveira KMC, Klein KL, Wolf J, Schaible A, Schröder K, Barker J, Marzi I, Leppik L, Henrich D. Pretreatment of Mesenchymal Stem Cells with Electrical Stimulation as a Strategy to Improve Bone Tissue Engineering Outcomes. Cells 2023; 12:2151. [PMID: 37681884 PMCID: PMC10487010 DOI: 10.3390/cells12172151] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Electrical stimulation (EStim), whether used alone or in combination with bone tissue engineering (BTE) approaches, has been shown to promote bone healing. In our previous in vitro studies, mesenchymal stem cells (MSCs) were exposed to EStim and a sustained, long-lasting increase in osteogenic activity was observed. Based on these findings, we hypothesized that pretreating MSC with EStim, in 2D or 3D cultures, before using them to treat large bone defects would improve BTE treatments. Critical size femur defects were created in 120 Sprague-Dawley rats and treated with scaffold granules seeded with MSCs that were pre-exposed or not (control group) to EStim 1 h/day for 7 days in 2D (MSCs alone) or 3D culture (MSCs + scaffolds). Bone healing was assessed at 1, 4, and 8 weeks post-surgery. In all groups, the percentage of new bone increased, while fibrous tissue and CD68+ cell count decreased over time. However, these and other healing features, like mineral density, bending stiffness, the amount of new bone and cartilage, and the gene expression of osteogenic markers, did not significantly differ between groups. Based on these findings, it appears that the bone healing environment could counteract the long-term, pro-osteogenic effects of EStim seen in our in vitro studies. Thus, EStim seems to be more effective when administered directly and continuously at the defect site during bone healing, as indicated by our previous studies.
Collapse
Affiliation(s)
- Santiago Bianconi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Karla M. C. Oliveira
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Kari-Leticia Klein
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Jakob Wolf
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Alexander Schaible
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Katrin Schröder
- Vascular Research Centre, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - John Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Goethe University Frankfurt, 60528 Frankfurt am Main, Germany;
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Liudmila Leppik
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany (K.-L.K.); (J.W.); (A.S.); (I.M.); (L.L.); (D.H.)
| |
Collapse
|
8
|
Truong NC, Phan TNM, Huynh NT, Pham KD, Van Pham P. Interferon-Gamma Increases the Immune Modulation of Umbilical Cord-Derived Mesenchymal Stem Cells but Decreases Their Chondrogenic Potential. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023. [PMID: 37291444 DOI: 10.1007/5584_2023_776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
INTRODUCTION The pro-inflammatory cytokine interferon-gamma (IFN-γ) is reported to be an agent that boosts the immune modulation of mesenchymal stem cells (MSCs). However, the effects of IFN-γ on the chondrogenic potential of treated MSCs have not been evaluated in depth. This study aimed to evaluate the effects of IFN-γ on the immune modulation and chondrogenic potential of human umbilical cord-derived MSCs (hUC-MSCs). METHODS UC-MSCs were isolated and expanded following published protocols. They were characterized as MSCs before their use in further experiments. The UC-MSCs were treated with IFN-γ at 10 ng/mL for 48 h. Changes in phenotype were investigated based on changes in MSC markers, immunomodulatory genes (TGF-β, IL-4, and IDO) for immune modulation, and cartilage-related genes during the induction of differentiation (Col1a2, Col2a1, Sox9, Runx2, and Acan) for chondrogenic potential. RESULTS IFN-γ-treated UC-MSCs maintained MSC markers and exhibited decreased expression of transcriptional regulatory factors in chondrogenesis (Sox9 and Runx2) and the extracellular matrix-specific genes Col1a2 and Acan but not Col2a1 compared to non-treated cells (p < 0.05). Furthermore, the immunomodulatory capability of IFN-γ-treated UC-MSCs was clearly revealed through their increased expression of IDO and IL-4 and decreased expression of TGF-β compared to non-treated cells (p < 0.05). CONCLUSION This study demonstrated that UC-MSCs treated with IFN-γ at 10 ng/mL had reduced expression of chondrocyte-specific genes; however, they maintained multi-lineage differentiation and exhibited immunomodulatory properties.
Collapse
Affiliation(s)
- Nhat Chau Truong
- Stem Cell Institute, University of Science, Ho Chi Minh City, Viet Nam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Thu Ngoc-Minh Phan
- Stem Cell Institute, University of Science, Ho Chi Minh City, Viet Nam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Nhi Thao Huynh
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City, Viet Nam
| | - Khuong Duy Pham
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City, Viet Nam
| | - Phuc Van Pham
- Stem Cell Institute, University of Science, Ho Chi Minh City, Viet Nam.
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Viet Nam.
- Laboratory of Cancer Research, University of Science, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
9
|
Du X, Cai L, Xie J, Zhou X. The role of TGF-beta3 in cartilage development and osteoarthritis. Bone Res 2023; 11:2. [PMID: 36588106 PMCID: PMC9806111 DOI: 10.1038/s41413-022-00239-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/25/2022] [Accepted: 11/03/2022] [Indexed: 01/03/2023] Open
Abstract
Articular cartilage serves as a low-friction, load-bearing tissue without the support with blood vessels, lymphatics and nerves, making its repair a big challenge. Transforming growth factor-beta 3 (TGF-β3), a vital member of the highly conserved TGF-β superfamily, plays a versatile role in cartilage physiology and pathology. TGF-β3 influences the whole life cycle of chondrocytes and mediates a series of cellular responses, including cell survival, proliferation, migration, and differentiation. Since TGF-β3 is involved in maintaining the balance between chondrogenic differentiation and chondrocyte hypertrophy, its regulatory role is especially important to cartilage development. Increased TGF-β3 plays a dual role: in healthy tissues, it can facilitate chondrocyte viability, but in osteoarthritic chondrocytes, it can accelerate the progression of disease. Recently, TGF-β3 has been recognized as a potential therapeutic target for osteoarthritis (OA) owing to its protective effect, which it confers by enhancing the recruitment of autologous mesenchymal stem cells (MSCs) to damaged cartilage. However, the biological mechanism of TGF-β3 action in cartilage development and OA is not well understood. In this review, we systematically summarize recent progress in the research on TGF-β3 in cartilage physiology and pathology, providing up-to-date strategies for cartilage repair and preventive treatment.
Collapse
Affiliation(s)
- Xinmei Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
10
|
Shojaee A, Ejeian F, Parham A, Nasr-Esfahani MH. Optimizing Tenogenic Differentiation of Equine Adipose-Derived Mesenchymal Stem Cells (eq-ASC) Using TGFB3 Along with BMP Antagonists. CELL JOURNAL 2022; 24:370-379. [PMID: 36043405 PMCID: PMC9428478 DOI: 10.22074/cellj.2022.7892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Tendon repair strategies usually are accompanied by pathological mineralization and scar tissue formation that increases the risk of re-injuries. This study aimed to establish an efficient tendon regeneration method simultaneously with a reduced risk of ectopic bone formation. MATERIALS AND METHODS In this experimental study, tenogenic differentiation was induced through transforming growth factor- β3 (TGFB3) treatment in combination with the inhibiting concentrations of bone morphogenetic proteins (BMP) antagonists, gremlin-2 (GREM2), and a Wnt inhibitor, namely sclerostin (SOST). The procedure's efficacy was evaluated using real-time polymerase chain reaction (qPCR) for expression analysis of tenogenic markers and osteochondrogenic marker genes. The expression level of two tenogenic markers, SCX and MKX, was also evaluated by immunocytochemistry. Sirius Red staining was performed to examine the amounts of collagen fibers. Moreover, to investigate the impact of the substrate on tenogenic differentiation, the nanofibrous scaffolds that highly resemble tendon extracellular matrix was employed. RESULTS Aggregated features formed in spontaneous normal culture conditions followed by up-regulation of tenogenic and osteogenic marker genes, including SCX, MKX, COL1A1, RUNX2, and CTNNB1. TGFB3 treatment exaggerated morphological changes and markedly amplified tenogenic differentiation in a shorter period of time. Along with TGFB3 treatment, inhibition of BMPs by GREM2 and SOST delayed migratory events to some extent and dramatically reduced osteo-chondrogenic markers synergistically. Nanofibrous scaffolds increased tenogenic markers while declining the expression of osteo-chondrogenic genes. CONCLUSION These findings revealed an appropriate in vitro potential of spontaneous tenogenic differentiation of eq- ASCs that can be improved by simultaneous activation of TGFB and inhibition of osteoinductive signaling pathways.
Collapse
Affiliation(s)
- Asiyeh Shojaee
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad,
Mashhad, Iran
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Abbas Parham
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad,
Mashhad, Iran,Stem Cell Biology and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad,
Mashhad, Iran,P.O.Box: 9177948974Division of PhysiologyDepartment of Basic SciencesFaculty of Veterinary MedicineFerdowsi
University of MashhadMashhadIranP.O.Box: 8159358686Department of Animal BiotechnologyReproductive Biomedicine Research CenterRoyan Institute for BiotechnologyACECRIsfahanIran
Emails:,
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran,P.O.Box: 9177948974Division of PhysiologyDepartment of Basic SciencesFaculty of Veterinary MedicineFerdowsi
University of MashhadMashhadIranP.O.Box: 8159358686Department of Animal BiotechnologyReproductive Biomedicine Research CenterRoyan Institute for BiotechnologyACECRIsfahanIran
Emails:,
| |
Collapse
|
11
|
Jankauskaite L, Malinauskas M, Aukstikalne L, Dabasinskaite L, Rimkunas A, Mickevicius T, Pockevičius A, Krugly E, Martuzevicius D, Ciuzas D, Baniukaitiene O, Usas A. Functionalized Electrospun Scaffold-Human-Muscle-Derived Stem Cell Construct Promotes In Vivo Neocartilage Formation. Polymers (Basel) 2022; 14:polym14122498. [PMID: 35746068 PMCID: PMC9229929 DOI: 10.3390/polym14122498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 02/05/2023] Open
Abstract
Polycaprolactone (PCL) is a non-cytotoxic, completely biodegradable biomaterial, ideal for cartilage tissue engineering. Despite drawbacks such as low hydrophilicity and lack of functional groups necessary for incorporating growth factors, it provides a proper environment for different cells, including stem cells. In our study, we aimed to improve properties of scaffolds for better cell adherence and cartilage regeneration. Thus, electrospun PCL–scaffolds were functionalized with ozone and loaded with TGF-β3. Together, human-muscle-derived stem cells (hMDSCs) were isolated and assessed for their phenotype and potential to differentiate into specific lineages. Then, hMDSCs were seeded on ozonated (O) and non-ozonated (“naïve” (NO)) scaffolds with or without protein and submitted for in vitro and in vivo experiments. In vitro studies showed that hMDSC and control cells (human chondrocyte) could be tracked for at least 14 days. We observed better proliferation of hMDSCs in O scaffolds compared to NO scaffolds from day 7 to day 28. Protein analysis revealed slightly higher expression of type II collagen (Coll2) on O scaffolds compared to NO on days 21 and 28. We detected more pronounced formation of glycosaminoglycans in the O scaffolds containing TGF-β3 and hMDSC compared to NO and scaffolds without TGF-β3 in in vivo animal experiments. Coll2-positive extracellular matrix was observed within O and NO scaffolds containing TGF-β3 and hMDSC for up to 8 weeks after implantation. These findings suggest that ozone-treated, TGF-β3-loaded scaffold with hMDSC is a promising tool in neocartilage formation.
Collapse
Affiliation(s)
- Lina Jankauskaite
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
- Correspondence:
| | - Mantas Malinauskas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Lauryna Aukstikalne
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Lauryna Dabasinskaite
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Augustinas Rimkunas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Tomas Mickevicius
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| | - Alius Pockevičius
- Pathology Center, Department of Veterinary Pathobiology, Veterinary Academy, Lithuanian University of Health Sciences, LT-47181 Kaunas, Lithuania;
| | - Edvinas Krugly
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Dainius Martuzevicius
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Darius Ciuzas
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Odeta Baniukaitiene
- Faculty of Chemical Technology, Kaunas University of Technology, LT-44029 Kaunas, Lithuania; (L.D.); (E.K.); (D.M.); (D.C.); (O.B.)
| | - Arvydas Usas
- Institute of Physiology and Pharmacology, Lithuanian University of Health Sciences, LT-49264 Kaunas, Lithuania; (M.M.); (L.A.); (A.R.); (T.M.); (A.U.)
| |
Collapse
|
12
|
Mikuła-Pietrasik J, Rutecki S, Książek K. The functional multipotency of transforming growth factor β signaling at the intersection of senescence and cancer. Cell Mol Life Sci 2022; 79:196. [PMID: 35305149 PMCID: PMC11073081 DOI: 10.1007/s00018-022-04236-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/22/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
The transforming growth factor β (TGF-β) family of cytokines comprises a group of proteins, their receptors, and effector molecules that, in a coordinated manner, modulate a plethora of physiological and pathophysiological processes. TGF-β1 is the best known and plausibly most active representative of this group. It acts as an immunosuppressant, contributes to extracellular matrix remodeling, and stimulates tissue fibrosis, differentiation, angiogenesis, and epithelial-mesenchymal transition. In recent years, this cytokine has been established as a vital regulator of organismal aging and cellular senescence. Finally, the role of TGF-β1 in cancer progression is no longer in question. Because this protein is involved in so many, often overlapping phenomena, the question arises whether it can be considered a molecular bridge linking some of these phenomena together and governing their reciprocal interactions. In this study, we reviewed the literature from the perspective of the role of various TGF-β family members as regulators of a complex mutual interplay between senescence and cancer. These aspects are then considered in a broader context of remaining TGF-β-related functions and coexisting processes. The main narrative axis in this work is centered around the interaction between the senescence of normal peritoneal cells and ovarian cancer cells. The discussion also includes examples of TGF-β activity at the interface of other normal and cancer cell types.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland
| | - Szymon Rutecki
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Długa ½ Str, Poznań University of Medical Sciences, 61-848, Poznań, Poland.
| |
Collapse
|
13
|
Inhibition of Autophagy Flux Promotes Secretion of Chondroitin Sulfate Proteoglycans in Primary Rat Astrocytes. Mol Neurobiol 2021; 58:6077-6091. [PMID: 34449046 DOI: 10.1007/s12035-021-02533-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
Following spinal cord injury (SCI), reactive astrocytes in the glial scar produce high levels of chondroitin sulfate proteoglycans (CSPGs), which are known to inhibit axonal regeneration. Transforming growth factor beta (TGFβ) is a well-known factor that induces the production of CSPGs, and in this study, we report a novel mechanism underlying TGFβ's effects on CSPG secretion in primary rat astrocytes. We observed increased TGFβ-induced secretion of the CSPGs neurocan and brevican, and this occurred simultaneously with inhibition of autophagy flux. In addition, we show that neurocan and brevican levels are further increased when TGFβ is administered in the presence of an autophagy inhibitor, Bafilomycin-A1, while they are reduced when cells are treated with a concentration of rapamycin that is not sufficient to induce autophagy. These findings suggest that TGFβ mediates its effects on CSPG secretion through autophagy pathways. They also represent a potential new approach to reduce CSPG secretion in vivo by targeting autophagy pathways, which could improve axonal regeneration after SCI.
Collapse
|
14
|
Li F, Xu J, Liu S. Cancer Stem Cells and Neovascularization. Cells 2021; 10:cells10051070. [PMID: 33946480 PMCID: PMC8147173 DOI: 10.3390/cells10051070] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) refer to a subpopulation of cancer cells responsible for tumorigenesis, metastasis, and drug resistance. Increasing evidence suggests that CSC-associated tumor neovascularization partially contributes to the failure of cancer treatment. In this review, we discuss the roles of CSCs on tumor-associated angiogenesis via trans-differentiation or forming the capillary-like vasculogenic mimicry, as well as the roles of CSCs on facilitating endothelial cell-involved angiogenesis to support tumor progression and metastasis. Furthermore, we discuss the underlying regulation mechanisms, including the intrinsic signals of CSCs and the extrinsic signals such as cytokines from the tumor microenvironment. Further research is required to identify and verify some novel targets to develop efficient therapeutic approaches for more efficient cancer treatment through interfering CSC-mediated neovascularization.
Collapse
Affiliation(s)
- Fengkai Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel.: +86-21-34771023
| |
Collapse
|
15
|
Xu L, Willumeit-Römer R, Luthringer-Feyerabend BJC. Mesenchymal Stem Cell and Oxygen Modulate the Cocultured Endothelial Cells in the Presence of Magnesium Degradation Products. ACS APPLIED BIO MATERIALS 2021; 4:2398-2407. [DOI: 10.1021/acsabm.0c01289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lei Xu
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | - Regine Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | | |
Collapse
|
16
|
Tseng SJ, Wu CC, Cheng CH, Lin JC. Studies of surface grafted collagen and transforming growth factor β1 combined with cyclic stretching as a dual chemical and physical stimuli approach for rat adipose-derived stem cells (rADSCs) chondrogenesis differentiation. J Mech Behav Biomed Mater 2020; 112:104062. [PMID: 32891975 DOI: 10.1016/j.jmbbm.2020.104062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
The adipose-derived stem cell has been used in various regenerative medicine research due to its multiple differentiation capabilities. Developing a stable and quick approach for the differentiation of stem cells is a critical issue in tissue regenerative field. In this investigation, rat adipose-derived stem cells (rADSCs) were seeded onto the type I collagen/transforming growth factor β1 (TGF-β1) immobilized polydimethylsiloxane (PDMS) substrate and then combined with short term dynamic stretching stimulation (intermittent or continuous stretching for 6 h) to induce the rADSCs chondrogenesis differentiation using the induction medium without growth factors added in vitro. Via regulating the extracellular chemical- and mechano-receptors of the cultured rADSCs, the chondrogenic differentiation was examined. After 72 h of static culture, proteoglycan secretion was noted on the surfaces modified by collagen with or without TGF-β1. After different stretching stimulations, significant proteoglycan secretion was noted on the surface modified by both collagen and collagen/TGF-β1, especially after the intermittent stretching culturing. Nonetheless, genetic expression of the chondrogenic markers: SOX-9, Col2a1, and aggrecan, instead, were dependent upon the surface grafted layer and the stretching mode utilized. These findings suggested that the surface chemical characteristics and external mechanical stimulation could synergistically affect the efficacy of chondrogenic differentiation of rADSCs.
Collapse
Affiliation(s)
- Shen-Jui Tseng
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Hui Cheng
- Department of Pediatrics, College of Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Jui-Che Lin
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
17
|
Gonçalves BÔP, De Andrade WP, Da Conceição Braga L, Fialho SL, Silva LM. Epithelial-to-mesenchymal transition markers are differentially expressed in epithelial cancer cell lines after everolimus treatment. Oncol Lett 2020; 20:158. [PMID: 32934726 PMCID: PMC7471649 DOI: 10.3892/ol.2020.12019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a phenomenon during which cancer epithelial cells undergo changes in plasticity and lose cell-cell adhesion with consequent remodeling of the extracellular matrix and development of mesenchymal characteristics. Long non-coding RNAs (lncRNAs) have been described as EMT modulation markers, becoming a promising target in the development of new therapies for cancer. The present study aimed to investigate the role of everolimus at 100 nM as inductor of the EMT phenomenon in cell lines derived from human breast (BT-549), colorectal (RKO-AS45-1) and ovary (TOV-21G) cancer. The integrity of cellular junctions was monitored using an in vitro model of epithelial resistance. The results demonstrated that the EMT genes ZEB1, TWIST1 and TGFB1 were differentially expressed in cells treated with everolimus compared with in untreated cells. lncRNA HOTAIR was upregulated post-treatment only in BT-549 cells compared with in untreated cells. After treatment with everolimus, the intensity of fluorescence of P-cadherin decreased, and that of fibronectin increased in RKO-AS45-1 and TOV-21G cells compared with control cells. The transepithelial electrical resistance at the RKO-AS45-1 monolayer treated with everolimus started to decrease at 48 h. The changes in the gene expression and epithelial resistance may confirm the role of everolimus in EMT.
Collapse
Affiliation(s)
- Bryan Ôrtero Perez Gonçalves
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Warne Pedro De Andrade
- Hematology and Oncology Nucleus, Grupo Oncoclinicas, Belo Horizonte, Minas Gerais 30140001, Brazil.,Department of Obstetrics and Gynecology, School of Medicine, São Paulo State University, Botucatu, São Paulo 18618687, Brazil
| | - Letícia Da Conceição Braga
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Sílvia Ligório Fialho
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Luciana Maria Silva
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| |
Collapse
|
18
|
Gonçalves BÔP, Fialho SL, Silvestrini BR, Sena IFG, Dos Santos GSP, Assis Gomes D, Silva LM. Central nervous system (CNS) tumor cell heterogeneity contributes to differential platinum-based response in an in vitro 2D and 3D cell culture approach. Exp Mol Pathol 2020; 116:104520. [PMID: 32828740 DOI: 10.1016/j.yexmp.2020.104520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022]
Abstract
One of the models that best explains the cellular heterogeneity observed in central nervous system (CNS) tumors is the presence of cancer stem cells (CSCs). CSCs can originate from differentiated adult cells that return to an undifferentiated stage through the mechanism known as epithelial-mesenchymal transition (EMT). In this paper, we evaluated cellular and molecular heterogeneity and the participation of the epithelial-mesenchymal transition (EMT) in glioblastoma (U-87 MG and LN-18) and neuroblastoma (KELLY and IMR-32) cell lines cultured in monolayer (2D) and neurosphere (CSC enrichment- 3D) models. For this, after treatment with cisplatin, we studied different cell subpopulations by immunophenotyping using neural stem cell/progenitor markers (ALDH, CD24, CD56, and CD133), mesenchymal stem cell markers (CD73, CD90, CD105, and CD146) and hematopoietic markers (CD14, CD19, CD34, CD45, and HLA-DR) and mRNA expression profiles of genes related to EMT, such as ZEB1, TWIST1, TGFB1, STAT3, and lncRNA HOTAIR. In addition, we evaluated the growth capacity of residual cells when treated with cisplatin using the chorioallantoic membrane (CAM) model to study disease relapse. After treatment with cisplatin, we found that the expression of STAT3 and TGFB1 genes markedly increased in the neurosphere of the IMR-32 cell line, and TWIST1 was upregulated in the neurosphere of LN-18. Only the nontreated monolayer of LN-18, KELLY, and IMR-32 amplified the lncRNA HOTAIR. The IMR-32 cell line exhibited an enrichment of CD24+/ALDH+ and this cell subset decreased after cisplatin treatment. We observed the loss of CD146+/CD73+ cell subpopulations in U-87 MG monolayer and neurosphere models, after cisplatin treatment, while in LN-18 monolayer cisplatin-treated cells, CD73+/CD90+ cell subpopulations increased. Neuroblastoma cell lines showed CD14+/HLA-DR- cell subpopulations representative of myeloid-derived suppressor cells (MDSCs). Tumors generated from residual cells, after exposure to cisplatin, grafted on CAM showed patterns of organization different from those of the controls. Thus, our findings strongly supported the idea that definitions of tumor phenotypic characteristics may help to establish better therapeutic strategies for the development of new drug targets.
Collapse
Affiliation(s)
- Bryan Ôrtero Perez Gonçalves
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil; Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Sílvia Ligório Fialho
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Bárbara Reis Silvestrini
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | | | | | - Dawidson Assis Gomes
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Luciana Maria Silva
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil.
| |
Collapse
|
19
|
Brooks AES, Iminitoff M, Williams E, Damani T, Jackson-Patel V, Fan V, James J, Dunbar PR, Feisst V, Sheppard HM. Ex Vivo Human Adipose Tissue Derived Mesenchymal Stromal Cells (ASC) Are a Heterogeneous Population That Demonstrate Rapid Culture-Induced Changes. Front Pharmacol 2020; 10:1695. [PMID: 32153389 PMCID: PMC7044177 DOI: 10.3389/fphar.2019.01695] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022] Open
Abstract
Human adipose-derived mesenchymal stromal cells (ASC) are showing clinical promise for the treatment of a range of inflammatory and degenerative conditions. These lipoaspirate-derived cells are part of the abundant and accessible source of heterogeneous stromal vascular fraction (SVF). They are typically isolated and expanded from the SVF via adherent cell culture for at least 2 weeks and as such represent a relatively undefined population of cells. We isolated ex vivo ASC directly from lipoaspirate using a cocktail of antibodies combined with immunomagnetic bead sorting. This method allowed for the rapid enrichment of a defined and untouched ex vivo ASC population (referred to as MACS-derived ASC) that were then compared to culture-derived ASC. This comparison found that MACS-derived ASC contain a greater proportion of cells with activity in in vitro differentiation assays. There were also significant differences in the secretion levels of some key paracrine molecules. Moreover, when the MACS-derived ASC were subjected to adherent tissue culture, rapid changes in gene expression were observed. This indicates that culturing cells may alter the clinical utility of these cells. Although MACS-derived ASC are more defined compared to culture-derived ASC, further investigations using a comprehensive multicolor flow cytometry panel revealed that this cell population is more heterogeneous than previously appreciated. Additional studies are therefore required to more precisely delineate phenotypically distinct ASC subsets with the most therapeutic potential. This research highlights the disparity between ex vivo MACS-derived and culture-derived ASC and the need for further characterization.
Collapse
Affiliation(s)
- Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Megan Iminitoff
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Eloise Williams
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Tanvi Damani
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | - Vicky Fan
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna James
- Department of Obstetrics and Gynecology, University of Auckland, Auckland, New Zealand
| | - P Rod Dunbar
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Vaughan Feisst
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Hilary M Sheppard
- School of Biological Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
El-Houseini ME, Ismail A, Abdelaal AA, El-Habashy AH, Abdallah ZF, Mohamed MZ, El-Hadidi M, Cho WCS, Ahmed H, Al-Shafie TA. Role of TGF-β1 and C-Kit Mutations in the Development of Hepatocellular Carcinoma in Hepatitis C Virus-Infected Patients: in vitro Study. BIOCHEMISTRY (MOSCOW) 2019; 84:941-953. [PMID: 31522676 DOI: 10.1134/s0006297919080108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Transforming growth factor beta (TGF-β) acts as a tumor-suppressing cytokine in healthy tissues and non-malignant tumors. Yet, in malignancy, TGF-β can exert the opposite effects that can promote proliferation of cancer cells. C-Kit plays a prominent role in stem cell activation and liver regeneration after injury. However, little is known about the cross-talk between TGF-β and C-Kit and its role in the progression of hepatocellular carcinoma (HCC). Here, we studied the effect of increasing doses of TGF-β1 on CD44+CD90+ liver stem cells (LSCs) and C-Kit gene expression in malignant and adjacent non-malignant liver tissues excised from 32 HCC patients. The percentage of LSCs in malignant tumors was two times higher compared to their counterparts from the non-malignant tissues. When treated with increasing doses of TGF-β1, proliferation of both malignant and non-malignant LSCs was progressively suppressed, but low TGF-β1 dose failed to suppress the growth of malignant LSCs. Moreover, C-Kit exons 9 and 11 were expressed in malignant LSCs, but not in their non-malignant counterparts. Analysis of C-Kit detected mutations in exon 9 (but not in exon 11) in some malignant liver cells resulting in the changes in the amino acid sequence and dysregulation of protein structure and function. Interestingly, in malignant liver cells, mutations in exon 9 were associated with high-viremia hepatitis C virus (HCV), and expression of this exon was not suppressed by the TGF-β1 treatment at all doses. To our knowledge, this is the first report that mutations in the C-Kit gene in HCC patients are associated with high- viremia HCV. Our study emphasizes the need for investigation of the TGF-β1 level and C-Kit mutations in patients with chronic HCV for HCC prevention and better therapy management.
Collapse
Affiliation(s)
- M E El-Houseini
- Cairo University, National Cancer Institute, Department of Cancer Biology, Cairo, 11796, Egypt
| | - A Ismail
- Ain Shams University, Faculty of Medicine, Department of Surgery, Cairo, 11566, Egypt
| | - A A Abdelaal
- Ain Shams University, Faculty of Medicine, Department of Surgery, Cairo, 11566, Egypt
| | - A H El-Habashy
- Cairo University, National Cancer Institute, Department of Pathology, Cairo, 11796, Egypt
| | - Z F Abdallah
- Cairo University, National Cancer Institute, Department of Cancer Biology, Cairo, 11796, Egypt
| | - M Z Mohamed
- Medical Center of Egyptian Railways, Department of Medical Laboratory, Cairo, 11669, Egypt
| | - M El-Hadidi
- Nile University, Center of Informatics Science, Giza, 12525, Egypt
| | - W C S Cho
- Queen Elizabeth Hospital, Department of Clinical Oncology, Kowloon, Hong Kong, China
| | - H Ahmed
- GlycoMantra, Inc., Baltimore, MD 21227, USA
| | - T A Al-Shafie
- Cairo University, National Cancer Institute, Department of Cancer Biology, Cairo, 11796, Egypt. .,Pharos University in Alexandria, Faculty of Pharmacy and Drug Manufacturing, Department of Pharmacology and Therapeutics, Alexandria, 21311, Egypt
| |
Collapse
|
21
|
Gasiūnienė M, Petkus G, Matuzevičius D, Navakauskas D, Navakauskienė R. Angiotensin II and TGF- β1 Induce Alterations in Human Amniotic Fluid-Derived Mesenchymal Stem Cells Leading to Cardiomyogenic Differentiation Initiation. Int J Stem Cells 2019; 12:251-264. [PMID: 31023001 PMCID: PMC6657950 DOI: 10.15283/ijsc18126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Background and Objectives Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) may be a valuable source for cardiovascular tissue engineering and cell therapy. The aim of this study is to verify angiotensin II and transforming growth factor-beta 1 (TGF-β1) as potential cardiomyogenic differentiation inducers of AF-MSCs. Methods and Results AF-MSCs were obtained from amniocentesis samples from second-trimester pregnant women, isolated and characterized by the expression of cell surface markers (CD44, CD90, CD105 positive; CD34 negative) and pluripotency genes (OCT4, SOX2, NANOG, REX1). Cardiomyogenic differentiation was induced using different concentrations of angiotensin II and TGF-β1. Successful initiation of differentiation was confirmed by alterations in cell morphology, upregulation of cardiac genes-markers NKX2-5, TBX5, GATA4, MYH6, TNNT2, DES and main cardiac ion channels genes (sodium, calcium, potassium) as determined by RT-qPCR. Western blot and immunofluorescence analysis revealed the increased expression of Connexin43, the main component of gap junctions, and Nkx2.5, the early cardiac transcription factor. Induced AF-MSCs switched their phenotype towards more energetic and started utilizing oxidative phosphorylation more than glycolysis for energy production as assessed using Agilent Seahorse XF analyzer. The immune analysis of chromatin-modifying enzymes DNMT1, HDAC1/2 and Polycomb repressive complex 1 and 2 (PRC1/2) proteins BMI1, EZH2 and SUZ12 as well as of modified histones H3 and H4 indicated global chromatin remodeling during the induced differentiation. Conclusions Angiotensin II and TGF-β1 are efficient cardiomyogenic inducers of human AF-MSCs; they initiate alterations at the gene and protein expression, metabolic and epigenetic levels in stem cells leading towards cardiomyocyte- like phenotype formation.
Collapse
Affiliation(s)
- Monika Gasiūnienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gintautas Petkus
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dalius Matuzevičius
- Electronic Systems Department, Electronics Faculty, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Dalius Navakauskas
- Electronic Systems Department, Electronics Faculty, Vilnius Gediminas Technical University, Vilnius, Lithuania
| | - Rūta Navakauskienė
- Department of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
22
|
Shojaee A, Parham A, Ejeian F, Nasr Esfahani MH. Equine adipose mesenchymal stem cells (eq-ASCs) appear to have higher potential for migration and musculoskeletal differentiation. Res Vet Sci 2019; 125:235-243. [PMID: 31310927 DOI: 10.1016/j.rvsc.2019.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 05/14/2019] [Accepted: 06/30/2019] [Indexed: 12/28/2022]
Abstract
Equine adipose-derived mesenchymal stem cells (eq-ASCs) possess excellent regeneration potential especially for treatment of musculoskeletal disorders. Besides their common characteristics, MSCs harvested from different species reveal some species-specific and donor-dependent behaviors. Hence, the molecular analysis of MSCs may shed more light on their future clinical application of these cells. This study aimed to investigate some behavioral aspects of eq-ASCs in vitro which may influence the efficacy of stem cell therapy. For this purpose, MSCs of a donor horse were isolated, characterized and expanded under normal culture conditions. During continuous culture condition, eq-ASCs were started to formed aggregated structures that was accompanied with the up-regulation of migratory related genes including transforming growth factor beta 1 (TGFB1) and its receptor 3 (TGFBR3), and snail family transcriptional repressor 1 (SNAI1), E-cadherin (CDH1) and β-catenin (CTNNB1). Moreover, the expression of a musculoskeletal progenitor marker, scleraxis bHLH transcription factor (SCX), was also increased after 3 days. In order to clarify the impact of TGFB signaling pathway on cultured cells, gain- and loss-of-function treatment by TGFB3 and SB431542 (TGFB inhibitor) were performed, respectively. We found that TGFB3 treatment exaggerated the aggregate formation effects, in some extend via induction of cytoskeletal actin rearrangement, while inhibition of TGFB signaling pathway by SB431542 reversed this phenomenon. Overall, our findings support the fact that eq-ASCs have an inherent capacity for migration, which was enhanced by TGFB3 treatment and, this ability may play crucial role in cell motility and wound healing of transplanted cells.
Collapse
Affiliation(s)
- Asiyeh Shojaee
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abbas Parham
- Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell Biology and Alternative Regenerative Medicine Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Fatemeh Ejeian
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
23
|
Li X, Yang H, Zhang Z, Yan Z, Lv H, Zhang Y, Wu B. Platelet‑rich fibrin exudate promotes the proliferation and osteogenic differentiation of human periodontal ligament cells in vitro. Mol Med Rep 2018; 18:4477-4485. [PMID: 30221718 PMCID: PMC6172397 DOI: 10.3892/mmr.2018.9472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/13/2018] [Indexed: 01/09/2023] Open
Abstract
The purpose of the present study was to evaluate the effects of platelet-rich fibrin (PRF) exudate on the proliferation, osteogenic differentiation and mineralization of human periodontal ligament cells (hPDLCs) in vitro. In the present study PRF was obtained with permission, from the peripheral blood of healthy donors and PRF exudates were collected on the 7th day of incubation. hPDLCs were obtained from healthy premolars, cultured by a tissue explant method and identified with anti-vimentin and anti-cytokeratin antibodies. PRF exudates were added to hPDLCs in different concentrations to evaluate cell proliferation and osteogenic differentiation. The proliferation of hPDLCs was measured using a colorimetric assay. Osteogenic differentiation and mineralization were determined by Alizarin red staining, alkaline phosphatase activity (ALP), western blotting and reverse transcription-quantitative polymerase chain reaction. Cell proliferation was enhanced by addition of the PRF exudate, which also promoted the formation of mineralized matrix nodules and upregulated ALP activity and osteoblast-associated levels of osteocalcin, runt-related transcription factor and osterix gene expression. As these stimulatory effects occurred in a dose-dependent manner, it was concluded that high concentrations of the PRF exudate served an essential role in the proliferation, osteogenic differentiation and mineralization of hPDLCs in vitro. The present study demonstrated that PRF exudate enhanced hPDLC proliferation, induced the osteoblastic differentiation of hPDLCs into mineralized tissue-formation cells in vitro, and may therefore provide potential benefits for periodontal tissue engineering; contributing to the primary processes of periodontal tissue regeneration. From the perspective of both economics and biology, PRF has greater clinical benefits than analogous growth factors.
Collapse
Affiliation(s)
- Xiaoju Li
- Department of Stomatology, The People's Hospital of Longhua, Shenzhen, Guangdong 518109, P.R. China
| | - Huixiao Yang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Zijiian Zhang
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Zhonghai Yan
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Huling Lv
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510140, P.R. China
| | - Yan Zhang
- Department of General Therapy Dentistry, Stomatology Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Bin Wu
- Department of Stomatology, The People's Hospital of Longhua, Shenzhen, Guangdong 518109, P.R. China
| |
Collapse
|
24
|
Abstract
Resistance to chemotherapy and cancer relapse are major clinical challenges attributed to a sub population of cancer stem cells (CSCs). The concept of CSCs has been the subject of intense research by the oncology community since evidence for their existence was first published over twenty years ago. Emerging data indicates that they are also able to evade novel therapies such as targeted agents, immunotherapies and anti-angiogenics. The inability to appropriately identify and isolate CSCs is a major hindrance to the field and novel technologies are now being utilized. Agents that target CSC-associated cell surface receptors and signaling pathways have generated promising pre-clinical results and are now entering clinical trial. Here we discuss and evaluate current therapeutic strategies to target CSCs.
Collapse
Affiliation(s)
- Stephanie Annett
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland
| | - Tracy Robson
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland.
| |
Collapse
|
25
|
Deriving vascular smooth muscle cells from mesenchymal stromal cells: Evolving differentiation strategies and current understanding of their mechanisms. Biomaterials 2017; 145:9-22. [PMID: 28843066 DOI: 10.1016/j.biomaterials.2017.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play essential roles in regulating blood vessel form and function. Regeneration of functional vascular smooth muscle tissue to repair vascular diseases is an area of intense research in tissue engineering and regenerative medicine. For functional vascular smooth muscle tissue regeneration to become a practical therapy over the next decade, the field will need to have access to VSMC sources that are effective, robust and safe. While pluripotent stem cells hold good future promise to this end, more immediate translation is expected to come from approaches that generate functional VSMCs from adult sources of multipotent adipose-derived and bone marrow-derived mesenchymal stromal cells (ASCs and BMSCs). The research to this end is extensive and is dominated by studies relating to classical biochemical signalling molecules used to induce differentiation of ASCs and BMSCs. However, prolonged use of the biochemical induction factors is costly and can cause potential endotoxin contamination in the culture. Over recent years several non-traditional differentiation approaches have been devised to mimic defined aspects of the native micro-environment in which VSMCs reside to contribute to the differentiation of VSMC-like cells from ASCs and BMSCs. In this review, the promises and limitations of several non-traditional culture approaches (e.g., co-culture, biomechanical, and biomaterial stimuli) targeting VSMC differentiation are discussed. The extensive crosstalk between the underlying signalling cascades are delineated and put into a translational context. It is expected that this review will not only provide significant insight into VSMC differentiation strategies for vascular smooth muscle tissue engineering applications, but will also highlight the fundamental importance of engineering the cellular microenvironment on multiple scales (with consideration of different combinatorial pathways) in order to direct cell differentiation fate and obtain cells of a desired and stable phenotype. These strategies may ultimately be applied to different sources of stem cells in the future for a range of biomaterial and tissue engineering disciplines.
Collapse
|
26
|
Rao S, Zaidi S, Banerjee J, Jogunoori W, Sebastian R, Mishra B, Nguyen BN, Wu RC, White J, Deng C, Amdur R, Li S, Mishra L. Transforming growth factor-β in liver cancer stem cells and regeneration. Hepatol Commun 2017; 1:477-493. [PMID: 29404474 PMCID: PMC5678904 DOI: 10.1002/hep4.1062] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells have established mechanisms that contribute to tumor heterogeneity as well as resistance to therapy. Over 40% of hepatocellular carcinomas (HCCs) are considered to be clonal and arise from a stem-like/cancer stem cell. Moreover, HCC is the second leading cause of cancer death worldwide, and an improved understanding of cancer stem cells and targeting these in this cancer are urgently needed. Multiple studies have revealed etiological patterns and multiple genes/pathways signifying initiation and progression of HCC; however, unlike the transforming growth factor β (TGF-β) pathway, loss of p53 and/or activation of β-catenin do not spontaneously drive HCC in animal models. Despite many advances in cancer genetics that include identifying the dominant role of TGF-β signaling in gastrointestinal cancers, we have not reached an integrated view of genetic mutations, copy number changes, driver pathways, and animal models that support effective targeted therapies for these common and lethal cancers. Moreover, pathways involved in stem cell transformation into gastrointestinal cancers remain largely undefined. Identifying the key mechanisms and developing models that reflect the human disease can lead to effective new treatment strategies. In this review, we dissect the evidence obtained from mouse and human liver regeneration, and mouse genetics, to provide insight into the role of TGF-β in regulating the cancer stem cell niche. (Hepatology Communications 2017;1:477-493).
Collapse
Affiliation(s)
- Shuyun Rao
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Sobia Zaidi
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Jaideep Banerjee
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Wilma Jogunoori
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Raul Sebastian
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Bibhuti Mishra
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| | - Bao-Ngoc Nguyen
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Ray-Chang Wu
- Department of Biochemistry and Molecular Medicine George Washington University Washington DC
| | - Jon White
- Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| | - Chuxia Deng
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Health Sciences University of Macau Taipa Macau China
| | - Richard Amdur
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Shulin Li
- Department of Pediatrics The University of Texas MD Anderson Cancer Center Houston TX
| | - Lopa Mishra
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| |
Collapse
|
27
|
Yang Y, Nian H, Tang X, Wang X, Liu R. Effects of the combined Herba Epimedii and Fructus Ligustri Lucidi on bone turnover and TGF-β1/Smads pathway in GIOP rats. JOURNAL OF ETHNOPHARMACOLOGY 2017; 201:91-99. [PMID: 28254481 DOI: 10.1016/j.jep.2017.02.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kidney deficiency is the main pathogenesis of osteoporosis based on the theory of "kidney governing bones" in traditional Chinese medicine (TCM). Combined Herba Epimedii and Fructus Ligustri Lucidi, based on traditional Chinese formula Er-Zhi pills, were frequently used in TCM formulas that were prescribed for kidney tonifying and bone strengthening. However, it is unclear whether the combination of the two herbs may have a protective influence on glucocorticoid-induced osteoporosis (GIOP). The objective of this study was to evaluate the therapeutic effects and the underlying molecular mechanism of the decoction and the active fractions of the combined herbs in GIOP rats. MATERIALS AND METHODS Male Sprague-Dawley rats were divided into seven groups, including the normal control (NC), GIOP model (MO), active fractions low (100mg/kg, LAF), active fractions high (200mg/kg, HAF), decoction low (3.5g/kg, LD), decoction high (7g/kg, HD) and Calcium with Vitamin D3 (0.2773g/kg, CaD)-treated group. The GIOP model was established by intramuscular injection of dexamethasone (1mg/kg) twice a week for 8 weeks. Different kinds of indicators were measured, including bone mineral density (BMD), bone biomechanical properties, serum bone alkaline phosphatase (b-ALP), serum bone γ-carboxyglutamic acid-containing protein (BGP), serum bone morphogenetic protein-2 (BMP-2), serum tartrate-resistant acid phosphatase (TRACP) and serum carboxy terminal cross linked telopeptide of typeⅠcollagen (ICTP), bone mineral content (BMC) and bone structured histomorphometry. The protein and mRNA expression of TGF-β1, Smad2, Smad3, Smad4 and Smad7 were detected by Western blotting (WB) and quantitative real time polymerase chain reaction (qRT-PCR), respectively. RESULTS Administration of combined Herba Epimedii and Fructus Ligustri Lucidi decoction and combined active fractions could significantly prevent GC-induced bone loss by increasing the contents of serum b-ALP, BGP and BMP-2 as the markers of bone formation, reducing the serum TRACP and ICTP contents to inhibit bone resorption and enhancing BMC. They could also attenuate biomechanical properties and BMD reduction, deterioration of trabecular architecture in MO rats. The mRNA and protein expressions of TGF-β1, smad2, smad3 and smad4 were up-regulated, and the mRNA and protein expression of Smad7 was down-regulated following combined Herba Epimedii and Fructus Ligustri Lucidi treatment. CONCLUSION Combination of Herba Epimedii and Fructus Ligustri Lucidi exhibited protective effects on promoting bone formation and precluding bone resorption. The underlying mechanism may be attributed to its regulations on TGF-β1/Smads pathway. The substance bases of the combined herbs on anti-osteoporosis were total flavonoids of Herba Epimedii, total iridoids and flavonoids of Fructus Ligustri Lucidi.
Collapse
Affiliation(s)
- Yan Yang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China
| | - Honglei Nian
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China
| | - Xiufeng Tang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China
| | - Xiujuan Wang
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Renhui Liu
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, No.10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| |
Collapse
|
28
|
Turajane T, Chaveewanakorn U, Fongsarun W, Aojanepong J, Papadopoulos KI. Avoidance of Total Knee Arthroplasty in Early Osteoarthritis of the Knee with Intra-Articular Implantation of Autologous Activated Peripheral Blood Stem Cells versus Hyaluronic Acid: A Randomized Controlled Trial with Differential Effects of Growth Factor Addition. Stem Cells Int 2017; 2017:8925132. [PMID: 29056974 PMCID: PMC5625803 DOI: 10.1155/2017/8925132] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/25/2017] [Accepted: 08/24/2017] [Indexed: 02/08/2023] Open
Abstract
In this randomized controlled trial, in early osteoarthritis (OA) that failed conservative intervention, the need for total knee arthroplasty (TKA) and WOMAC scores were evaluated, following a combination of arthroscopic microdrilling mesenchymal cell stimulation (MCS) and repeated intra-articular (IA) autologous activated peripheral blood stem cells (AAPBSCs) with growth factor addition (GFA) and hyaluronic acid (HA) versus IA-HA alone. Leukapheresis-harvested AAPBSCs were administered as three weekly IA injections combined with HA and GFA (platelet-rich plasma [PRP] and granulocyte colony-stimulating factor [hG-CSF]) and MCS in group 1 and in group 2 but without hG-CSF while group 3 received IA-HA alone. Each group of 20 patients was evaluated at baseline and at 1, 6, and, 12 months. At 12 months, all patients in the AAPBSC groups were surgical intervention free compared to three patients needing TKA in group 3 (p < 0.033). Total WOMAC scores showed statistically significant improvements at 6 and 12 months for the AAPBSC groups versus controls. There were no notable adverse events. We have shown avoidance of TKA in the AAPBSC groups at 12 months and potent, early, and sustained symptom alleviation through GFA versus HA alone. Differential effects of hG-CSF were noted with an earlier onset of symptom alleviation throughout.
Collapse
Affiliation(s)
- Thana Turajane
- 1Department of Orthopedic Surgery, Police General Hospital, Bangkok, Thailand
| | | | | | - Jongjate Aojanepong
- 3Department of Gynecology and Obstetrics, Police General Hospital, Bangkok, Thailand
| | | |
Collapse
|
29
|
Leshansky L, Aberdam D, Itskovitz-Eldor J, Berrih-Aknin S. Human embryonic stem cells prevent T-cell activation by suppressing dendritic cells function via TGF-beta signaling pathway. Stem Cells 2015; 32:3137-49. [PMID: 25186014 DOI: 10.1002/stem.1833] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 07/23/2014] [Indexed: 12/25/2022]
Abstract
Human embryonic stem cells (hESCs) represent a potential source of transplantable cells for regenerative medicine, but development of teratoma even in syngenic recipients represents a critical obstacle to safe stem cell-based therapies. We hypothesized that hESCs escape the immune surveillance by regulating the environmental immune system. Using cocultures of hESCs with allogenic peripheral blood mononuclear cells, we demonstrated that hESCs prevent proliferation and activation of human CD4+ T lymphocytes, an effect dependent upon monocytes. Altered expression of key signaling molecules responsible for the crosstalk of monocytes with T cells was detected in the presence of hESCs. Analyzing the mechanism of action, we demonstrated that hESCs were able to downregulate intracellular glutathione levels in both monocytes and CD4+ cells by suppressing glutamate cysteine ligase expression and to alter MHCII and CD80 expression in monocytes. These effects were achieved at least partially via TGF-beta signaling, and both monocyte phenotype and GCLC expression were affected by Caspase-3 proteolytic activity. Altogether, our results demonstrate a novel immune-suppressive mechanism used by hESCs.
Collapse
Affiliation(s)
- Lucy Leshansky
- INSERTECH Stem Cell Center, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Stem Cell Center, Ruth and Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | |
Collapse
|
30
|
Inhibition of Transforming Growth Factor-β Receptor signaling promotes culture expansion of undifferentiated human Endometrial Mesenchymal Stem/stromal Cells. Sci Rep 2015; 5:15042. [PMID: 26461813 PMCID: PMC4602195 DOI: 10.1038/srep15042] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
Human endometrial MSC (eMSC) are a novel source of MSC easily harvested from the highly regenerative uterine lining. We have developed protocols for eMSC isolation from single cell suspensions using magnetic bead-sorting using a perivascular marker antibody to SUSD2 and culture expansion in serum free medium (SFM). Similar to other MSC, eMSC spontaneously differentiate into fibroblasts during culture expansion decreasing their purity and efficacy. The aim of this study was to determine if A83-01, a TGF-β receptor inhibitor prevents eMSC differentiation in culture. SUSD2+ eMSC were cultured in SFM with bFGF/EGF in 5% O2/5% CO2. At passage 6, eMSC were incubated with or without A83-01 for 7 days, then analysed for MSC properties. A83-01 dose dependently promoted SUSD2+ eMSC proliferation and blocked apoptosis via the SMAD 2/3 pathway. Fewer A83-01 treated cells were autofluorescent or stained with β-galactosidase, indicating reduced senescence. A83-01-treated cells had higher cloning efficiency, differentiated into mesodermal lineages and expressed MSC phenotypic markers. These data suggest that A83-01 maintains SUSD2+ eMSC stemness, promoting proliferation by blocking senescence and apoptosis in late passage cultures through binding to TGF-β receptors. Small molecules such as A83-01 may enable the expansion of undifferentiated MSC for use in tissue engineering and cell-based therapies.
Collapse
|
31
|
Downey J, Lauzier D, Kloen P, Klarskov K, Richter M, Hamdy R, Faucheux N, Scimè A, Balg F, Grenier G. Prospective heterotopic ossification progenitors in adult human skeletal muscle. Bone 2015; 71:164-70. [PMID: 25445454 DOI: 10.1016/j.bone.2014.10.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/11/2014] [Accepted: 10/25/2014] [Indexed: 11/16/2022]
Abstract
Skeletal muscle has strong regenerative capabilities. However, failed regeneration can lead to complications where aberrant tissue forms as is the case with heterotopic ossification (HO), in which chondrocytes, osteoblasts and white and brown adipocytes can arise following severe trauma. In humans, the various HO cell types likely originate from multipotent mesenchymal stromal cells (MSCs) in skeletal muscle, which have not been identified in humans until now. In the present study, adherent cells from freshly digested skeletal muscle tissue were expanded in defined culture medium and were FACS-enriched for the CD73(+)CD105(+)CD90(-) population, which displayed robust multilineage potential. Clonal differentiation assays confirmed that all three lineages originated from a single multipotent progenitor. In addition to differentiating into typical HO lineages, human muscle resident MSCs (hmrMSCs) also differentiated into brown adipocytes expressing uncoupling protein 1 (UCP1). Characterizing this novel multipotent hmrMSC population with a brown adipocyte differentiation capacity has enhanced our understanding of the contribution of non-myogenic progenitor cells to regeneration and aberrant tissue formation in human skeletal muscle.
Collapse
Affiliation(s)
- Jennifer Downey
- CHUS Clinical Research Centre, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Peter Kloen
- Department of Orthopedic Surgery, Academic Medical Centre, Amsterdam, The Netherlands
| | - Klaus Klarskov
- CHUS Clinical Research Centre, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Pharmacology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Martin Richter
- CHUS Clinical Research Centre, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Pediatrics, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Reggie Hamdy
- Shriners Hospital for Children, Montreal, QC, Canada; Department of Surgery, Orthopedic Surgery Division, McGill University, Montreal, QC, Canada
| | - Nathalie Faucheux
- CHUS Clinical Research Centre, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Anthony Scimè
- Stem Cell Research Group, Faculty of Health, York University, Toronto, ON, Canada
| | - Frédéric Balg
- CHUS Clinical Research Centre, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Orthopedic Surgery, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Guillaume Grenier
- CHUS Clinical Research Centre, Université de Sherbrooke, Sherbrooke, QC, Canada; Department of Orthopedic Surgery, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
32
|
Tissue extracellular matrix nanoparticle presentation in electrospun nanofibers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:469120. [PMID: 24971329 PMCID: PMC4058126 DOI: 10.1155/2014/469120] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 04/09/2014] [Indexed: 12/27/2022]
Abstract
Biomaterials derived from the decellularization of mature tissues retain biological and architectural features that profoundly influence cellular activity. However, the clinical utility of such materials remains limited as the shape and physical properties are difficult to control. In contrast, scaffolds based on synthetic polymers can be engineered to exhibit specific physical properties, yet often suffer from limited biological functionality. This study characterizes composite materials that present decellularized extracellular matrix (DECM) particles in combination with synthetic nanofibers and examines the ability of these materials to influence stem cell differentiation. Mechanical processing of decellularized tissues yielded particles with diameters ranging from 71 to 334 nm. Nanofiber scaffolds containing up to 10% DECM particles (wt/wt) derived from six different tissues were engineered and evaluated to confirm DECM particle incorporation and to measure bioactivity. Scaffolds containing bone, cartilage, and fat promoted osteogenesis at 1 and 3 weeks compared to controls. In contrast, spleen and lung DECM significantly reduced osteogenic outcomes compared to controls. These findings highlight the potential to incorporate appropriate source DECM nanoparticles within nanofiber composites to design a scaffold with bioactivity targeted to specific applications.
Collapse
|
33
|
Karamichos D, Funderburgh ML, Hutcheon AEK, Zieske JD, Du Y, Wu J, Funderburgh JL. A role for topographic cues in the organization of collagenous matrix by corneal fibroblasts and stem cells. PLoS One 2014; 9:e86260. [PMID: 24465995 PMCID: PMC3897697 DOI: 10.1371/journal.pone.0086260] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022] Open
Abstract
Human corneal fibroblasts (HCF) and corneal stromal stem cells (CSSC) each secrete and organize a thick stroma-like extracellular matrix in response to different substrata, but neither cell type organizes matrix on tissue-culture polystyrene. This study compared cell differentiation and extracellular matrix secreted by these two cell types when they were cultured on identical substrata, polycarbonate Transwell filters. After 4 weeks in culture, both cell types upregulated expression of genes marking differentiated keratocytes (KERA, CHST6, AQP1, B3GNT7). Absolute expression levels of these genes and secretion of keratan sulfate proteoglycans were significantly greater in CSSC than HCF. Both cultures produced extensive extracellular matrix of aligned collagen fibrils types I and V, exhibiting cornea-like lamellar structure. Unlike HCF, CSSC produced little matrix in the presence of serum. Construct thickness and collagen organization was enhanced by TGF-ß3. Scanning electron microscopic examination of the polycarbonate membrane revealed shallow parallel grooves with spacing of 200–300 nm, similar to the topography of aligned nanofiber substratum which we previously showed to induce matrix organization by CSSC. These results demonstrate that both corneal fibroblasts and stromal stem cells respond to a specific pattern of topographical cues by secreting highly organized extracellular matrix typical of corneal stroma. The data also suggest that the potential for matrix secretion and organization may not be directly related to the expression of molecular markers used to identify differentiated keratocytes.
Collapse
Affiliation(s)
- Dimitrios Karamichos
- Schepens Eye Research Institute/Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Martha L. Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Audrey E. K. Hutcheon
- Schepens Eye Research Institute/Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - James D. Zieske
- Schepens Eye Research Institute/Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jian Wu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - James L. Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
34
|
Felthaus O, Gosau M, Ettl T, Prantl L, Morsczeck C. Migration of human dental follicle cells in vitro. J Periodontal Res 2013; 49:205-12. [PMID: 23710611 DOI: 10.1111/jre.12096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND OBJECTIVES The objective of this study was to elucidate the effects of different growth factors on the migration of dental follicle cells (DFCs). DFCs are ectomesenchymally derived easily accessible multipotent stem cells. Cell migration is a crucial step in many biological processes but also for tissue engineering. Growth factors such as epidermal growth factor (EGF), bone morphogenetic protein-2 (BMP2) or transforming growth factor β1 (TGF-β1) can be used to modify the behavior of cells. MATERIAL AND METHODS We used different migration assays (gel spot assay, scratch assay, transwell assay) to evaluate the influence of EGF, BMP2 and TGF-β1 on the migration of DFCs. We investigated the expression of migration-related genes after growth factor stimulation using the PCR array human cell motility. RESULTS DFCs treated with BMP2 or TGF-β1 migrated faster than DFCs treated with EGF. Additionally, more migration-related genes are regulated after treatment with BMP2 or TGF-β1 than with EGF. TGF-β1 additionally functions as a chemoattractant for DFCs. Osteogenic differentiation markers were regulated after BMP2 treatment only. CONCLUSION Whereas the strong migration induced by BMP2 was accompanied by beginning osteogenic differentiation the strong migration induced by TGF-β1 was directional. EGF exhibited not only the weakest migration stimulation but also the weakest induction of differentiation into mineralizing cells.
Collapse
Affiliation(s)
- O Felthaus
- Department of Cranio- and Maxillofacial Surgery, University of Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|