1
|
Otsuka H. Nanofabrication Technologies to Control Cell and Tissue Function in Three-Dimension. Gels 2023; 9:gels9030203. [PMID: 36975652 PMCID: PMC10048556 DOI: 10.3390/gels9030203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/29/2023] Open
Abstract
In the 2000s, advances in cellular micropatterning using microfabrication contributed to the development of cell-based biosensors for the functional evaluation of newly synthesized drugs, resulting in a revolutionary evolution in drug screening. To this end, it is essential to utilize cell patterning to control the morphology of adherent cells and to understand contact and paracrine-mediated interactions between heterogeneous cells. This suggests that the regulation of the cellular environment by means of microfabricated synthetic surfaces is not only a valuable endeavor for basic research in biology and histology, but is also highly useful to engineer artificial cell scaffolds for tissue regeneration. This review particularly focuses on surface engineering techniques for the cellular micropatterning of three-dimensional (3D) spheroids. To establish cell microarrays, composed of a cell adhesive region surrounded by a cell non-adherent surface, it is quite important to control a protein-repellent surface in the micro-scale. Thus, this review is focused on the surface chemistries of the biologically inspired micropatterning of two-dimensional non-fouling characters. As cells are formed into spheroids, their survival, functions, and engraftment in the transplanted site are significantly improved compared to single-cell transplantation. To improve the therapeutic effect of cell spheroids even further, various biomaterials (e.g., fibers and hydrogels) have been developed for spheroid engineering. These biomaterials not only can control the overall spheroid formation (e.g., size, shape, aggregation speed, and degree of compaction), but also can regulate cell-to-cell and cell-to-matrix interactions in spheroids. These important approaches to cell engineering result in their applications to tissue regeneration, where the cell-biomaterial composite is injected into diseased area. This approach allows the operating surgeon to implant the cell and polymer combinations with minimum invasiveness. The polymers utilized in hydrogels are structurally similar to components of the extracellular matrix in vivo, and are considered biocompatible. This review will provide an overview of the critical design to make hydrogels when used as cell scaffolds for tissue engineering. In addition, the new strategy of injectable hydrogel will be discussed as future directions.
Collapse
Affiliation(s)
- Hidenori Otsuka
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, 1-3 Kagurazaka, Shinjuku-ku, Tokyo 162-8601, Japan
| |
Collapse
|
2
|
Gao B, Gao W, Wu Z, Zhou T, Qiu X, Wang X, Lian C, Peng Y, Liang A, Qiu J, Zhu Y, Xu C, Li Y, Su P, Huang D. Melatonin rescued interleukin 1β-impaired chondrogenesis of human mesenchymal stem cells. Stem Cell Res Ther 2018; 9:162. [PMID: 29898779 PMCID: PMC6001057 DOI: 10.1186/s13287-018-0892-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/15/2018] [Accepted: 05/02/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a widespread arthritic disease and a primary cause of disability. Increasing evidence suggests that inflammation has a pivotal part in its pathogenesis. Interleukin-1β (IL-1β) is a primary mediator of local inflammatory processes in OA. Current therapies for OA mainly focus on the symptoms of the advanced stage of the disease. The possible utilization of bone marrow mesenchymal stem cells (BMSCs) to regenerate cartilage is an appealing method, but in the case of OA requires chondrogenesis to take place within an inflamed environment. Our previous study showed that melatonin (MLT) can promote chondrogenic differentiation of MSCs, but whether MLT can rescue IL-1β-impaired chondrogenesis in human BMSCs has not yet been established. MLT, which can have anti-inflammatory and prochondrogenic effects, has demonstrated potential in defeating IL-1β-induced inhibition of chondrogenesis and further study should be conducted. METHODS Human bone marrow-derived MSCs were separated and cultured based on our system that was already documented. A high-density micromass culture system was used for the chondrogenic differentiation of human BMSCs, which was also described previously. Human BMSCs were induced for chondrogenesis for 7, 14, and 21 days with the treatment of IL-1β and MLT. The cultured cartilage pellets were then evaluated by morphology, extracellular matrix accumulation, and chondrogenic, metabolic, and apoptotic marker expression. Furthermore, cell apoptosis was assessed by TUNEL assay. The phosphorylation level P65 and IκBα of the NF-κB pathway activity was explored on day 21 of chondrogenic differentiation of BMSCs. RESULTS The current evaluation showed that MLT can save IL-1β-impaired chondrogenesis of human BMSCs in different aspects. Firstly, MLT can restore the chondrogenic pellet size, and rescue matrix synthesis and accumulation. Secondly, MLT can upregulate chondrogenic marker COL2A1 expression at both mRNA and protein levels, and also regulate the expression levels of other chondrogenic markers like ACAN, SOX9, and COL10A1 in the presence of IL-1β. Thirdly, MLT can maintain the metabolic balance of the chondrogenic process by suppressing expression of catabolic genes, such as MMP, MMP13, and ADAMTS4. Furthermore, MLT can subdue IL-1β-induced cell apoptosis of BMSCs throughout chondrogenesis. Meanwhile, MLT suppressed the phosphorylation level of P65 and IκBα, which were elevated by IL-1β treatment, indicating that MLT can attenuate the IL-1β-induced activation of NF-κB signaling. CONCLUSION The current evaluation showed that MLT can save IL-1β-impaired chondrogenesis of human BMSCs by restoring the pellet size and matrix accumulation, and maintaining the metabolic balance, reducing cell apoptosis. Our study also showed that MLT can attenuate the IL-1β-induced activation of the NF-κB signaling pathway, which is the most important pathway downstream of IL-1β, and plays a crucial role in inflammation, apoptosis, and metabolism. Thus, MLT has prospects for treating OA due to its multifaceted functions, such as mitigating inflammation, maintaining metabolic balance, and mitigating apoptosis.
Collapse
Affiliation(s)
- Bo Gao
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China.,Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zizhao Wu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Taifeng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China
| | - Xianjian Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Xudong Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Chengjie Lian
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China
| | - Yan Peng
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Jincheng Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Yuanxin Zhu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China
| | - Caixia Xu
- Research Centre for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yibing Li
- Department of Spine Surgery, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Peiqiang Su
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, #58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China.
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, #107 West Yan Jiang Road, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
3
|
Nazim UM, Moon JH, Lee YJ, Seol JW, Kim YJ, Park SY. Glipizide sensitizes lung cancer cells to TRAIL-induced apoptosis via Akt/mTOR/autophagy pathways. Oncotarget 2017; 8:100021-100033. [PMID: 29245957 PMCID: PMC5724999 DOI: 10.18632/oncotarget.21754] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 05/31/2017] [Indexed: 12/11/2022] Open
Abstract
The combination of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) with subsidiary agents is a promising anticancer strategy to conquer TRAIL resistance in malignant cells. Glipizide is a second-generation oral hypoglycemic medicine for the cure of type II diabetes because of its capability to selectively stimulate insulin secretion from β-cells. In this study, we revealed that glipizide could trigger TRAIL-mediated apoptotic cell death in human lung adenocarcinoma cells. Pretreatment with glipizide downregulation of p-Akt and p-mTOR in different concentrations. In addition, LC3-II and p-Akt was suppressed in the presence of LY294002, a well-known inhibitor of P13K. Treatment with glipizide commenced in a slight increase in conversion rate of LC3-I to LC3-II and significantly decreased p62 expression levels in a dose-dependent manner. This indicates that glipizide encouraged autophagy flux activation in human lung cancer cells. Inhibition of autophagy flux applying a specific inhibitor and genetically modified ATG5 siRNA enclosed glipizide-mediated enhancing effect of TRAIL. These data demonstrate that inhibition of Akt/mTOR by glipizide sensitizes TRAIL-induced tumor cell death through activating autophagy flux and also suggest that glipizide may be a combination therapeutic target with TRAIL protein in TRAIL-resistant cancer cells.
Collapse
Affiliation(s)
- Uddin Md Nazim
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Ji-Hong Moon
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - You-Jin Lee
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Yong Ju Kim
- Department of Herbal Medicine Resources, College of Environmental and Bioresources, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, South Korea
| |
Collapse
|
4
|
Ayerst BI, Merry CLR, Day AJ. The Good the Bad and the Ugly of Glycosaminoglycans in Tissue Engineering Applications. Pharmaceuticals (Basel) 2017; 10:E54. [PMID: 28608822 PMCID: PMC5490411 DOI: 10.3390/ph10020054] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022] Open
Abstract
High sulfation, low cost, and the status of heparin as an already FDA- and EMA- approved product, mean that its inclusion in tissue engineering (TE) strategies is becoming increasingly popular. However, the use of heparin may represent a naïve approach. This is because tissue formation is a highly orchestrated process, involving the temporal expression of numerous growth factors and complex signaling networks. While heparin may enhance the retention and activity of certain growth factors under particular conditions, its binding 'promiscuity' means that it may also inhibit other factors that, for example, play an important role in tissue maintenance and repair. Within this review we focus on articular cartilage, highlighting the complexities and highly regulated processes that are involved in its formation, and the challenges that exist in trying to effectively engineer this tissue. Here we discuss the opportunities that glycosaminoglycans (GAGs) may provide in advancing this important area of regenerative medicine, placing emphasis on the need to move away from the common use of heparin, and instead focus research towards the utility of specific GAG preparations that are able to modulate the activity of growth factors in a more controlled and defined manner, with less off-target effects.
Collapse
Affiliation(s)
- Bethanie I Ayerst
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| | - Catherine L R Merry
- Stem Cell Glycobiology Group, Wolfson Centre for Stem Cells, Tissue Engineering & Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| |
Collapse
|
5
|
Goldberg A, Mitchell K, Soans J, Kim L, Zaidi R. The use of mesenchymal stem cells for cartilage repair and regeneration: a systematic review. J Orthop Surg Res 2017; 12:39. [PMID: 28279182 PMCID: PMC5345159 DOI: 10.1186/s13018-017-0534-y] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/13/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The management of articular cartilage defects presents many clinical challenges due to its avascular, aneural and alymphatic nature. Bone marrow stimulation techniques, such as microfracture, are the most frequently used method in clinical practice however the resulting mixed fibrocartilage tissue which is inferior to native hyaline cartilage. Other methods have shown promise but are far from perfect. There is an unmet need and growing interest in regenerative medicine and tissue engineering to improve the outcome for patients requiring cartilage repair. Many published reviews on cartilage repair only list human clinical trials, underestimating the wealth of basic sciences and animal studies that are precursors to future research. We therefore set out to perform a systematic review of the literature to assess the translation of stem cell therapy to explore what research had been carried out at each of the stages of translation from bench-top (in vitro), animal (pre-clinical) and human studies (clinical) and assemble an evidence-based cascade for the responsible introduction of stem cell therapy for cartilage defects. This review was conducted in accordance to PRISMA guidelines using CINHAL, MEDLINE, EMBASE, Scopus and Web of Knowledge databases from 1st January 1900 to 30th June 2015. In total, there were 2880 studies identified of which 252 studies were included for analysis (100 articles for in vitro studies, 111 studies for animal studies; and 31 studies for human studies). There was a huge variance in cell source in pre-clinical studies both of terms of animal used, location of harvest (fat, marrow, blood or synovium) and allogeneicity. The use of scaffolds, growth factors, number of cell passages and number of cells used was hugely heterogeneous. SHORT CONCLUSIONS This review offers a comprehensive assessment of the evidence behind the translation of basic science to the clinical practice of cartilage repair. It has revealed a lack of connectivity between the in vitro, pre-clinical and human data and a patchwork quilt of synergistic evidence. Drivers for progress in this space are largely driven by patient demand, surgeon inquisition and a regulatory framework that is learning at the same pace as new developments take place.
Collapse
Affiliation(s)
- Andy Goldberg
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| | - Katrina Mitchell
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| | - Julian Soans
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| | - Louise Kim
- Joint Research and Enterprise Office, St George’s University of London and St George’s University Hospitals NHS Foundation Trust, Hunter Wing, Cranmer Terrace, London, SW17 0RE UK
| | - Razi Zaidi
- Institute of Orthopaedics and Musculoskeletal Science, Royal National Orthopaedic Hospital (RNOH), Brockley Hill Stanmore, London, HA7 4LP UK
| |
Collapse
|
6
|
Rodrigues MN, Oliveira MB, Costa RR, Mano JF. Chitosan/Chondroitin Sulfate Membranes Produced by Polyelectrolyte Complexation for Cartilage Engineering. Biomacromolecules 2016; 17:2178-88. [DOI: 10.1021/acs.biomac.6b00399] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Mariana N. Rodrigues
- 3B’s
Research Group, Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence of Tissue Engineering and Regenerative
Medicine, Avepark − Parque de
Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s,
PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Mariana B. Oliveira
- 3B’s
Research Group, Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence of Tissue Engineering and Regenerative
Medicine, Avepark − Parque de
Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s,
PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - Rui R. Costa
- 3B’s
Research Group, Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence of Tissue Engineering and Regenerative
Medicine, Avepark − Parque de
Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s,
PT Government Associated Laboratory, Braga/Guimarães, Portugal
| | - João F. Mano
- 3B’s
Research Group, Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence of Tissue Engineering and Regenerative
Medicine, Avepark − Parque de
Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s,
PT Government Associated Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
7
|
Colombier P, Clouet J, Boyer C, Ruel M, Bonin G, Lesoeur J, Moreau A, Fellah BH, Weiss P, Lescaudron L, Camus A, Guicheux J. TGF-β1 and GDF5 Act Synergistically to Drive the Differentiation of Human Adipose Stromal Cells towardNucleus Pulposus-like Cells. Stem Cells 2015; 34:653-67. [DOI: 10.1002/stem.2249] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 10/09/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Pauline Colombier
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
| | - Johann Clouet
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
- Université de Nantes, UFR Sciences Biologiques et Pharmaceutiques; Nantes France
- CHU Nantes, Pharmacie Centrale, PHU 11; Nantes France
| | - Cécile Boyer
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
| | - Maëva Ruel
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
| | - Gaëlle Bonin
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
| | - Julie Lesoeur
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
| | - Anne Moreau
- Université de Nantes, UFR Médecine; Nantes France
- CHU Nantes, Service d'Anatomopathologie; Nantes France
| | - Borhane-Hakim Fellah
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
- CRIP, Centre de Recherche et d'Investigations Précliniques, ONIRIS; Nantes France
| | - Pierre Weiss
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
- CHU Nantes, PHU 4 OTONN; Nantes France
| | - Laurent Lescaudron
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
- Université de Nantes, UFR Sciences et Techniques; Nantes France
| | - Anne Camus
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
| | - Jérôme Guicheux
- INSERM UMRS 791, Laboratoire d'Ingénierie Osteo Articulaire et Dentaire (LIOAD); Nantes France
- Université de Nantes, UFR Odontologie; Nantes France
- CHU Nantes, PHU 4 OTONN; Nantes France
| |
Collapse
|
8
|
Dang PN, Solorio LD, Alsberg E. Driving cartilage formation in high-density human adipose-derived stem cell aggregate and sheet constructs without exogenous growth factor delivery. Tissue Eng Part A 2015; 20:3163-75. [PMID: 24873753 DOI: 10.1089/ten.tea.2012.0551] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
An attractive cell source for cartilage tissue engineering, human adipose-derived stem cells (hASCs) can be easily expanded and signaled to differentiate into chondrocytes. This study explores the influence of growth factor distribution and release kinetics on cartilage formation within 3D hASC constructs incorporated with transforming growth factor-β1 (TGF-β1)-loaded gelatin microspheres. The amounts of microspheres, TGF-β1 concentration, and polymer degradation rate were varied within hASC aggregates. Microsphere and TGF-β1 loading concentrations were identified that resulted in glycosaminoglycan (GAG) production comparable to those of control aggregates cultured in TGF-β1-containing medium. Self-assembling hASC sheets were then engineered for the production of larger, more clinically relevant constructs. Chondrogenesis was observed in hASC-only sheets cultured with exogenous TGF-β1 at 3 weeks. Importantly, sheets with incorporated TGF-β1-loaded microspheres achieved GAG production similar to sheets treated with exogenous TGF-β1. Cartilage formation was confirmed histologically via observation of cartilage-like morphology and GAG staining. This is the first demonstration of the self-assembly of hASCs into high-density cell sheets capable of forming cartilage in the presence of exogenous TGF-β1 or with TGF-β1-releasing microspheres. Microsphere incorporation may bypass the need for extended in vitro culture, potentially enabling hASC sheets to be implanted more rapidly into defects to regenerate cartilage in vivo.
Collapse
Affiliation(s)
- Phuong N Dang
- 1 Department of Biomedical Engineering, Case Western Reserve University , Cleveland, Ohio
| | | | | |
Collapse
|
9
|
Qi C, Zhou Q, Li B, Yang Y, Cao L, Ye Y, Li J, Ding Y, Wang H, Wang J, He X, Zhang Q, Lan T, Lee KKH, Li W, Song X, Zhou J, Yang X, Wang L. Glipizide, an antidiabetic drug, suppresses tumor growth and metastasis by inhibiting angiogenesis. Oncotarget 2015; 5:9966-79. [PMID: 25294818 PMCID: PMC4259451 DOI: 10.18632/oncotarget.2483] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Angiogenesis is involved in the development, progression and metastasis of various human cancers. Herein, we report the discovery of glipizide, a widely used drug for type 2 diabetes mellitus, as a promising anticancer agent through the inhibition of tumor angiogenesis. By high-throughput screening (HTS) of an FDA approved drug library utilizing our in vivo chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) models, glipizide has been identified to significantly inhibit blood vessel formation and development. Moreover, glipizide was found to suppress tumor angiogenesis, tumor growth and metastasis using xenograft tumor and MMTV-PyMT transgenic mouse models. We further revealed that the anticancer capability of glipizide is not attributed to its antiproliferative effects, which are not significant against various human cancer cell lines. To investigate whether its anticancer efficacy is associated with the glucose level alteration induced by glipizide application, glimepiride, another medium to long-acting sulfonylurea antidiabetic drug in the same class, was employed for the comparison studies in the same fashion. Interestingly, glimepiride has demonstrated no significant impact on the tumor growth and metastasis, indicating that the anticancer effects of glipizide is not ascribed to its antidiabetic properties. Furthermore, glipizide suppresses endothelial cell migration and the formation of tubular structures, thereby inhibiting angiogenesis by up-regulating the expression of natriuretic peptide receptor A. These findings uncover a novel mechanism of glipizide as a potential cancer therapy, and also for the first time, provide direct evidence to support that treatment with glipizide may reduce the cancer risk for diabetic patients.
Collapse
Affiliation(s)
- Cuiling Qi
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China. These authors contributed equally to this work
| | - Qin Zhou
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China. These authors contributed equally to this work
| | - Bin Li
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yang Yang
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, China Medical University, He Ping District, Shen Yang City, Liao Ning Province, China
| | - Yuxiang Ye
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiangchao Li
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Ding
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huiping Wang
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jintao Wang
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaodong He
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qianqian Zhang
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tian Lan
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kenneth Ka Ho Lee
- Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Weidong Li
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoyu Song
- Key Laboratory of Medical Cell Biology, China Medical University, He Ping District, Shen Yang City, Liao Ning Province, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou, China
| | - Lijing Wang
- Vascular Biology Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
10
|
HE HONGHUI, CHEN KE, WANG FANG, ZHAO LILING, WAN XINXING, WANG LINGHAO, MO ZHAOHUI. miR-204-5p promotes the adipogenic differentiation of human adipose-derived mesenchymal stem cells by modulating DVL3 expression and suppressing Wnt/β-catenin signaling. Int J Mol Med 2015; 35:1587-95. [PMID: 25847080 PMCID: PMC4432921 DOI: 10.3892/ijmm.2015.2160] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 03/27/2015] [Indexed: 11/10/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) play an important regulatory role during adipogenesis, and have been studied extensively in this regard. Specifically, the switch between the differentiation of mesenchymal stem cells (MSCs) towards adipogenic vs. osteogenic lineages is regulated by miR-204 which controls the expression of Runx2. However, the association between miR-204-5p and the Wnt/β-catenin signaling pathway during adipogenesis has not yet been clarified. In the present study, we demonstrate that miR-204-5p regulates the in vitro adipogenesis of human adipose-derived mesenchymal stem cells (hADSCs). The level of miR-204-5p was shown to be gradually upregulated during adipocytic differentiation, together with the mRNA expression of the critical adipogenic transcription factors, cytidine-cytidine-adenosine-adenosine-thymidine (CCAAT) enhancer binding protein α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ), and the mature adipogenic marker, fatty acid binding protein 4 (FABP4). We further demonstrate that while the overexpression of miR-204-5p promotes adipogenesis, its knockdown causes the inhibition of this process. We then used bioinformatics tools and luciferase reporter assay to establish that dishevelled segment polarity protein 3 (DVL3), a key regulator of the Wnt/β-catenin signaling pathway, is a direct target of miR-204-5p. In addition, the overexpression of DVL3 led to an increase in β-catenin and cyclin D1 (CCND1) expression and, by contrast, the knockdown of DVL3 led to a decrease in the expression of β-catenin and CCND1. The knockdown of DVL3 significantly promoted adipogenesis. Finally, we demonstrated that the overexpression of miR-204-5p induced the downregulation of β-catenin and the canonical Wnt target gene, CCND1, in mature adipoctyes, while its knockdown led to their upregulation. Taken together, our data suggest that miR-204-5p regulates adipogenesis by controlling DVL3 expression and subsequently inhibiting the activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - ZHAOHUI MO
- Correspondence to: Professor Zhaohui Mo, Department of Endocrinology, Third Xiangya Hospital of Central South University, 138 Tong Zi Po Road, Changsha, Hunan 410013, P.R. China, E-mail:
| |
Collapse
|
11
|
Liu Y, Wang DA. Viral vector-mediated transgenic cell therapy in regenerative medicine: safety of the process. Expert Opin Biol Ther 2014; 15:559-67. [PMID: 25528865 DOI: 10.1517/14712598.2015.995086] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|