1
|
Harper CV, Eccles L, Henstock J, Charnock JC. Trophoblast-derived factors drive human mesenchymal stem cell differentiation along an endothelial lineage: A model of early placental vasculogenesis. Reprod Biol 2025; 25:100994. [PMID: 39823693 DOI: 10.1016/j.repbio.2025.100994] [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: 10/08/2024] [Revised: 12/19/2024] [Accepted: 01/04/2025] [Indexed: 01/20/2025]
Abstract
Mechanisms controlling the process and patterning of blood vessel development in the placenta remain largely unknown. The close physical proximity of early blood vessels observed in the placenta and the cytotrophoblast, as well as the reported production of vasculogenic growth factors by the latter, suggests that signalling between these two niches may be important. Here, we have developed an in vitro model to address the hypothesis that the cytotrophoblast, by the secretion of soluble factors, drives differentiation of resident sub-trophoblastic mesenchymal stem cells (MSCs) along a vascular lineage, thereby establishing feto-placental circulation. BM-MSCs (a readily available model for placental stem cells) were treated with conditioned medium containing the secretome from human BeWo trophoblast cells, or endothelial growth medium (EGM2) supplemented with exogenous growth factors (VEGF, IGF1 and EGF) for 10-12 days. Trophoblast-conditioned media, found to contain detectable concentrations of cytokines including VEGF, uPAR, TIMP-1, TIMP-2, IL6 and placental growth factor, induced the expression of the endothelial genes CD31, von Willibrand factor (vWF), FLT-1, VEGFR2 and VE-Cadherin. Upregulation of vWF protein was also detected following growth in trophoblast-conditioned media, using immunocytochemistry. Wound healing (migration assay) and Matrigel-tube formation assays confirmed that the BM-MSCs cultured in trophoblast-conditioned media exhibited functional measures of endothelial cells in addition to expressing relevant markers. Identification of key trophoblast-secreted factors and their promotion of endothelial differentiation in BM-MSCs helps advance our theories regarding the close relationship of the mesenchymal stem cell-cytotrophoblast niche in coordinating the complex angiogenic events that occur in the placenta. The in vitro model presented here provides an accessible and reproducible tool for further investigations into placental development.
Collapse
Affiliation(s)
| | - Leah Eccles
- Department of Biology, Edge Hill University, L39 4QP, UK
| | - James Henstock
- Faculty of Health & Life Sciences, Northumbria University, Newcastle-upon-Tyne NE1 8SU, UK
| | | |
Collapse
|
2
|
Prince E. Designing Biomimetic Strain-Stiffening into Synthetic Hydrogels. Biomacromolecules 2024; 25:6283-6295. [PMID: 39356204 DOI: 10.1021/acs.biomac.4c00756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Biological tissues are mechanoresponsive; that is, their properties dynamically change in response to mechanical stimuli. For example, in response to shear or elongational strain, collagen, fibrin, actin, and other filamentous biomaterials undergo dramatic strain-stiffening. Above a critical strain, their stiffness increases over orders of magnitude. While it is widely accepted that the stiffness of biological tissues impacts cell phenotype and several diseases, the biological impact of strain-stiffening remains understudied. Synthetic hydrogels that mimic the mechanoresponsive nature of biological tissues could serve as an in vitro platform for these studies. This review highlights recent efforts to mimic the strain-stiffening behavior of biological materials in synthetic hydrogels. We discuss the design principles for imparting synthetic hydrogels with biomimetic strain-stiffening, critically compare designs of strain-stiffening hydrogels that have been reported thus far, and discuss their use as in vitro platforms to probe how strain-stiffening impacts cell behavior, diseases, and other biological processes.
Collapse
Affiliation(s)
- Elisabeth Prince
- Department of Chemical Engineering, University of Waterloo, 200 University Ave. West, N2L 3G1 Waterloo, ON, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Ave. West, N2L 3G1 Waterloo, ON, Canada
| |
Collapse
|
3
|
Dalal S, Ramirez-Gomez J, Sharma B, Devara D, Kumar S. MicroRNAs and synapse turnover in Alzheimer's disease. Ageing Res Rev 2024; 99:102377. [PMID: 38871301 DOI: 10.1016/j.arr.2024.102377] [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: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-beta plaques and neurofibrillary tangles in the brain, leading to synaptic dysfunction and cognitive decline. Healthy synapses are the crucial for normal brain function, memory restoration and other neurophysiological function. Synapse loss and synaptic dysfunction are two primary events that occur during AD initiation. Synapse lifecycle and/or synapse turnover is divided into five key stages and several sub-stages such as synapse formation, synapse assembly, synapse maturation, synapse transmission and synapse termination. In normal state, the synapse turnover is regulated by various biological and molecular factors for a healthy neurotransmission. In AD, the different stages of synapse turnover are affected by AD-related toxic proteins. MicroRNAs (miRNAs) have emerged as critical regulators of gene expression and have been implicated in various neurological diseases, including AD. Deregulation of miRNAs modulate the synaptic proteins and affect the synapse turnover at different stages. In this review, we discussed the key milestones of synapse turnover and how they are affected in AD. Further, we discussed the involvement of miRNAs in synaptic turnover, focusing specifically on their role in AD pathogenesis. We also emphasized the regulatory mechanisms by which miRNAs modulate the synaptic turnover stages in AD. Current studies will help to understand the synaptic life-cycle and role of miRNAs in each stage that is deregulated in AD, further allowing for a better understanding of the pathogenesis of devastating disease.
Collapse
Affiliation(s)
- Sarthak Dalal
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Jaime Ramirez-Gomez
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Bhupender Sharma
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Davin Devara
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Subodh Kumar
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA; L. Frederick Francis Graduate School of Biomedicael Sciences, Texas Tech University Health Sciences Center, El Paso, TX, USA.
| |
Collapse
|
4
|
Koç-Demir A, Elçin AE, Elçin YM. Magnetic biocomposite scaffold based on decellularized tendon ECM and MNP-deposited halloysite nanotubes: physicochemical, thermal, rheological, mechanical and in vitrobiological evaluations. Biomed Mater 2024; 19:035027. [PMID: 38537375 DOI: 10.1088/1748-605x/ad38ab] [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: 10/18/2023] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
The development of new three-dimensional biomaterials with advanced versatile properties is critical to the success of tissue engineering (TE) applications. Here, (a) bioactive decellularized tendon extracellular matrix (dECM) with a sol-gel transition feature at physiological temperature, (b) halloysite nanotubes (HNT) with known mechanical properties and bioactivity, and (c) magnetic nanoparticles (MNP) with superparamagnetic and osteogenic properties were combined to develop a new scaffold that could be used in prospective bone TE applications. Deposition of MNPs on HNTs resulted in magnetic nanostructures without agglomeration of MNPs. A completely cell-free, collagen- and glycosaminoglycan- rich dECM was obtained and characterized. dECM-based scaffolds incorporated with 1%, 2% and 4% MNP-HNT were analysed for their physical, chemical, andin vitrobiological properties. Fourier-transform infrared spectroscopy, x-ray powder diffractometry and vibrating sample magnetometry analyses confirmed the presence of dECM, HNT and MNP in all scaffold types. The capacity to form apatite layer upon incubation in simulated body fluid revealed that dECM-MNP-HNT is a bioactive material. Combining dECM with MNP-HNT improved the thermal stability and compressive strength of the macroporous scaffolds upto 2% MNP-HNT.In vitrocytotoxicity and hemolysis experiments showed that the scaffolds were essentially biocompatible. Human bone marrow mesenchymal stem cells adhered and proliferated well on the macroporous constructs containing 1% and 2% MNP-HNT; and remained metabolically active for at least 21 din vitro. Collectively, the findings support the idea that magnetic nanocomposite dECM scaffolds containing MNP-HNT could be a potential template for TE applications.
Collapse
Affiliation(s)
- Aysel Koç-Demir
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Ayşe Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
- Biovalda Health Technologies, Inc., Ankara, Turkey
| |
Collapse
|
5
|
Lopes V, Moreira G, Bramini M, Capasso A. The potential of graphene coatings as neural interfaces. NANOSCALE HORIZONS 2024; 9:384-406. [PMID: 38231692 DOI: 10.1039/d3nh00461a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Recent advances in nanotechnology design and fabrication have shaped the landscape for the development of ideal cell interfaces based on biomaterials. A holistic evaluation of the requirements for a cell interface is a highly complex task. Biocompatibility is a crucial requirement which is affected by the interface's properties, including elemental composition, morphology, and surface chemistry. This review explores the current state-of-the-art on graphene coatings produced by chemical vapor deposition (CVD) and applied as neural interfaces, detailing the key properties required to design an interface capable of physiologically interacting with neural cells. The interfaces are classified into substrates and scaffolds to differentiate the planar and three-dimensional environments where the cells can adhere and proliferate. The role of specific features such as mechanical properties, porosity and wettability are investigated. We further report on the specific brain-interface applications where CVD graphene paved the way to revolutionary advances in biomedicine. Future studies on the long-term effects of graphene-based materials in vivo will unlock even more potentially disruptive neuro-applications.
Collapse
Affiliation(s)
- Vicente Lopes
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal.
| | - Gabriel Moreira
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal.
| | - Mattia Bramini
- Department of Cell Biology, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain.
| | - Andrea Capasso
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal.
| |
Collapse
|
6
|
Xu X, Feng Q, Ma X, Deng Y, Zhang K, Ooi HS, Yang B, Zhang ZY, Feng B, Bian L. Dynamic gelatin-based hydrogels promote the proliferation and self-renewal of embryonic stem cells in long-term 3D culture. Biomaterials 2022; 289:121802. [PMID: 36152514 DOI: 10.1016/j.biomaterials.2022.121802] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/12/2022] [Accepted: 09/09/2022] [Indexed: 11/02/2022]
Abstract
Long-term maintenance of embryonic stem cells (ESCs) in the undifferentiated state is still challenging. Compared with traditional 2D culture methods, 3D culture in biomaterials such as hydrogels is expected to better support the long-term self-renewal of ESCs by emulating the biophysical and biochemical properties of the extracellular matrix (ECM). Although prior studies showed that soft and degradable hydrogels favor the 3D growth of ESCs, few studies have examined the impact of the structural dynamics of the hydrogel matrix on ESC behaviors. Herein, we report a gelatin-based structurally dynamic hydrogel (GelCD hydrogel) that emulates the intrinsic structural dynamics of the ECM. Compared with covalently crosslinked gelatin hydrogels (GelMA hydrogels) with similar stiffness and biodegradability, GelCD hydrogels significantly promote the clonal expansion and viability of encapsulated mouse ESCs (mESCs) independent of MMP-mediated hydrogel degradation. Furthermore, GelCD hydrogels better maintain the pluripotency of encapsulated mESCs than do traditional 2D culture methods that use MEF feeder cells or medium supplementation with GSK3β and MEK 1/2 inhibitors (2i). When cultured in GelCD hydrogels for an extended period (over 2 months) with cell passaging every 7 days, mESCs preserve their normal morphology and maintain their pluripotency and full differentiation capability. Our findings highlight the critical role of the structural dynamics of the hydrogel matrix in accommodating the volume expansion that occurs during clonal ESC growth, and we believe that our dynamic hydrogels represent a valuable tool to support the long-term 3D culture of ESCs.
Collapse
Affiliation(s)
- Xiayi Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China.
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Chongqing, 400044, China
| | - Xun Ma
- Center for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences Limited, Hong Kong SAR, 999077, China; School of Biomedical Sciences, Faculty of Medicine, Institute for Tissue Engineering and Regenerative Medicine (iTERM), CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yingrui Deng
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Kunyu Zhang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, China
| | - Hon Son Ooi
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, 510150, China.
| | - Bo Feng
- Center for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences Limited, Hong Kong SAR, 999077, China; School of Biomedical Sciences, Faculty of Medicine, Institute for Tissue Engineering and Regenerative Medicine (iTERM), CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China; Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
7
|
Roberts RM, Ezashi T, Temple J, Owen JR, Soncin F, Parast MM. The role of BMP4 signaling in trophoblast emergence from pluripotency. Cell Mol Life Sci 2022; 79:447. [PMID: 35877048 PMCID: PMC10243463 DOI: 10.1007/s00018-022-04478-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/24/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
The Bone Morphogenetic Protein (BMP) signaling pathway has established roles in early embryonic morphogenesis, particularly in the epiblast. More recently, however, it has also been implicated in development of extraembryonic lineages, including trophectoderm (TE), in both mouse and human. In this review, we will provide an overview of this signaling pathway, with a focus on BMP4, and its role in emergence and development of TE in both early mouse and human embryogenesis. Subsequently, we will build on these in vivo data and discuss the utility of BMP4-based protocols for in vitro conversion of primed vs. naïve pluripotent stem cells (PSC) into trophoblast, and specifically into trophoblast stem cells (TSC). PSC-derived TSC could provide an abundant, reproducible, and ethically acceptable source of cells for modeling placental development.
Collapse
Affiliation(s)
- R Michael Roberts
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Toshihiko Ezashi
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Colorado Center for Reproductive Medicine, 10290 Ridgegate Circle, Lone Tree, CO, 80124, USA
| | - Jasmine Temple
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Joseph R Owen
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
| |
Collapse
|
8
|
Rosellini E, Cascone MG. Biomimetic Strategies to Develop Bioactive Scaffolds for Myocardial Tissue Engineering. Open Biomed Eng J 2022. [DOI: 10.2174/18741207-v16-e2205090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this paper is to provide an overview of the results of the research activity carried out in our laboratories, over the last 10 years, in relation to the development of strategies for the production of biomimetic and bioactive scaffolds for myocardial tissue engineering. Biomimetic and bioactive polymeric scaffolds for cardiac regeneration were designed and manufactured in our laboratories and their morphological, physicochemical, mechanical and biological properties were investigated by different techniques, such as scanning electron microscopy, infrared chemical imaging, swelling test, in vitro degradation assessment, dynamic mechanical analysis, in vitro and in vivo biological tests. Biomimetic scaffolds, able to favor tissue regeneration by mimicking nature, were engineered by different strategies, comprising: (i) the imitation of the composition and interactions among components of the natural extracellular matrix (ECM), by mixing of proteins and polysaccharides; (ii) the material surface modification, using both traditional and innovative techniques, such as molecular imprinting; (iii) the incorporation and release of specific active agents and (iv) the production of scaffolds with a microarchitecture similar to that of native ECM. All the developed strategies were found to be effective in creating materials able to influence cellular behavior and therefore to favor the process of new tissue formation. In particular, the approach based on the combination of different strategies aimed at creating a system capable of communicating with the cells and promoting specific cellular responses, as the ECM does, has appeared particularly promising, in view to favor the formation of a tissue equivalent to the cardiac one.
Collapse
|
9
|
Bonet F, Inácio JM, Bover O, Añez SB, Belo JA. CCBE1 in Cardiac Development and Disease. Front Genet 2022; 13:836694. [PMID: 35222551 PMCID: PMC8864227 DOI: 10.3389/fgene.2022.836694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 12/04/2022] Open
Abstract
The collagen- and calcium-binding EGF-like domains 1 (CCBE1) is a secreted protein extensively described as indispensable for lymphangiogenesis during development enhancing VEGF-C signaling. In human patients, mutations in CCBE1 have been found to cause Hennekam syndrome, an inherited disease characterized by malformation of the lymphatic system that presents a wide variety of symptoms such as primary lymphedema, lymphangiectasia, and heart defects. Importantly, over the last decade, an essential role for CCBE1 during heart development is being uncovered. In mice, Ccbe1 expression was initially detected in distinct cardiac progenitors such as first and second heart field, and the proepicardium. More recently, Ccbe1 expression was identified in the epicardium and sinus venosus (SV) myocardium at E11.5–E13.5, the stage when SV endocardium–derived (VEGF-C dependent) coronary vessels start to form. Concordantly, CCBE1 is required for the correct formation of the coronary vessels and the coronary artery stem in the mouse. Additionally, Ccbe1 was found to be enriched in mouse embryonic stem cells (ESC) and revealed as a new essential gene for the differentiation of ESC-derived early cardiac precursor cell lineages. Here, we bring an up-to-date review on the role of CCBE1 in cardiac development, function, and human disease implications. Finally, we envisage the potential of this molecule’s functions from a regenerative medicine perspective, particularly novel therapeutic strategies for heart disease.
Collapse
Affiliation(s)
- Fernando Bonet
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
- Medicine Department, School of Medicine, University of Cádiz (UCA), Cádiz, Spain
- Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - José M. Inácio
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Oriol Bover
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Sabrina B. Añez
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - José A. Belo
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
- *Correspondence: José A. Belo,
| |
Collapse
|
10
|
Nihad M, Shenoy P S, Bose B. Cell therapy research for Diabetes: Pancreatic β cell differentiation from pluripotent stem cells. Diabetes Res Clin Pract 2021; 181:109084. [PMID: 34673084 DOI: 10.1016/j.diabres.2021.109084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (PSCs), both embryonic and induced pluripotent stem cells (iPSCs), have been differentiated into pancreatic β isletsin vitrofor more than a decade. The idea is to get enough β cells for cell transplantation for diabetics. Finding a standard cell therapy for diabetes is essential because of the logarithmic increase in the global population of people with diabetes and the insufficient availability of the human cadaveric pancreas. Moreover, with better insights into developmental biology, thein vitroβ cell differentiation protocols have depended on thein vivoβ cell organogenesis. Various protocols for pancreatic β cell differentiation have been developed. Such protocols are based on the modulation of cell signalling pathways with growth factors, small molecules, RNAi approaches, directed differentiation using transcription factors, genome editing. Growth factor free differentiation protocols, epigenetic modulations, 3D differentiation approaches, and encapsulation strategies have also been reported for better glycemic control and endocrine modulations. Here, we have reviewed various aforementionedin vitroβ cell differentiation protocols from human PSCs, their respective comparisons, challenges, past, present, and future. The literature has been reviewed primarily from PubMed from the year 2000 till date using the mentioned keywords.
Collapse
Affiliation(s)
- Muhammad Nihad
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India.
| |
Collapse
|
11
|
Shim NY, Heo JS. Performance of the Polydopamine-Graphene Oxide Composite Substrate in the Osteogenic Differentiation of Mouse Embryonic Stem Cells. Int J Mol Sci 2021; 22:ijms22147323. [PMID: 34298943 PMCID: PMC8303500 DOI: 10.3390/ijms22147323] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Graphene oxide (GO) is a biocompatible material considered a favorable stem cell culture substrate. In this study, GO was modified with polydopamine (PDA) to facilitate depositing GO onto a tissue culture polystyrene (PT) surface, and the osteogenic performance of the PDA/GO composite in pluripotent embryonic stem cells (ESCs) was investigated. The surface chemistry of the PDA/GO-coated PT surface was analyzed by scanning electron microscopy (SEM) and X-ray photoelectron spectroscopy (XPS). A high cell viability of ESCs cultured on the PDA/GO composite-coated surface was initially ensured. Then, the osteogenic differentiation of the ESCs in response to the PDA/GO substrate was assessed by alkaline phosphatase (ALP) activity, intracellular calcium levels, matrix mineralization assay, and evaluation of the mRNA and protein levels of osteogenic factors. The culture of ESCs on the PDA/GO substrate presented higher osteogenic potency than that on the uncoated control surface. ESCs cultured on the PDA/GO substrate expressed significantly higher levels of integrin α5 and β1, as well as bone morphogenetic protein receptor (BMPR) types I and II, compared with the control groups. The phosphorylation of extracellular signal-regulated kinase (ERK)1/2, p38, and c-Jun-N-terminal kinase (JNK) mitogen-activated protein kinases (MAPKs) was observed in ESCs culture on the PDA/GO substrate. Moreover, BMP signal transduction by SMAD1/5/8 phosphorylation was increased more in cells on PDA/GO than in the control. The nuclear translocation of SMAD1/5/8 in cells was also processed in response to the PDA/GO substrate. Blocking activation of the integrin α5/β1, MAPK, or SMAD signaling pathways downregulated the PDA/GO-induced osteogenic differentiation of ESCs. These results suggest that the PDA/GO composite stimulates the osteogenic differentiation of ESCs via the integrin α5/β1, MAPK, and BMPR/SMAD signaling pathways.
Collapse
|
12
|
Fukunaga I, Oe Y, Chen C, Danzaki K, Ohta S, Koike A, Ikeda K, Kamiya K. Activin/Nodal/TGF-β Pathway Inhibitor Accelerates BMP4-Induced Cochlear Gap Junction Formation During in vitro Differentiation of Embryonic Stem Cells. Front Cell Dev Biol 2021; 9:602197. [PMID: 33968919 PMCID: PMC8097046 DOI: 10.3389/fcell.2021.602197] [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: 09/02/2020] [Accepted: 03/30/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in gap junction beta-2 (GJB2), the gene that encodes connexin 26 (CX26), are the most frequent cause of hereditary deafness worldwide. We recently developed an in vitro model of GJB2-related deafness (induced CX26 gap junction-forming cells; iCX26GJCs) from mouse induced pluripotent stem cells (iPSCs) by using Bone morphogenetic protein 4 (BMP4) signaling-based floating cultures (serum-free culture of embryoid body-like aggregates with quick aggregation cultures; hereafter, SFEBq cultures) and adherent cultures. However, to use these cells as a disease model platform for high-throughput drug screening or regenerative therapy, cell yields must be substantially increased. In addition to BMP4, other factors may also induce CX26 gap junction formation. In the SFEBq cultures, the combination of BMP4 and the Activin/Nodal/TGF-β pathway inhibitor SB431542 (SB) resulted in greater production of isolatable CX26-expressing cell mass (CX26+ vesicles) and higher Gjb2 mRNA levels than BMP4 treatment alone, suggesting that SB may promote BMP4-mediated production of CX26+ vesicles in a dose-dependent manner, thereby increasing the yield of highly purified iCX26GJCs. This is the first study to demonstrate that SB accelerates BMP4-induced iCX26GJC differentiation during stem cell floating culture. By controlling the concentration of SB supplementation in combination with CX26+ vesicle purification, large-scale production of highly purified iCX26GJCs suitable for high-throughput drug screening or regenerative therapy for GJB2-related deafness may be possible.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kazusaku Kamiya
- Department of Otorhinolaryngology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Akbarzadeh A, Kianmanesh M, Fendereski K, Ebadi M, Daryabari SS, Masoomi A, Ghazisaeedi F, Seyyed Hossein Beigi R, Sheikh R, Kajbafzadeh AM. Decellularised whole ovine testis as a potential bio-scaffold for tissue engineering. Reprod Fertil Dev 2020; 31:1665-1673. [PMID: 31217071 DOI: 10.1071/rd19070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to determine an efficient whole-organ decellularisation protocol of a human-sized testis by perfusion through the testicular arteries. In the first step of this study, we determined the most efficient detergent agent, whereas the second phase delineated the optimal time required for the decellularisation process. Initially sheep testes were decellularised by one of three different detergent agents: sodium dodecyl sulphate (SDS), Triton X-100 and trypsin-ethylenediamine tetraacetic acid (EDTA) solutions, each perfused for 6h. In the second phase, the selected detergent agent was applied for different time periods. A total number of 20 organs were processed during this investigation. The efficacy of the decellularisation process and the preservation of the extracellular matrix components and structure were evaluated by histopathological examinations, 4',6'-diamidino-2-phenylindole (DAPI) staining, DNA quantification, hydroxyproline measurement, magnetic resonance imaging and scanning electron microscopy. Organ perfusion with 1% SDS solution for 6 to 8h demonstrated the most desirable outcomes regarding decellularisation and extracellular matrix preservation. The 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay was used to determine the toxicity of the scaffold and its potential for further application in tissue-engineering investigations. This investigation introduces an efficient method to produce a three-dimensional testicular bio-scaffold resembling the properties of the native organ that could be employed in tissue-engineering studies.
Collapse
Affiliation(s)
- Aram Akbarzadeh
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Maral Kianmanesh
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Kiarad Fendereski
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Maryam Ebadi
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Seyedeh Sima Daryabari
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Ahmad Masoomi
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Fereshteh Ghazisaeedi
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Reza Seyyed Hossein Beigi
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Reyhaneh Sheikh
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Paediatric Urology and Regenerative Medicine Research Centre, Children's Medical Centre, Tehran University of Medical Sciences, No. 62, Dr. Gharib Street, Keshavarz Boulevard, Tehran, 1419733151, Iran; and Corresponding author.
| |
Collapse
|
14
|
Sun S, Shi H, Moore S, Wang C, Ash-Shakoor A, Mather PT, Henderson JH, Ma Z. Progressive Myofibril Reorganization of Human Cardiomyocytes on a Dynamic Nanotopographic Substrate. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21450-21462. [PMID: 32326701 DOI: 10.1021/acsami.0c03464] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cardiomyocyte (CM) alignment with striated myofibril organization is developed during early cardiac organogenesis. Previous work has successfully achieved in vitro CM alignment using a variety of biomaterial scaffolds and substrates with static topographic features. However, the cellular processes that occur during the response of CMs to dynamic surface topographic changes, which may provide a model of in vivo developmental progress of CM alignment within embryonic myocardium, remains poorly understood. To gain insights into these cellular processes involved in the response of CMs to dynamic topographic changes, we developed a dynamic topographic substrate that employs a shape memory polymer coated with polyelectrolyte multilayers to produce a flat-to-wrinkle surface transition when triggered by a change in incubation temperature. Using this system, we investigated cellular morphological alignment and intracellular myofibril reorganization in response to the dynamic wrinkle formation. Hence, we identified the progressive cellular processes of human-induced pluripotent stem cell-CMs in a time-dependent manner, which could provide a foundation for a mechanistic model of cardiac myofibril reorganization in response to extracellular microenvironment changes.
Collapse
Affiliation(s)
- Shiyang Sun
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Sarah Moore
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Ariel Ash-Shakoor
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Patrick T Mather
- Department of Chemical Engineering, Bucknell University, Lewisburg, Pennsylvania 17837, United States
| | - James H Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
15
|
Sevari SP, Shahnazi F, Chen C, Mitchell JC, Ansari S, Moshaverinia A. Bioactive glass‐containing hydrogel delivery system for osteogenic differentiation of human dental pulp stem cells. J Biomed Mater Res A 2019; 108:557-564. [DOI: 10.1002/jbm.a.36836] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Sevda P. Sevari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry University of California Los Angeles California
| | - Faezeh Shahnazi
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry University of California Los Angeles California
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine University of Pennsylvania Philadelphia Pennsylvania
| | - John C. Mitchell
- Biomaterial Research Center Midwestern University Glendale Arizona
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry University of California Los Angeles California
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry University of California Los Angeles California
- California NanoSystems Institute University of California Los Angeles California
- Center for Minimally Invasive Therapeutics (C‐MIT) University of California Los Angeles California
| |
Collapse
|
16
|
Yamamoto T, Hatabayashi K, Arita M, Yajima N, Takenaka C, Suzuki T, Takahashi M, Oshima Y, Hara K, Kagawa K, Kawamata S. Kynurenine signaling through the aryl hydrocarbon receptor maintains the undifferentiated state of human embryonic stem cells. Sci Signal 2019; 12:12/587/eaaw3306. [PMID: 31239324 DOI: 10.1126/scisignal.aaw3306] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kynurenine, which is generated from tryptophan by indoleamine 2,3-dioxygenase 1 (IDO1), binds to the aryl hydrocarbon receptor (AhR). Here, we report that kynurenine was produced by undifferentiated human embryonic stem cells (hESCs) and by induced pluripotent stem cells (iPSCs). In undifferentiated hESCs, kynurenine stimulated the AhR to promote the expression of self-renewal genes. The kynurenine-AhR complex also stimulated the expression of IDO1 and AHR, activating a positive feedback loop. Inhibition of IDO1 activity reduced the proliferation of undifferentiated ESCs but did not stimulate their differentiation. Substantial amounts of free kynurenine were present in the culture medium, providing a paracrine signal for maintenance of the undifferentiated state. Kynurenine was not present in the medium of differentiated ESCs or iPSCs. When ESCs were induced to undergo ectodermal differentiation, the abundance of kynurenine in the medium was reduced through activation of the main kynurenine catabolic pathway mediated by kynurenine aminotransferase 2 (KAT2, also known as AADAT), resulting in the secretion of 2-aminoadipic acid (2-AAA) into the culture medium. Inhibition of KAT2 activity blocked ectodermal differentiation. Thus, kynurenine metabolism plays an important role in the maintenance of the undifferentiated state and in ectodermal differentiation. Furthermore, kynurenine in the culture medium is a biomarker for the undifferentiated state, whereas the presence of 2-AAA in the culture medium is a biomarker of ESCs and iPSCs that have committed to differentiate along the ectoderm lineage.
Collapse
Affiliation(s)
- Takako Yamamoto
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation, 2-2 Minatojima Minami-machi, Chuo-ku, Kobe 650-0047, Japan
| | - Kunitada Hatabayashi
- Innovative Technology Planning Department, Tokyo Electron Limited, Akasaka Biz Tower, 5-3-1 Akasaka, Minato-Ku, Tokyo 107-6325, Japan
| | - Mao Arita
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation, 2-2 Minatojima Minami-machi, Chuo-ku, Kobe 650-0047, Japan
| | - Nobuyuki Yajima
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation, 2-2 Minatojima Minami-machi, Chuo-ku, Kobe 650-0047, Japan
| | - Chiemi Takenaka
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation, 2-2 Minatojima Minami-machi, Chuo-ku, Kobe 650-0047, Japan
| | - Takashi Suzuki
- Analytical and Measuring Instruments Division, Shimadzu Corporation, 1 Nishinokyo, Kuwahara-cho, Nagagyo-ku, Kyoto 604-8511, Japan
| | - Masatoshi Takahashi
- Analytical and Measuring Instruments Division, Shimadzu Corporation, 1 Nishinokyo, Kuwahara-cho, Nagagyo-ku, Kyoto 604-8511, Japan
| | - Yasuhiro Oshima
- Innovative Technology Planning Department, Tokyo Electron Limited, Akasaka Biz Tower, 5-3-1 Akasaka, Minato-Ku, Tokyo 107-6325, Japan
| | - Keisuke Hara
- Innovative Technology Planning Department, Tokyo Electron Limited, Akasaka Biz Tower, 5-3-1 Akasaka, Minato-Ku, Tokyo 107-6325, Japan
| | - Kenichi Kagawa
- Innovative Technology Planning Department, Tokyo Electron Limited, Akasaka Biz Tower, 5-3-1 Akasaka, Minato-Ku, Tokyo 107-6325, Japan
| | - Shin Kawamata
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation, 2-2 Minatojima Minami-machi, Chuo-ku, Kobe 650-0047, Japan. .,Riken Center for Developmental Biology, 2-1 Minatojima Minami-machi, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
17
|
Wen H, Xiao W, Biswas S, Cong ZQ, Liu XM, Lam KS, Liao YH, Deng W. Alginate Hydrogel Modified with a Ligand Interacting with α3β1 Integrin Receptor Promotes the Differentiation of 3D Neural Spheroids toward Oligodendrocytes in Vitro. ACS APPLIED MATERIALS & INTERFACES 2019; 11:5821-5833. [PMID: 30645095 DOI: 10.1021/acsami.8b19438] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this study, we established a long-term three-dimensional (3D) culture system by using integrin ligand modified alginate hydrogels to encapsulate and differentiate neural progenitor cells (NPCs) toward oligodendrocyte (OL) lineage cells. The porosity of the hydrogel was optimized by varying the alginate concentrations and then characterized by scanning electronic microscopy (SEM). The surface plasmon resonance (SPR) test was used to confirm the ligand-integrin interactions indicating adherence between the NPC surfaces and the hydrogels. Following encapsulation in the hydrogels, both mouse and human NPC sphere cultures could be maintained up to 90 days. Mouse NPC spheres were differentiated into viable neurons, astrocytes and mature OLs by day 60 in all groups whereas human NPC spheres were differentiated into neurons and later into GFAP positive astrocytes and O4 positive pre-OL within 90 days. The species difference in the timeline of OL development between mouse and human was reflected in this system. The ligand LXY30 interacting with the α3β1 integrin receptor was more effective in promoting the differentiation of hNPCs to OL lineage cells compared with the ligand LXW64 interacting with the αvβ3 integrin receptor, hyaluronic acid interacting with CD44 receptor or without any ligand. This study is the first to differentiate O4+ pre-OLs from hNPCs in a LXY30-α3β1 (integrin-ligand) modified alginate 3D hydrogel culture. This 3D platform could serve as a valuable tool in disease modeling, drug discovery, and NPC transplantation.
Collapse
Affiliation(s)
- Han Wen
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
| | - Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
- School of Pharmaceutical Sciences, Shenzhen Campus , Sun Yat-Sen University , 135 Xingang Xi Road , Guangzhou 510275 , China
| | - Zhao-Qing Cong
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Xin-Min Liu
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
| | - Yong-Hong Liao
- Institute of Medicinal Plant Development , Chinese Academy of Medical Sciences and Peking Union Medical College , 151 Malianwa North Road , Haidian District, Beijing 100193 , China
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine , University of California , 2700 Stockton Blvd , Davis , California 95817 , United States
- Institute for Pediatric Regenerative Medicine , Shriners Hospitals for Children-Northern California , 2425 Stockton Blvd , Sacramento , California 95817 , United States
- School of Pharmaceutical Sciences, Shenzhen Campus , Sun Yat-Sen University , 135 Xingang Xi Road , Guangzhou 510275 , China
| |
Collapse
|
18
|
Kwon SG, Kwon YW, Lee TW, Park GT, Kim JH. Recent advances in stem cell therapeutics and tissue engineering strategies. Biomater Res 2018; 22:36. [PMID: 30598836 PMCID: PMC6299977 DOI: 10.1186/s40824-018-0148-4] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/07/2018] [Indexed: 12/19/2022] Open
Abstract
Background Tissue regeneration includes delivering specific types of cells or cell products to injured tissues or organs for restoration of tissue and organ function. Stem cell therapy has drawn considerable attention since transplantation of stem cells can overcome the limitations of autologous transplantation of patient’s tissues; however, it is not perfect for treating diseases. To overcome the hurdles associated with stem cell therapy, tissue engineering techniques have been developed. Development of stem cell technology in combination with tissue engineering has opened new ways of producing engineered tissue substitutes. Several studies have shown that this combination of tissue engineering and stem cell technologies enhances cell viability, differentiation, and therapeutic efficacy of transplanted stem cells. Main body Stem cells that can be used for tissue regeneration include mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells. Transplantation of stem cells alone into injured tissues exhibited low therapeutic efficacy due to poor viability and diminished regenerative activity of transplanted cells. In this review, we will discuss the progress of biomedical engineering, including scaffolds, biomaterials, and tissue engineering techniques to overcome the low therapeutic efficacy of stem cells and to treat human diseases. Conclusion The combination of stem cell and tissue engineering techniques overcomes the limitations of stem cells in therapy of human diseases, and presents a new path toward regeneration of injured tissues.
Collapse
Affiliation(s)
- Seong Gyu Kwon
- 1Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612 Gyeongsangnam-do Republic of Korea
| | - Yang Woo Kwon
- 1Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612 Gyeongsangnam-do Republic of Korea
| | - Tae Wook Lee
- 1Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612 Gyeongsangnam-do Republic of Korea
| | - Gyu Tae Park
- 1Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612 Gyeongsangnam-do Republic of Korea
| | - Jae Ho Kim
- 1Department of Physiology, Pusan National University School of Medicine, Yangsan, 50612 Gyeongsangnam-do Republic of Korea.,2Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, 50612 Republic of Korea
| |
Collapse
|
19
|
Shibata S, Hayashi R, Okubo T, Kudo Y, Katayama T, Ishikawa Y, Toga J, Yagi E, Honma Y, Quantock AJ, Sekiguchi K, Nishida K. Selective Laminin-Directed Differentiation of Human Induced Pluripotent Stem Cells into Distinct Ocular Lineages. Cell Rep 2018; 25:1668-1679.e5. [DOI: 10.1016/j.celrep.2018.10.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 09/10/2018] [Accepted: 10/05/2018] [Indexed: 12/22/2022] Open
|
20
|
Bover O, Justo T, Pereira PNG, Facucho-Oliveira J, Inácio JM, Ramalho JS, Domian IJ, Belo JA. Loss of Ccbe1 affects cardiac-specification and cardiomyocyte differentiation in mouse embryonic stem cells. PLoS One 2018; 13:e0205108. [PMID: 30281646 PMCID: PMC6169972 DOI: 10.1371/journal.pone.0205108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 09/19/2018] [Indexed: 01/27/2023] Open
Abstract
Understanding the molecular pathways regulating cardiogenesis is crucial for the early diagnosis of heart diseases and improvement of cardiovascular disease. During normal mammalian cardiac development, collagen and calcium-binding EGF domain-1 (Ccbe1) is expressed in the first and second heart field progenitors as well as in the proepicardium, but its role in early cardiac commitment remains unknown. Here we demonstrate that during mouse embryonic stem cell (ESC) differentiation Ccbe1 is upregulated upon emergence of Isl1- and Nkx2.5- positive cardiac progenitors. Ccbe1 is markedly enriched in Isl1-positive cardiac progenitors isolated from ESCs differentiating in vitro or embryonic hearts developing in vivo. Disruption of Ccbe1 activity by shRNA knockdown or blockade with a neutralizing antibody results in impaired differentiation of embryonic stem cells along the cardiac mesoderm lineage resulting in a decreased expression of mature cardiomyocyte markers. In addition, knockdown of Ccbe1 leads to smaller embryoid bodies. Collectively, our results show that CCBE1 is essential for the commitment of cardiac mesoderm and consequently, for the formation of cardiac myocytes in differentiating mouse ESCs.
Collapse
Affiliation(s)
- Oriol Bover
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Tiago Justo
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
- Center for Biomedical Research, Campus de Gambelas, University of Algarve, Faro, Portugal
| | - Paulo N. G. Pereira
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - João Facucho-Oliveira
- Center for Biomedical Research, Campus de Gambelas, University of Algarve, Faro, Portugal
| | - José M. Inácio
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - José S. Ramalho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ibrahim J. Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Cambridge, Massachusetts, United States of America
| | - José António Belo
- Stem Cells and Development Laboratory, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
- * E-mail:
| |
Collapse
|
21
|
Kajbafzadeh AM, Khorramirouz R, Masoumi A, Keihani S, Nabavizadeh B. Decellularized human fetal intestine as a bioscaffold for regeneration of the rabbit bladder submucosa. J Pediatr Surg 2018; 53:1781-1788. [PMID: 29459044 DOI: 10.1016/j.jpedsurg.2018.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/15/2017] [Accepted: 01/26/2018] [Indexed: 10/18/2022]
Abstract
PURPOSE We aim to report a method to create a natural acellular scaffold from human fetal small intestine for augmentation cystoplasty in rabbits. METHODS Fetal intestines were decellularized by immersion in a hypotonic solution. The success of this protocol was evaluated by histological analysis, scanning electron microscopy and measurement of collagen and sulfated glycosaminoglycan of the acellular tissues. Eight mature rabbits were selected and acellular scaffolds were implanted on the exposed urothelium. Urodynamic studies and cystography were performed after six months. At 14, 120 and 180days animals were sacrificed and augmented bladders were resected. RESULTS Histological analysis revealed formation of muscular layer and blood vessels in implanted scaffolds similar to normal bladder. These findings demonstrate the effective seeding of scaffold by host bladder cells. The tissue architecture of recellularized scaffold was similar to the native bladder. CONCLUSIONS Fetal intestine acellular matrix could be an exceptional scaffold for bladder augmentation cystoplasty and may pave the road for future studies in order to be used for clinical application.
Collapse
Affiliation(s)
- Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Khorramirouz
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Masoumi
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorena Keihani
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Nabavizadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Farhat W, Hasan A, Lucia L, Becquart F, Ayoub A, Kobeissy F. Hydrogels for Advanced Stem Cell Therapies: A Biomimetic Materials Approach for Enhancing Natural Tissue Function. IEEE Rev Biomed Eng 2018; 12:333-351. [PMID: 29993840 DOI: 10.1109/rbme.2018.2824335] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Stem-cell-based therapy is a promising approach for the treatment of a myriad of diseases and injuries. However, the low rate of cell survival and the uncontrolled differentiation of the injected stem cells currently remain key challenges in advancing stem cell therapeutics. Hydrogels are biomaterials that are potentially highly effective candidates for scaffold systems for stem cells and other molecular encapsulation approaches to target in vivo delivery. Hydrogel-based strategies can potentially address several current challenges in stem cell therapy. We present a concise overview of the recent advances in applications of hydrogels in stem cell therapies, with a focus particularly on the recent advances in the design and approaches for application of hydrogels in tissue engineering. The capability of hydrogels to either enhance the function of the transplanted stem cells by promoting their controlled differentiation or enhance the recruitment of endogenous adult stem cells to the injury site for repair is also reviewed. Finally, the importance of impacts and the desired relationship between the scaffold system and the encapsulated stem cells are discussed.
Collapse
|
23
|
Nojehdehian H, Moztarzadeh F, Baharvand H, Mehrjerdi NZ, Nazarian H, Tahriri M. Effect of Poly-L-lysine Coating on Retinoic Acid-Loaded PLGA Microspheres in the Differentiation of Carcinoma Stem Cells into Neural Cells. Int J Artif Organs 2018. [DOI: 10.1177/039139881003301005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this study, PLGA microspheres were prepared using a water-in-oil-in-water emulsion/solvent evaporation technique. Some microspheres were coated with poly-L-lysine (an extracellular matrix (ECM) component), and then pluripotent P19 embryonic carcinoma cells were seeded on them. P19 cells attached onto the PLGA microspheres; subsequently, by adding retinoic acid (RA) to cell culture medium as a neurogenic inducer (RA was released from the microspheres), the cells differentiated into neural cells. Size and morphology of PLGA microspheres was characterized by scanning electron microscopy (SEM). Neurogenic differentiation was studied by immunofluorescent staining, real-time polymerase chain reaction (RT-PCR), and light microscopy. Histological assay showed that more cells attached onto microspheres coated with poly-L-lysine than the uncoated group. Immunofluoresent staining and RT-PCR analysis for β-Tubulin, Nestin and Pax6 genes indicated differentiation of P19 cells into neural cells on both coated and uncoated microspheres. It was found that a high surface area of microspheres improves cell attachment and expansion, which was significantly increased in those coated with poly-L-lysine. Finally, these results highlight the versatility of these sample scaffolds as a model system for nerve tissue engineering.
Collapse
Affiliation(s)
- Hanieh Nojehdehian
- Stem Cell Department, Cell Science Research Center, Royan Institute, Tehran - Iran
- Biomaterials Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran - Iran
- Faculty of Biomedical Engineering, Azad University, Tehran - Iran
| | - Fathollah Moztarzadeh
- Biomaterials Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran - Iran
| | - Hossein Baharvand
- Stem Cell Department, Cell Science Research Center, Royan Institute, Tehran - Iran
| | | | - Hamid Nazarian
- Stem Cell Department, Cell Science Research Center, Royan Institute, Tehran - Iran
| | - Mohammadreza Tahriri
- Biomaterials Group, Faculty of Biomedical Engineering (Center of Excellence), Amirkabir University of Technology, Tehran - Iran
| |
Collapse
|
24
|
Rosellini E, Zhang YS, Migliori B, Barbani N, Lazzeri L, Shin SR, Dokmeci MR, Cascone MG. Protein/polysaccharide-based scaffolds mimicking native extracellular matrix for cardiac tissue engineering applications. J Biomed Mater Res A 2018; 106:769-781. [PMID: 29052369 PMCID: PMC5845858 DOI: 10.1002/jbm.a.36272] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 11/07/2022]
Abstract
Tissue engineering has emerged as a viable approach to treat disease or repair damage in tissues and organs. One of the key elements for the success of tissue engineering is the use of a scaffold serving as artificial extracellular matrix (ECM). The ECM hosts the cells and improves their survival, proliferation, and differentiation, enabling the formation of new tissue. Here, we propose the development of a class of protein/polysaccharide-based porous scaffolds for use as ECM substitutes in cardiac tissue engineering. Scaffolds based on blends of a protein component, collagen or gelatin, with a polysaccharide component, alginate, were produced by freeze-drying and subsequent ionic and chemical crosslinking. Their morphological, physicochemical, and mechanical properties were determined and compared with those of natural porcine myocardium. We demonstrated that our scaffolds possessed highly porous and interconnected structures, and the chemical homogeneity of the natural ECM was well reproduced in both types of scaffolds. Furthermore, the alginate/gelatin (AG) scaffolds better mimicked the native tissue in terms of interactions between components and protein secondary structure, and in terms of swelling behavior. The AG scaffolds also showed superior mechanical properties for the desired application and supported better adhesion, growth, and differentiation of myoblasts under static conditions. The AG scaffolds were subsequently used for culturing neonatal rat cardiomyocytes, where high viability of the resulting cardiac constructs was observed under dynamic flow culture in a microfluidic bioreactor. We therefore propose our protein/polysaccharide scaffolds as a viable ECM substitute for applications in cardiac tissue engineering. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 769-781, 2018.
Collapse
Affiliation(s)
- Elisabetta Rosellini
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Bianca Migliori
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Niccoletta Barbani
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| | - Luigi Lazzeri
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Mehmet Remzi Dokmeci
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139
| | - Maria Grazia Cascone
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino, Pisa, 56126, Italy
| |
Collapse
|
25
|
Abstract
In this chapter we illustrate protocols to investigate growth and neurotrophic factors in human and rodent (rat and mouse)-derived embryonic stem cells. The conventional two-dimensional cell monolayer system to grow embryonic stem cells is presented, focusing on the coating strategies also using extracellular matrix components. Then, different approaches for three-dimensional stem cell culture are presented, using hydrogels and scaffolds. Quantitative polymerase chain reaction, immunocytochemistry, immunoenzymatic ELISA assay, and multiparametric assays to quantify growth and neurotrophic factor production are presented.
Collapse
|
26
|
Madaboosi N, Uhlig K, Schmidt S, Vikulina AS, Möhwald H, Duschl C, Volodkin D. A “Cell-Friendly” Window for the Interaction of Cells with Hyaluronic Acid/Poly-l
-Lysine Multilayers. Macromol Biosci 2017; 18. [DOI: 10.1002/mabi.201700319] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/27/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Narayanan Madaboosi
- Fraunhofer Institute for Cell Therapy and Immunology; Branch Bioanalytics and Bioprocesses (Fraunhofer IZI-BB); Department Cellular Biotechnology & Biochips; Am Mühlenberg 13 14476 Potsdam-Golm Germany
- Max Planck Institute for Colloids and Interfaces; Am Mühlenberg 1 14476 Potsdam-Golm Germany
| | - Katja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology; Branch Bioanalytics and Bioprocesses (Fraunhofer IZI-BB); Department Cellular Biotechnology & Biochips; Am Mühlenberg 13 14476 Potsdam-Golm Germany
| | - Stephan Schmidt
- Fraunhofer Institute for Cell Therapy and Immunology; Branch Bioanalytics and Bioprocesses (Fraunhofer IZI-BB); Department Cellular Biotechnology & Biochips; Am Mühlenberg 13 14476 Potsdam-Golm Germany
- Heinrich-Heine-Universität Düsseldorf; Institut für Organische und Makromolekulare Chemie; Universiätsstr.1 40225 Düsseldorf Germany
| | - Anna S. Vikulina
- School of Science and Technology; Nottingham Trent University; Clifton Lane Nottingham NG11 8NS UK
| | - Helmuth Möhwald
- Max Planck Institute for Colloids and Interfaces; Am Mühlenberg 1 14476 Potsdam-Golm Germany
| | - Claus Duschl
- Fraunhofer Institute for Cell Therapy and Immunology; Branch Bioanalytics and Bioprocesses (Fraunhofer IZI-BB); Department Cellular Biotechnology & Biochips; Am Mühlenberg 13 14476 Potsdam-Golm Germany
| | - Dmitry Volodkin
- Fraunhofer Institute for Cell Therapy and Immunology; Branch Bioanalytics and Bioprocesses (Fraunhofer IZI-BB); Department Cellular Biotechnology & Biochips; Am Mühlenberg 13 14476 Potsdam-Golm Germany
- School of Science and Technology; Nottingham Trent University; Clifton Lane Nottingham NG11 8NS UK
| |
Collapse
|
27
|
Ansari S, Sarrion P, Hasani-Sadrabdi MM, Aghaloo T, Wu BM, Moshaverinia A. Regulation of the fate of dental-derived mesenchymal stem cells using engineered alginate-GelMA hydrogels. J Biomed Mater Res A 2017; 105:2957-2967. [PMID: 28639378 PMCID: PMC5623163 DOI: 10.1002/jbm.a.36148] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 11/11/2022]
Abstract
Mesenchymal stem cells (MSCs) derived from dental and orofacial tissues provide an alternative therapeutic option for craniofacial bone tissue regeneration. However, there is still a need to improve stem cell delivery vehicles to regulate the fate of the encapsulated MSCs for high quality tissue regeneration. Matrix elasticity plays a vital role in MSC fate determination. Here, we have prepared various hydrogel formulations based on alginate and gelatin methacryloyl (GelMA) and have encapsulated gingival mesenchymal stem cells (GMSCs) and human bone marrow MSCs (hBMMSCs) within these fabricated hydrogels. We demonstrate that addition of the GelMA to alginate hydrogel reduces the elasticity of the hydrogel mixture. While presence of GelMA in an alginate-based scaffold significantly increased the viability of encapsulated MSCs, increasing the concentration of GelMA downregulated the osteogenic differentiation of encapsulated MSCs in vitro due to decrease in the stiffness of the hydrogel matrix. The osteogenic suppression was rescued by addition of a potent osteogenic growth factor such as rh-BMP-2. In contrast, MSCs encapsulated in alginate hydrogel without GelMA were successfully osteo-differentiated without the aid of additional growth factors, as confirmed by expression of osteogenic markers (Runx2 and OCN), as well as positive staining using Xylenol orange. Interestingly, after two weeks of osteo-differentiation, hBMMSCs and GMSCs encapsulated in alginate/GelMA hydrogels still expressed CD146, an MSC surface marker, while MSCs encapsulated in alginate hydrogel failed to express any positive staining. Altogether, our findings suggest that it is possible to control the fate of encapsulated MSCs within hydrogels by tuning the mechanical properties of the matrix. We also reconfirmed the important role of the presence of inductive signals in guiding MSC differentiation. These findings may enable the design of new multifunctional scaffolds for spatial and temporal control over the fate and function of stem cells even post-transplantation. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2957-2967, 2017.
Collapse
Affiliation(s)
- Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA
| | - Patricia Sarrion
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA
| | - Mohammad Mahdi Hasani-Sadrabdi
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA
- Parker H. Petit Institute for Bioengineering and Bioscience, G. W. Woodruff School of Mechanical Engineering and School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA
| | - Benjamin M Wu
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA
| |
Collapse
|
28
|
Ansari S, Diniz IM, Chen C, Aghaloo T, Wu BM, Shi S, Moshaverinia A. Alginate/hyaluronic acid hydrogel delivery system characteristics regulate the differentiation of periodontal ligament stem cells toward chondrogenic lineage. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:162. [PMID: 28914392 DOI: 10.1007/s10856-017-5974-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/01/2017] [Indexed: 06/07/2023]
Abstract
Cartilage tissue regeneration often presents a challenging clinical situation. Recently, it has been shown that Periodontal Ligament Stem Cells (PDLSCs) possess high chondrogenic differentiation capacity. In this study, we developed a stem cell delivery system based on alginate/hyaluronic acid (HA) loaded with TGF-β1 ligand, encapsulating PDLSCs; and investigated the chondrogenic differentiation of encapsulated cells in alginate/HA hydrogel microspheres in vitro and in vivo. The results showed that PDLSCs, as well as human bone marrow mesenchymal stem cells (hBMMSCs), as the positive control, were stained positive for both toluidine blue and alcian blue staining, while exhibiting high levels of gene expression related to chondrogenesis (Col II, Aggrecan and Sox-9), as assessed via qPCR. The quantitative PCR analyses exhibited that the chondrogenic differentiation of encapsulated MSCs can be regulated by the modulus of elasticity of hydrogel delivery system, confirming the vital role of the microenvironment, and the presence of inductive signals for viability and differentiation of MSCs. In vivo, histological and immunofluorescence staining for chondrogenic specific protein markers confirmed ectopic cartilage-like tissue regeneration inside transplanted hydrogels. PDLSCs presented significantly greater capability for chondrogenic differentiation than hBMMSCs (P < 0.05). Altogether, our findings confirmed that alginate/HA hydrogels encapsulating PDLSCs are a promising candidate for cartilage regeneration.
Collapse
Affiliation(s)
- Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Ivana M Diniz
- Departamento de Odontologia Restauradora, Faculdade de Odontologia da UFMG, Belo Horizonte, Brazil
| | - Chider Chen
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Benjamin M Wu
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Songtao Shi
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, CA, USA.
| |
Collapse
|
29
|
Huma T, Hu X, Ma Y, Willden A, Rizak J, Shahab M, Wang Z. Kisspeptin-10 treatment generated specific GnRH expression in cells differentiated from rhesus monkey derived Lyon NSCs. Neuroscience 2017; 349:318-329. [DOI: 10.1016/j.neuroscience.2017.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 01/01/2023]
|
30
|
Collagen Sponge Functionalized with Chimeric Anti-BMP-2 Monoclonal Antibody Mediates Repair of Critical-Size Mandibular Continuity Defects in a Nonhuman Primate Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8094152. [PMID: 28401163 PMCID: PMC5376406 DOI: 10.1155/2017/8094152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/05/2017] [Accepted: 01/19/2017] [Indexed: 11/25/2022]
Abstract
Antibody-mediated osseous regeneration (AMOR) has been introduced by our research group as a tissue engineering approach to capture of endogenous growth factors through the application of specific monoclonal antibodies (mAbs) immobilized on a scaffold. Specifically, anti-Bone Morphogenetic Protein- (BMP-) 2 mAbs have been demonstrated to be efficacious in mediating bone repair in a number of bone defects. The present study sought to investigate the application of AMOR for repair of mandibular continuity defect in nonhuman primates. Critical-sized mandibular continuity defects were created in Macaca fascicularis locally implanted with absorbable collagen sponges (ACS) functionalized with chimeric anti-BMP-2 mAb or isotype control mAb. 2D and 3D analysis of cone beam computed tomography (CBCT) imaging demonstrated increased bone density and volume observed within mandibular continuity defects implanted with collagen scaffolds functionalized with anti-BMP-2 mAb, compared with isotype-matched control mAb. Both CBCT imaging and histologic examination demonstrated de novo bone formation that was in direct apposition to the margins of the resected bone. It is hypothesized that bone injury may be necessary for AMOR. This is evidenced by de novo bone formation adjacent to resected bone margins, which may be the source of endogenous BMPs captured by anti-BMP-2 mAb, in turn mediating bone repair.
Collapse
|
31
|
Dzobo K, Vogelsang M, Parker MI. Wnt/β-Catenin and MEK-ERK Signaling are Required for Fibroblast-Derived Extracellular Matrix-Mediated Endoderm Differentiation of Embryonic Stem Cells. Stem Cell Rev Rep 2016; 11:761-73. [PMID: 26022506 DOI: 10.1007/s12015-015-9598-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human embryonic stem cells (hESCs) have the potential to differentiate into all cells of the three germ layers, thus making them an attractive source of cells for use in regenerative medicine. The greatest challenge lies in regulating the differentiation of hESCs into specific cell lineages by both intrinsic and extrinsic factors. In this study we determined the effect of a fibroblast-derived extracellular matrix (fd-ECM) on hESCs differentiation. We demonstrate that growth of hESCs on fd-ECM results in hESCs losing their stemness and proliferation potential. As the stem cells differentiate they attain gene expression profiles similar to the primitive streak of the in vivo embryo. The activation of both the MEK-ERK and Wnt/β-catenin signaling pathways is required for the fd-ECM-mediated differentiation of hESCs towards the endoderm and involves integrins α1, α2, α3 and β1. This study illustrates the importance of the cellular microenvironment in directing stem cell fate and that the nature and composition of the extracellular matrix is a crucial determining factor.
Collapse
Affiliation(s)
- Kevin Dzobo
- Cape Town Component, Wernher and Beit Building (South), UCT Campus, International Centre for Genetic Engineering and Biotechnology (ICGEB), Anzio Road, Observatory, 7925, Cape Town, South Africa
| | | | | |
Collapse
|
32
|
Dehdilani N, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Amoughli Tabrizi B, Parsa H, Sabagi F. Improved Survival and Hematopoietic Differentiation of Murine Embryonic Stem Cells on Electrospun Polycaprolactone Nanofiber. CELL JOURNAL 2016; 17:629-38. [PMID: 26862522 PMCID: PMC4746413 DOI: 10.22074/cellj.2016.3835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 11/13/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Three-dimensional (3D) biomimetic nanofiber scaffolds have widespread ap- plications in biomedical tissue engineering. They provide a suitable environment for cel- lular adhesion, survival, proliferation and differentiation, guide new tissue formation and development, and are one of the outstanding goals of tissue engineering. Electrospinning has recently emerged as a leading technique for producing biomimetic scaffolds with mi- cro to nanoscale topography and a high porosity similar to the natural extracellular matrix (ECM). These scaffolds are comprised of synthetic and natural polymers for tissue engi- neering applications. Several kinds of cells such as human embryonic stem cells (hESCs) and mouse ESCs (mESCs) have been cultured and differentiated on nanofiber scaffolds. mESCs can be induced to differentiate into a particular cell lineage when cultured as em- bryoid bodies (EBs) on nano-sized scaffolds. MATERIALS AND METHODS We cultured mESCs (2500 cells/100 µl) in 96-well plates with knockout Dulbecco's modified eagle medium (DMEM-KO) and Roswell Park Memorial Institute-1640 (RPMI-1640), both supplemented with 20% ESC grade fetal bovine serum (FBS) and essential factors in the presence of leukemia inhibitory factor (LIF). mESCs were seeded at a density of 2500 cells/100 µl onto electrospun polycaprolactone (PCL) nanofibers in 96-well plates. The control group comprised mESCs grown on tissue cul- ture plates (TCP) at a density of 2500 cells/100 µl. Differentiation of mESCs into mouse hematopoietic stem cells (mHSCs) was performed by stem cell factor (SCF), interleukin-3 (IL-3), IL-6 and Fms-related tyrosine kinase ligand (Flt3-L) cytokines for both the PCL and TCP groups. We performed an experimental study of mESCs differentiation. RESULTS PCL was compared to conventional TCP for survival and differentiation of mESCs to mHSCs. There were significantly more mESCs in the PCL group. Flowcyto- metric analysis revealed differences in hematopoietic differentiation between the PCL and TCP culture systems. There were more CD34+(Sca1+) and CD133+cells subpopulations in the PCL group compared to the conventional TCP culture system. CONCLUSION The nanofiber scaffold, as an effective surface, improves survival and differentiation of mESCs into mHSCs compared to gelatin coated TCP. More studies are necessary to understand how the topographical features of electrospun fibers af- fect cell growth and behavior. This can be achieved by designing biomimetic scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Nima Dehdilani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahram Amoughli Tabrizi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Hamed Parsa
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabagi
- Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
33
|
Blaber EA, Finkelstein H, Dvorochkin N, Sato KY, Yousuf R, Burns BP, Globus RK, Almeida EAC. Microgravity Reduces the Differentiation and Regenerative Potential of Embryonic Stem Cells. Stem Cells Dev 2015; 24:2605-21. [PMID: 26414276 PMCID: PMC4652210 DOI: 10.1089/scd.2015.0218] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mechanical unloading in microgravity is thought to induce tissue degeneration by various mechanisms, including inhibition of regenerative stem cell differentiation. To address this hypothesis, we investigated the effects of microgravity on early lineage commitment of mouse embryonic stem cells (mESCs) using the embryoid body (EB) model of tissue differentiation. We found that exposure to microgravity for 15 days inhibits mESC differentiation and expression of terminal germ layer lineage markers in EBs. Additionally, microgravity-unloaded EBs retained stem cell self-renewal markers, suggesting that mechanical loading at Earth's gravity is required for normal differentiation of mESCs. Finally, cells recovered from microgravity-unloaded EBs and then cultured at Earth's gravity showed greater stemness, differentiating more readily into contractile cardiomyocyte colonies. These results indicate that mechanical unloading of stem cells in microgravity inhibits their differentiation and preserves stemness, possibly providing a cellular mechanistic basis for the inhibition of tissue regeneration in space and in disuse conditions on earth.
Collapse
Affiliation(s)
- Elizabeth A Blaber
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California.,2 School of Biotechnology and Biomolecular Sciences, University of New South Wales , Sydney, Australia
| | - Hayley Finkelstein
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Natalya Dvorochkin
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Kevin Y Sato
- 3 FILMSS Wyle, Space Biology, NASA Ames Research Center , Moffett Field, California
| | - Rukhsana Yousuf
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Brendan P Burns
- 2 School of Biotechnology and Biomolecular Sciences, University of New South Wales , Sydney, Australia .,4 Australian Centre for Astrobiology, University of New South Wales , Sydney, Australia
| | - Ruth K Globus
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| | - Eduardo A C Almeida
- 1 Space Biosciences Division, NASA Ames Research Center , Moffett Field, California
| |
Collapse
|
34
|
Johnson MD, Reeder JE, O’Connell M. Bone morphogenetic protein-4 and 7 and receptors regulate vascular endothelial growth factor and receptors in human fetal leptomeninges. Neurosci Lett 2015; 606:225-30. [DOI: 10.1016/j.neulet.2015.08.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/09/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
|
35
|
Wang H, Luo X, Leighton J. Extracellular Matrix and Integrins in Embryonic Stem Cell Differentiation. BIOCHEMISTRY INSIGHTS 2015; 8:15-21. [PMID: 26462244 PMCID: PMC4589090 DOI: 10.4137/bci.s30377] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 12/17/2022]
Abstract
Embryonic stem cells (ESCs) are pluripotent cells with great therapeutic potentials. The in vitro differentiation of ESC was designed by recapitulating embryogenesis. Significant progress has been made to improve the in vitro differentiation protocols by toning soluble maintenance factors. However, more robust methods for lineage-specific differentiation and maturation are still under development. Considering the complexity of in vivo embryogenesis environment, extracellular matrix (ECM) cues should be considered besides growth factor cues. ECM proteins bind to cells and act as ligands of integrin receptors on cell surfaces. Here, we summarize the role of the ECM and integrins in the formation of three germ layer progenies. Various ECM–integrin interactions were found, facilitating differentiation toward definitive endoderm, hepatocyte-like cells, pancreatic beta cells, early mesodermal progenitors, cardiomyocytes, neuroectoderm lineages, and epidermal cells, such as keratinocytes and melanocytes. In the future, ECM combinations for the optimal ESC differentiation environment will require substantial study.
Collapse
Affiliation(s)
- Han Wang
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xie Luo
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jake Leighton
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
36
|
zur Nieden NI, Turgman CC, Lang X, Larsen JM, Granelli J, Hwang YJ, Lyubovitsky JG. Fluorescent hydrogels for embryoid body formation and osteogenic differentiation of embryonic stem cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:10599-10605. [PMID: 25905907 DOI: 10.1021/acsami.5b02368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Substrate mechanics (e.g., stiffness and topography of the microenvironment) are likely critical for driving normal morphogenesis and tissue development. As such, substrate mechanics imposed by hydrogels have been exploited to guide the lineage differentiation of stem cells and to drive stemness. In this work, we chemically modified gelatin hydrogels through glyceraldehyde cross-linking to render them suitable for cell culture. The modified hydrogels proved to be ideal for embryonic stem cell osteogenesis, initially providing a soft nonadhesive surface for the formation of embryoid bodies. They subsequently degraded in culture to afford a harder surface during osteoblast differentiation. The gels synthesized are highly fluorescent, relatively easy to prepare, and can potentially aid in overcoming the challenge of imaging changes to the microenvironments of cells during three-dimensional cell culture. Exploiting these materials could lead to the development of tissue-engineered products of increased complexity and rational treatment strategies.
Collapse
|
37
|
Geuss LR, Wu DC, Ramamoorthy D, Alford CD, Suggs LJ. Paramagnetic beads and magnetically mediated strain enhance cardiomyogenesis in mouse embryoid bodies. PLoS One 2014; 9:e113982. [PMID: 25501004 PMCID: PMC4264692 DOI: 10.1371/journal.pone.0113982] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/25/2014] [Indexed: 01/16/2023] Open
Abstract
Mechanical forces play an important role in proper embryologic development, and similarly such forces can directly impact pluripotency and differentiation of mouse embryonic stem cells (mESC) in vitro. In addition, manipulation of the embryoid body (EB) microenvironment, such as by incorporation of microspheres or microparticles, can similarly influence fate determination. In this study, we developed a mechanical stimulation regimen using permanent neodymium magnets to magnetically attract cells within an EB. Arginine-Glycine-Aspartic Acid (RGD)-conjugated paramagnetic beads were incorporated into the interior of the EBs during aggregation, allowing us to exert force on individual cells using short-term magnetization. EBs were stimulated for one hour at different magnetic field strengths, subsequently exerting a range of force intensity on the cells at different stages of early EB development. Our results demonstrated that following exposure to a 0.2 Tesla magnetic field, ESCs respond to magnetically mediated strain by activating Protein Kinase A (PKA) and increasing phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) expression. The timing of stimulation can also be tailored to guide ESC differentiation: the combination of bone morphogenetic protein 4 (BMP4) supplementation with one hour of magnetic attraction on Day 3 enhances cardiomyogenesis by increasing contractile activity and the percentage of sarcomeric α-actin-expressing cells compared to control samples with BMP4 alone. Interestingly, we also observed that the beads alone had some impact on differentiation by increasingly slightly, albeit not significantly, the percentage of cardiomyocytes. Together these results suggest that magnetically mediated strain can be used to enhance the percentage of mouse ESC-derived cardiomyocytes over current differentiation protocols.
Collapse
Affiliation(s)
- Laura R. Geuss
- The University of Texas at Austin, Institute of Cell and Molecular Biology, Austin, Texas, United States of America
| | - Douglas C. Wu
- The University of Texas at Austin, Institute of Cell and Molecular Biology, Austin, Texas, United States of America
| | - Divya Ramamoorthy
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States of America
| | - Corinne D. Alford
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States of America
| | - Laura J. Suggs
- The University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Khorsandi L, Nejad-Dehbashi F, Ahangarpour A, Hashemitabar M. Three-dimensional differentiation of bone marrow-derived mesenchymal stem cells into insulin-producing cells. Tissue Cell 2014; 47:66-72. [PMID: 25554603 DOI: 10.1016/j.tice.2014.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/22/2014] [Accepted: 11/23/2014] [Indexed: 12/20/2022]
Abstract
Fibrin glue (FG) is used in a variety of clinical applications and in the laboratory for localized and sustained release of factors potentially important for tissue engineering. The aim of this study was to evaluate FG scaffold effect on differentiation of insulin-producing cells (IPCs) from bone marrow-derived mesenchymal stem cells (BM-MSCs). In this experimental study BM-MSCs were cultured and the cells characterized by analysis of cell surface markers using flow cytometry. BM-MSCs were seeded in FG scaffold (3D culture) and then treated with induction media. After induction, the presence of IPCs was demonstrated using gene expression profiles for pancreatic cell differentiation markers (PDX-1, GLUT-2 and insulin) and insulin detection in cytoplasm. Release of insulin by these cells was confirmed by radioimmunoassay. Expression of the islet-associated genes PDX-1, GLUT-2 and Insulin genes in 3D cultured cells was markedly higher than the 2D cultured cells exposure differentiation media. Compared to 2D culture of BM-MSCs-derived IPCs, the insulin release from 3D BM-MSCs-derived IPCs showed a nearly 3 fold (p<0.05) increase when exposed to a high glucose (25 mM) medium. Percentage of insulin positive cells in 3D experimental group showed an approximately 3.5-fold increase in compared to 2D experimental culture cells. The results of this study demonstrated that FG scaffold can enhance the differentiation of IPCs from rats BM-MSCs.
Collapse
Affiliation(s)
- Layasadat Khorsandi
- Cell & Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Fereshteh Nejad-Dehbashi
- Cell & Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Diabetes Research Center, Health research institute and Department of Physiology, School of Medicine, Jundishapur University of Medical Sciences, Ahvaz 61335-189, Iran
| | - Mahmoud Hashemitabar
- Cell & Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
39
|
Deng WP, Yang CC, Yang LY, Chen CWD, Chen WH, Yang CB, Chen YH, Lai WFT, Renshaw PF. Extracellular matrix-regulated neural differentiation of human multipotent marrow progenitor cells enhances functional recovery after spinal cord injury. Spine J 2014; 14:2488-99. [PMID: 24792783 PMCID: PMC4692164 DOI: 10.1016/j.spinee.2014.04.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 04/01/2014] [Accepted: 04/15/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Recent advanced studies have demonstrated that cytokines and extracellular matrix (ECM) could trigger various types of neural differentiation. However, the efficacy of differentiation and in vivo transplantation has not yet thoroughly been investigated. PURPOSE To highlight the current understanding of the effects of ECM on neural differentiation of human bone marrow-derived multipotent progenitor cells (MPCs), regarding state-of-art cure for the animal with acute spinal cord injury (SCI), and explore future treatments aimed at neural repair. STUDY DESIGN A selective overview of the literature pertaining to the neural differentiation of the MSCs and experimental animals aimed at improved repair of SCI. METHODS Extracellular matrix proteins, tenascin-cytotactin (TN-C), tenascin-restrictin (TN-R), and chondroitin sulfate (CS), with the cytokines, nerve growth factor (NGF)/brain-derived neurotrophic factor (BDNF)/retinoic acid (RA) (NBR), were incorporated to induce transdifferentiation of human MPCs. Cells were treated with NBR for 7 days, and then TN-C, TN-R, or CS was added for 2 days. The medium was changed every 2 days. Twenty-four animals were randomly assigned to four groups with six animals in each group: one experimental and three controls. Animals received two (bilateral) injections of vehicle, MPCs, NBR-induced MPCs, or NBR/TN-C-induced MPCs into the lesion sites after SCI. Functional assessment was measured using the Basso, Beattie, and Bresnahan locomotor rating score. Data were analyzed using analysis of variance followed by Student-Newman-Keuls (SNK) post hoc tests. RESULTS Results showed that MPCs with the transdifferentiation of human MPCs to neurons were associated with increased messenger-RNA (mRNA) expression of neuronal markers including nestin, microtubule-associated protein (MAP) 2, glial fibrillary acidic protein, βIII tubulin, and NGF. Greater amounts of neuronal morphology appeared in cultures incorporated with TN-C and TN-R than those with CS. The addition of TN-C enhanced mRNA expressions of MAP2, βIII tubulin, and NGF, whereas TN-R did not significantly change. Conversely, CS exposure decreased MAP2, βIII tubulin, and NGF expressions. The TN-C-treated MSCs significantly and functionally repaired SCI-induced rats at Day 42. Present results indicate that ECM components, such as tenascins and CS in addition to cytokines, may play functional roles in regulating neurogenesis by human MPCs. CONCLUSIONS These findings suggest that the combined use of TN-C, NBR, and human MPCs offers a new feasible method for nerve repair.
Collapse
Affiliation(s)
- Win-Ping Deng
- Graduate Institute of Biomedical Materials and Engineering, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Chi-Chiang Yang
- Department of Neurology, Tungs’ Taichung Metroharbor Hospital, 699 Taiwan Blvd. 8 Sec., Taitung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Chun-Wei D. Chen
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 415 E. 68th Street, New York 10065, NY, USA
| | - Wei-Hong Chen
- Graduate Institute of Biomedical Materials and Engineering, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Charn-Bing Yang
- Orthopedic Section Department, New Taipei City Hospital, 198 Yin-His Rd., Banquiao District, New Taipei City, Taiwan
| | - Yu-Hsin Chen
- Department of Physiology, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Wen-Fu T. Lai
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 415 E. 68th Street, New York 10065, NY, USA,International Center of Nano Biomedicine Research, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan,Brain McLean Imaging Center, McLean Hospital/Harvard Medical School, 115 Mill Strret, Belmont 02115, MA, USA,Corresponding author. Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan. Tel.: (886)2-23916632; fax: (886)2-23967262. (W.-F.T. Lai)
| | - Perry F. Renshaw
- The Brain Institute, The University of Utah, 201 Presidents Cir, Salt Lake City 84112, UT, USA
| |
Collapse
|
40
|
Shen X, Yang Q, Jin P, Li X. Alpha-lipoic acid enhances DMSO-induced cardiomyogenic differentiation of P19 cells. Acta Biochim Biophys Sin (Shanghai) 2014; 46:766-73. [PMID: 25112287 DOI: 10.1093/abbs/gmu057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alpha-lipoic acid (α-LA) is a potent antioxidant that acts as an essential cofactor in mitochondrial dehydrogenase reactions. α-LA has been shown to possess anti-inflammatory and cytoprotective properties, and is used to improve symptoms of diabetic neuropathy. However, the role of α-LA in stem cell differentiation and the underlying molecular mechanisms remain unknown. In the present study, we showed that α-LA significantly promoted dimethyl sulfoxide (DMSO)-induced cardiomyogenic differentiation of mouse embryonic carcinoma P19 cells. α-LA dose dependently increased beating embryonic body (EB) percentages of DMSO-differentiated P19 cells. The expressions of cardiac specific genes TNNT2, Nkx2.5, GATA4, MEF2C, and MLC2V and cardiac isoform of troponin T (cTnT)-positively stained cell population were significantly up-regulated by the addition of α-LA. We also demonstrated that the differentiation time after EB formation was critical for α-LA to take effect. Interestingly, without DMSO treatment, α-LA did not stimulate the cardiomyogenic differentiation of P19 cells. Further investigation indicated that collagen synthesis-enhancing activity, instead of the antioxidative property, plays a significant role in the cardiomyogenic differentiation-promoting function of α-LA. These findings highlight the potential use of α-LA for regenerative therapies in heart diseases.
Collapse
Affiliation(s)
- Xinghua Shen
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Qinghui Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Peng Jin
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Xueqi Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| |
Collapse
|
41
|
Mashinchian O, Bonakdar S, Taghinejad H, Satarifard V, Heidari M, Majidi M, Sharifi S, Peirovi A, Saffar S, Taghinejad M, Abdolahad M, Mohajerzadeh S, Shokrgozar MA, Rezayat SM, Ejtehadi MR, Dalby MJ, Mahmoudi M. Cell-imprinted substrates act as an artificial niche for skin regeneration. ACS APPLIED MATERIALS & INTERFACES 2014; 6:13280-13292. [PMID: 24967724 DOI: 10.1021/am503045b] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Bioinspired materials can mimic the stem cell environment and modulate stem cell differentiation and proliferation. In this study, biomimetic micro/nanoenvironments were fabricated by cell-imprinted substrates based on mature human keratinocyte morphological templates. The data obtained from atomic force microscopy and field emission scanning electron microscopy revealed that the keratinocyte-cell-imprinted poly(dimethylsiloxane) casting procedure could imitate the surface morphology of the plasma membrane, ranging from the nanoscale to the macroscale, which may provide the required topographical cell fingerprints to induce differentiation. Gene expression levels of the genes analyzed (involucrin, collagen type I, and keratin 10) together with protein expression data showed that human adipose-derived stem cells (ADSCs) seeded on these cell-imprinted substrates were driven to adopt the specific shape and characteristics of keratinocytes. The observed morphology of the ADSCs grown on the keratinocyte casts was noticeably different from that of stem cells cultivated on the stem-cell-imprinted substrates. Since the shape and geometry of the nucleus could potentially alter the gene expression, we used molecular dynamics to probe the effect of the confining geometry on the chain arrangement of simulated chromatin fibers in the nuclei. The results obtained suggested that induction of mature cell shapes onto stem cells can influence nucleus deformation of the stem cells followed by regulation of target genes. This might pave the way for a reliable, efficient, and cheap approach of controlling stem cell differentiation toward skin cells for wound healing applications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATiM), Tehran University of Medical Sciences , P.O. Box 14177-55469, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Archacka K, Denkis A, Brzóska E, Świerczek B, Tarczyluk M, Jańczyk-Ilach K, Ciemerych MA, Moraczewski J. Competence of in vitro cultured mouse embryonic stem cells for myogenic differentiation and fusion with myoblasts. Stem Cells Dev 2014; 23:2455-68. [PMID: 24940624 DOI: 10.1089/scd.2013.0582] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pluripotent stem cells are a potential source of various cell types for use in regenerative medicine. Despite accumulating knowledge, there is currently no efficient and reproducible protocol that does not require genetic manipulation for generation of myogenic cells from pluripotent stem cells. Here, we examined whether mouse embryonic stem (ES) cells are able to undergo myogenic differentiation and fusion in response to signals released by differentiating myoblasts. Using ES cells expressing the histone 2B-green fluorescent fusion protein, we were able to detect hybrid myotubes formed by ES cells and differentiating myoblasts. ES cells that fused with myoblasts downregulated the expression of pluripotency markers and induced the expression of myogenic markers, while unfused ES cells did not exhibit this expression pattern. Thus, the signals released by myoblasts were not sufficient to induce myogenic differentiation of ES cells. Although ES cells synthesize many proteins involved in myoblast adhesion and fusion, we did not observe any myotubes formed exclusively by ES cells. We found that ES cells lacked M-cadherin and vascular cell adhesion molecule-1, which may account for the low frequency of hybrid myotube formation in ES cell-myoblast co-cultures and the inability of ES cells alone to form myotubes.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw , Warsaw, Poland
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Three-Dimensional (3D) Cell Culture Conditions, Present and Future Improvements. RAZAVI INTERNATIONAL JOURNAL OF MEDICINE 2014. [DOI: 10.5812/rijm.17803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
44
|
Disrupted WNT Signaling in Mouse Embryonic Stem Cells in the Absence of Calreticulin. Stem Cell Rev Rep 2014; 10:191-206. [DOI: 10.1007/s12015-013-9488-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
Moshaverinia A, Xu X, Chen C, Ansari S, Zadeh HH, Snead ML, Shi S. Application of stem cells derived from the periodontal ligament or gingival tissue sources for tendon tissue regeneration. Biomaterials 2014; 35:2642-50. [PMID: 24397989 DOI: 10.1016/j.biomaterials.2013.12.053] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 12/19/2013] [Indexed: 12/24/2022]
Abstract
Tendon injuries are often associated with significant dysfunction and disability due to tendinous tissue's very limited self-repair capacity and propensity for scar formation. Dental-derived mesenchymal stem cells (MSCs) in combination with appropriate scaffold material present an alternative therapeutic option for tendon repair/regeneration that may be advantageous compared to other current treatment modalities. The MSC delivery vehicle is the principal determinant for successful implementation of MSC-mediated regenerative therapies. In the current study, a co-delivery system based on TGF-β3-loaded RGD-coupled alginate microspheres was developed for encapsulating periodontal ligament stem cells (PDLSCs) or gingival mesenchymal stem cells (GMSCs). The capacity of encapsulated dental MSCs to differentiate into tendon tissue was investigated in vitro and in vivo. Encapsulated dental-derived MSCs were transplanted subcutaneously into immunocompromised mice. Our results revealed that after 4 weeks of differentiation in vitro, PDLSCs and GMSCs as well as the positive control human bone marrow mesenchymal stem cells (hBMMSCs) exhibited high levels of mRNA expression for gene markers related to tendon regeneration (Scx, DCn, Tnmd, and Bgy) via qPCR measurement. In a corresponding in vivo animal model, ectopic neo-tendon regeneration was observed in subcutaneous transplanted MSC-alginate constructs, as confirmed by histological and immunohistochemical staining for protein markers specific for tendons. Interestingly, in our quantitative PCR and in vivo histomorphometric analyses, PDLSCs showed significantly greater capacity for tendon regeneration than GMSCs or hBMMSCs (P < 0.05). Altogether, these findings indicate that periodontal ligament and gingival tissues can be considered as suitable stem cell sources for tendon engineering. PDLSCs and GMSCs encapsulated in TGF-β3-loaded RGD-modified alginate microspheres are promising candidates for tendon regeneration.
Collapse
Affiliation(s)
- Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA.
| | - Xingtian Xu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Chider Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Sahar Ansari
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Homayoun H Zadeh
- Laboratory for Immunoregulation and Tissue Engineering (LITE), Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Malcolm L Snead
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Songtao Shi
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
46
|
Kisspeptin-10 modulates the proliferation and differentiation of the rhesus monkey derived stem cell line: R366.4. ScientificWorldJournal 2013; 2013:135470. [PMID: 24381507 PMCID: PMC3863535 DOI: 10.1155/2013/135470] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 10/09/2013] [Indexed: 01/07/2023] Open
Abstract
The rhesus monkey embryonic stem cell line R366.4 has been identified to differentiate into a number of cell types. However, it has not been well characterized for its response to drugs affecting reproductive endocrinology. Kisspeptins (KPs) are ligands for the GPR-54, which is known to modulate reproductive function. The current study was designed to determine the effect of the KP-10 peptide on R366.4 cells and to investigate the role of KP-GPR54 in the cell proliferation process. Four different doses (0.1, 1, 10, and 100 nM) of KP-10 and control were selected to evaluate cell growth parameters and cellular morphological changes over a 72 hr period. The cells were treated with kisspeptin-10 during the early rosette stage. Proliferation rates, analyzed by flow cytometry and cell count methods, were found to be decreased after treatment. Moreover, the number of rosettes was found to decrease following KP-10 treatments. Morphological changes consisting of neuronal projections were also witnessed. This suggested that KP-10 had an antiproliferative effect on R366.4 cells leading to a differentiation state and morphological changes consistent with neuronal stem cell development. The R366.4 stem cell line differentiates based on kisspeptin signaling and may be used to investigate reproductive cell endocrinology in vitro.
Collapse
|
47
|
Prabhakaran MP, Mobarakeh LG, Kai D, Karbalaie K, Nasr-Esfahani MH, Ramakrishna S. Differentiation of embryonic stem cells to cardiomyocytes on electrospun nanofibrous substrates. J Biomed Mater Res B Appl Biomater 2013; 102:447-54. [PMID: 24039141 DOI: 10.1002/jbm.b.33022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 06/24/2013] [Accepted: 08/01/2013] [Indexed: 12/31/2022]
Abstract
The potential of pluripotent embryonic stem cells (ESCs) isolated from the inner mass of blastocysts are investigated for its ability to differentiate on biocompatible electrospun nanofibers, for regeneration of the myocardially infracted heart. Nanostructured poly(d,l-lactide-co-glycolide)/collagen (PLGA/Col) scaffolds with fiber diameters in the range of 300 ± 65 nm, was fabricated by electrospinning to mimic the extracellular matrix of the native tissue. During the culture of embryoid bodies outgrowth on the scaffolds, and further differentiation of ESCs to cardiomyocytes, the PLGA/Col nanofibers was found better than that of the electrospun PLGA nanofibers, where a better interaction and growth of ESC differentiated cardiomyocytes was observed on the composite scaffolds. The phenotypical characteristics of ESC-derived cardiomyocytes and molecular protein expression were carried out by scanning electron microscopy and immunocytochemistry, respectively. Our studies highlight the significance of a suitable material, its architecture, and cell-biomaterial interactions that is essential at a nanoscale level signifying the application of a bioengineered cardiac graft for stem cell differentiation and transplantation, which could be an intriguing strategy for cardiac regeneration.
Collapse
Affiliation(s)
- Molamma P Prabhakaran
- Center for Nanofibers and Nanotechnology, E3-05-14, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, Singapore, 117576
| | | | | | | | | | | |
Collapse
|
48
|
A microparticle approach to morphogen delivery within pluripotent stem cell aggregates. Biomaterials 2013; 34:7227-35. [PMID: 23827184 DOI: 10.1016/j.biomaterials.2013.05.079] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 05/30/2013] [Indexed: 01/23/2023]
Abstract
Stem cell fate and specification is largely controlled by extrinsic cues that comprise the 3D microenvironment. Biomaterials can serve to control the spatial and temporal presentation of morphogenic molecules in order to direct stem cell fate decisions. Here we describe a microparticle (MP)-based approach to deliver growth factors within multicellular aggregates to direct pluripotent stem cell differentiation. Compared to conventional soluble delivery methods, gelatin MPs laden with BMP4 or noggin induced efficient gene expression of mesoderm and ectoderm lineages, respectively, despite using nearly 12-fold less total growth factor. BMP4-laden MPs increased the percentage of cells expressing GFP under the control of the Brachyury-T promoter as visualized by whole-mount confocal imaging and quantified by flow cytometry. Furthermore, the ability to localize MPs laden with different morphogens within a particular hemisphere of stem cell aggregates allowed for spatial control of differentiation within 3D cultures. Overall, localized delivery of growth factors within multicellular aggregates from microparticle delivery vehicles is an important step towards scalable differentiation technologies and the study of morphogen gradients in pluripotent stem cell differentiation.
Collapse
|
49
|
Moshaverinia A, Ansari S, Chen C, Xu X, Akiyama K, Snead ML, Zadeh HH, Shi S. Co-encapsulation of anti-BMP2 monoclonal antibody and mesenchymal stem cells in alginate microspheres for bone tissue engineering. Biomaterials 2013; 34:6572-9. [PMID: 23773817 DOI: 10.1016/j.biomaterials.2013.05.048] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/23/2013] [Indexed: 12/19/2022]
Abstract
Recently, it has been shown that tethered anti-BMP2 monoclonal antibodies (mAbs) can trap BMP ligands and thus provide BMP inductive signals for osteo-differentiation of progenitor cells. The objectives of this study were to: (1) develop a co-delivery system based on murine anti-BMP2 mAb-loaded alginate microspheres encapsulating human bone marrow mesenchymal stem cells (hBMMSCs); and (2) investigate osteogenic differentiation of encapsulated stem cells in alginate microspheres in vitro and in vivo. Alginate microspheres of 1 ± 0.1 mm diameter were fabricated with 2 × 10(6) hBMMSCs per mL of alginate. Critical-size calvarial defects (5 mm diameter) were created in immune-compromised mice and alginate microspheres preloaded with anti-BMP mAb encapsulating hBMMSCs were transplanted into defect sites. Alginate microspheres pre-loaded with isotype-matched non-specific antibody were used as the negative control. After 8 weeks, micro CT and histologic analyses were used to analyze bone formation. In vitro analysis demonstrated that anti-BMP2 mAbs tethered BMP2 ligands that can activate the BMP receptors on hBMMSCs. The co-delivery system described herein, significantly enhanced hBMMSC-mediated osteogenesis, as confirmed by the presence of BMP signal pathway-activated osteoblast determinants Runx2 and ALP. Our results highlight the importance of engineering the microenvironment for stem cells, and particularly the value of presenting inductive signals for osteo-differentiation of hBMMSCs by tethering BMP ligands using mAbs. This strategy of engineering the microenvironment with captured BMP signals is a promising modality for repair and regeneration of craniofacial, axial and appendicular bone defects.
Collapse
Affiliation(s)
- Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kothapalli CR, Kamm RD. 3D matrix microenvironment for targeted differentiation of embryonic stem cells into neural and glial lineages. Biomaterials 2013; 34:5995-6007. [PMID: 23694902 DOI: 10.1016/j.biomaterials.2013.04.042] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 04/23/2013] [Indexed: 12/13/2022]
Abstract
The onset of neurodegenerative disorders is characterized by the progressive dysfunction and loss of subpopulations of specialized cells within specific regions of the central nervous system (CNS). Since CNS has a limited ability for self-repair and regeneration under such conditions, clinical transplantation of stem cells has been explored as an alternative. Although embryonic stem cells (ESCs) offer a promising therapeutic platform to treat a variety of neurodegenerative disorders, the niche microenvironment, which could regulate their differentiation into specialized lineages on demand, needs to be optimized for successful clinical transplantation. Here, we evaluated the synergistic role of matrix microenvironment (type, architecture, composition, stiffness) and signaling molecules (type, dosage) on murine ESC differentiation into specific neural and glial lineages. ESCs were cultured as embryoid bodies on either 2D substrates or within 3D scaffolds, in the presence or absence of retinoic acid (RA) and sonic hedgehog (Shh). Results showed that ESCs maintained their stemness even after 4 days in the absence of exogenous signaling molecules, as evidenced by Oct-4 staining. RA at 1 μM dosage was deemed optimal for neural differentiation and neurite outgrowth on collagen-1 coated substrates. Significant neural differentiation with robust neurite outgrowth and branching was evident only on collagen-1 coated 2D substrates and within 3D matrigel scaffolds, in the presence of 1 μM RA. Blocking α6 or β1 integrin subunits on differentiating cells inhibited matrigel-induced effects on neural differentiation and neurite outgrowth. Hydrogel concentration strongly regulated formation of neural and astrocyte lineages in 1 μM RA additive cultures. When RA and Shh were provided, either alone or together, 3D collagen-1 scaffolds enhanced significant motor neuron formation, while 3D matrigel stimulated dopaminergic neuron differentiation. These results suggest a synergistic role of microenvironmental cues for ESC differentiation and maturation, with potential applications in cell transplantation therapy.
Collapse
Affiliation(s)
- Chandrasekhar R Kothapalli
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH 44115, USA.
| | | |
Collapse
|