1051
|
Zhang X, Jaramillo M, Singh S, Kumta P, Banerjee I. Analysis of regulatory network involved in mechanical induction of embryonic stem cell differentiation. PLoS One 2012; 7:e35700. [PMID: 22558203 PMCID: PMC3338716 DOI: 10.1371/journal.pone.0035700] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 03/20/2012] [Indexed: 01/14/2023] Open
Abstract
Embryonic stem cells are conventionally differentiated by modulating specific growth factors in the cell culture media. Recently the effect of cellular mechanical microenvironment in inducing phenotype specific differentiation has attracted considerable attention. We have shown the possibility of inducing endoderm differentiation by culturing the stem cells on fibrin substrates of specific stiffness. Here, we analyze the regulatory network involved in such mechanically induced endoderm differentiation under two different experimental configurations of 2-dimensional and 3-dimensional culture, respectively. Mouse embryonic stem cells are differentiated on an array of substrates of varying mechanical properties and analyzed for relevant endoderm markers. The experimental data set is further analyzed for identification of co-regulated transcription factors across different substrate conditions using the technique of bi-clustering. Overlapped bi-clusters are identified following an optimization formulation, which is solved using an evolutionary algorithm. While typically such analysis is performed at the mean value of expression data across experimental repeats, the variability of stem cell systems reduces the confidence on such analysis of mean data. Bootstrapping technique is thus integrated with the bi-clustering algorithm to determine sets of robust bi-clusters, which is found to differ significantly from corresponding bi-clusters at the mean data value. Analysis of robust bi-clusters reveals an overall similar network interaction as has been reported for chemically induced endoderm or endodermal organs but with differences in patterning between 2-dimensional and 3-dimensional culture. Such analysis sheds light on the pathway of stem cell differentiation indicating the prospect of the two culture configurations for further maturation.
Collapse
Affiliation(s)
- Xinan Zhang
- School of Mathematics and Statistics, Central China Normal University, Wuhan, China
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Maria Jaramillo
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Satish Singh
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Prashant Kumta
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Complex Engineered Multifunctional Materials, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ipsita Banerjee
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
1052
|
Kilian KA, Mrksich M. Directing Stem Cell Fate by Controlling the Affinity and Density of Ligand-Receptor Interactions at the Biomaterials Interface. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201108746] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
1053
|
Kilian KA, Mrksich M. Directing stem cell fate by controlling the affinity and density of ligand-receptor interactions at the biomaterials interface. Angew Chem Int Ed Engl 2012; 51:4891-5. [PMID: 22505230 DOI: 10.1002/anie.201108746] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Indexed: 01/24/2023]
Abstract
Sticky situation: the differentiation of mesenchymal stem cells can be influenced by the affinity and density of an immobilized ligand for the integrin receptors. Cells adherent to monolayers that present the high-affinity, cyclic-RGD peptide (left) show increased expression of osteogenic markers, while cells on monolayers presenting the lower-affinity, linear-RGD peptide (right) express early markers of myogenesis at a high density and neurogenesis at a low density of the ligand.
Collapse
|
1054
|
D'Angelo F, Armentano I, Cacciotti I, Tiribuzi R, Quattrocelli M, Del Gaudio C, Fortunati E, Saino E, Caraffa A, Cerulli GG, Visai L, Kenny JM, Sampaolesi M, Bianco A, Martino S, Orlacchio A. Tuning multi/pluri-potent stem cell fate by electrospun poly(L-lactic acid)-calcium-deficient hydroxyapatite nanocomposite mats. Biomacromolecules 2012; 13:1350-60. [PMID: 22449037 DOI: 10.1021/bm3000716] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this study, we investigated whether multipotent (human-bone-marrow-derived mesenchymal stem cells [hBM-MSCs]) and pluripotent stem cells (murine-induced pluripotent stem cells [iPSCs] and murine embryonic stem cells [ESCs]) respond to nanocomposite fibrous mats of poly(L-lactic acid) (PLLA) loaded with 1 or 8 wt % of calcium-deficient nanohydroxyapatite (d-HAp). Remarkably, the dispersion of different amounts of d-HAp to PLLA produced a set of materials (PLLA/d-HAp) with similar architectures and tunable mechanical properties. After 3 weeks of culture in the absence of soluble osteogenic factors, we observed the expression of osteogenic markers, including the deposition of bone matrix proteins, in multi/pluripotent cells only grown on PLLA/d-HAp nanocomposites, whereas the osteogenic differentiation was absent on stem-cell-neat PLLA cultures. Interestingly, this phenomenon was confined only in hBM-MSCs, murine iPSCs, and ESCs grown on direct contact with the PLLA/d-HAp mats. Altogether, these results indicate that the osteogenic differentiation effect of these electrospun PLLA/d-HAp nanocomposites was independent of the stem cell type and highlight the direct interaction of stem cell-polymeric nanocomposite and the mechanical properties acquired by the PLLA/d-HAp nanocomposites as key steps for the differentiation process.
Collapse
Affiliation(s)
- Francesco D'Angelo
- Department of Experimental Medicine and Biochemical Sciences, Section of Biochemistry and Molecular Biology, University of Perugia, Via del Giochetto, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1055
|
Kim WS, Mooney DJ, Arany PR, Lee K, Huebsch N, Kim J. Adipose Tissue Engineering Using Injectable, Oxidized Alginate Hydrogels. Tissue Eng Part A 2012; 18:737-43. [DOI: 10.1089/ten.tea.2011.0250] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Woo Seob Kim
- Department of Plastic Surgery, College of Medicine, Chung-Ang University, Heuk Seok-Dong, Dong Jak-Gu, Seoul, Korea
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
| | - David J. Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
| | - Praveen R. Arany
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
- Program in Oral and Maxillofacial Pathology, Leder Human Biology and Translational Medicine and Biological Sciences in Dental Medicine, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Kangwon Lee
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
| | - Nathaniel Huebsch
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Cambridge, Massachusetts
| | - Jaeyun Kim
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
1056
|
Mohamed A, Xing MM. Nanomaterials and nanotechnology for skin tissue engineering. INTERNATIONAL JOURNAL OF BURNS AND TRAUMA 2012; 2:29-41. [PMID: 22928165 PMCID: PMC3415966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 02/20/2012] [Indexed: 06/01/2023]
Abstract
A recent literature review of the field shows that tissue-engineered skin has been in clinical use for the last several decades and that, over this time the technology has advanced rapidly. Despite this progress no synthetic skin yet produced has completely replicated normal, healthy skin. Therefore, researchers must continue to develop materials that successfully overcome the problems with current skin tissue substitutes. This paper is a comprehensive review of the prospects for nanotechnology and nanomaterials to close this gap by mimicking surface properties for reconstruction of a variety of skin tissues. In addition, a number of commercially available products that regenerate different layers of the burn-damaged or chronically wounded skin are reviewed.
Collapse
Affiliation(s)
- Aezeden Mohamed
- Nano-Medicine and Tissue Engineering Lab, Department of Mechanical and Manufacturing Engineering, Faculty of Engineering, Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba and Manitoba Institute of Child Health Winnipeg, Manitoba, Canada
| | | |
Collapse
|
1057
|
González-García C, Moratal D, Oreffo ROC, Dalby MJ, Salmerón-Sánchez M. Surface mobility regulates skeletal stem cell differentiation. Integr Biol (Camb) 2012; 4:531-9. [PMID: 22395101 DOI: 10.1039/c2ib00139j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A family of polymer substrates which consists of a vinyl backbone chain with the side groups -COO(CH(2))(x)H, with x = 1, 2, 4, was prepared. Substrates with similar chemical groups but decreasing stiffness, characterized by their elastic modulus at 37 °C, as well as surface mobility, characterized by the glass transition temperature, were obtained. We have investigated whether these subtle variations in polymer chemistry lead to alterations in fibronectin (FN) adsorption and mesenchymal stem cell response. The same FN density was adsorbed on every substrate (∼450 ng cm(-2)) although the supramolecular organization of the protein at the material interface, as obtained with AFM, was different for x = 1 and the other two surfaces (x = 2, 4). Consequently, this allows one to investigate the effect of physical properties of the matrix on stem cell differentiation after ruling out any influence of protein activity. Cell adhesion was quantified by calculating the size distribution of focal adhesions. Mesenchymal stem cell differentiation to the osteoblastic lineage was determined by quantifying protein levels for osteocalcin, osteopontin and Runx2, in the absence of any additional osteogenic soluble factors in the culture media, but as a direct effect of material properties. The findings indicate the potential to modulate skeletal progenitor cell commitment to the osteoblastic lineage through surface mobility of the underlying material surface.
Collapse
Affiliation(s)
- Cristina González-García
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, 46022 Valencia, Spain
| | | | | | | | | |
Collapse
|
1058
|
Zhao L, Liu L, Wu Z, Zhang Y, Chu PK. Effects of micropitted/nanotubular titania topographies on bone mesenchymal stem cell osteogenic differentiation. Biomaterials 2012; 33:2629-41. [PMID: 22204980 DOI: 10.1016/j.biomaterials.2011.12.024] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 12/10/2011] [Indexed: 01/14/2023]
Abstract
Micro/nanotopographical modification of biomaterials constitutes a promising approach to direct stem cell osteogenic differentiation to promote osseointegration. In this work, titania nanotubes (NTs) 25 and 80 nm in size with the acid-etched Ti topography (AcidTi) and hierarchical hybrid micropitted/nanotubular topographies (Micro/5VNT and Micro/20VNT) are produced to mimic the structure of the natural bone extracellular matrix (ECM). The effects on bone mesenchymal stem cell (MSC) osteogenic differentiation are studied systematically by various microscopic and biological characterization techniques. Cell adhesion is assayed by nucleus fluorescence staining and cell proliferation is studied by CCK-8 assay and flow cytometry. Osteogenic differentiation is assayed by alkaline phosphatase (ALP) expression, collagen secretion, matrix mineralization, and quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis on the osteogenesis related gene expression. All the topographies are observed to induce MSC osteogenic differentiation in the absence of osteogenic supplements. The nanotube surfaces significantly promote cell attachment and spread, collagen secretion and ECM mineralization, as well as osteogenesis-related gene expression. Among them, Micro/20VNT shows the best ability to simultaneously promote MSC proliferation and osteogenic differentiation. Our results unambiguously demonstrate their excellent ability to support MSC proliferation and induce MSC osteogenic differentiation, especially those with the micropitted topography.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Periodontology and Oral Medicine, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an 710032, China
| | | | | | | | | |
Collapse
|
1059
|
Sun Y, Chen CS, Fu J. Forcing stem cells to behave: a biophysical perspective of the cellular microenvironment. Annu Rev Biophys 2012; 41:519-42. [PMID: 22404680 DOI: 10.1146/annurev-biophys-042910-155306] [Citation(s) in RCA: 297] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Physical factors in the local cellular microenvironment, including cell shape and geometry, matrix mechanics, external mechanical forces, and nanotopographical features of the extracellular matrix, can all have strong influences on regulating stem cell fate. Stem cells sense and respond to these insoluble biophysical signals through integrin-mediated adhesions and the force balance between intracellular cytoskeletal contractility and the resistant forces originated from the extracellular matrix. Importantly, these mechanotransduction processes can couple with many other potent growth-factor-mediated signaling pathways to regulate stem cell fate. Different bioengineering tools and microscale/nanoscale devices have been successfully developed to engineer the physical aspects of the cellular microenvironment for stem cells, and these tools and devices have proven extremely powerful for identifying the extrinsic physical factors and their downstream intracellular signaling pathways that control stem cell functions.
Collapse
Affiliation(s)
- Yubing Sun
- Integrated Biosystems and Biomechanics Laboratory, Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
1060
|
Weiss MS, Bernabé BP, Shikanov A, Bluver DA, Mui MD, Shin S, Broadbelt LJ, Shea LD. The impact of adhesion peptides within hydrogels on the phenotype and signaling of normal and cancerous mammary epithelial cells. Biomaterials 2012; 33:3548-59. [PMID: 22341213 DOI: 10.1016/j.biomaterials.2012.01.055] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 01/29/2012] [Indexed: 11/26/2022]
Abstract
The microenviroment contributes to directing mammary epithelial cell (MEC) development and the progression of breast cancer. Three-dimensional culture models have been used to support formation of structures that display varying degrees of disorganization that parallel the degree of cancer. Synthetic hydrogels were employed to investigate the mechanisms by which specific adhesion signals in the microenvironment directed development. Polyethylene glycol-based hydrogels supported 3D growth of MECs and directed formation of a range of phenotypes that were functions of genotype, and identity and concentration of adhesion peptides RGD and YIGSR. Non-cancerous and cancerous MECs responded differentially to the same adhesion cues and produced variable structural organizations. An analysis of dynamic signaling pathways revealed differential activities of transcription factors within the MAPK and JAK/STAT pathways in response to genotype and adhesion. These results directly implicate adhesion in cancer development and demonstrate that AP1, CREB, STAT1, and STAT3 all contribute to the genotype dependence of cellular response to adhesion peptides. The tools presented in this work could be applied to other systems and connect extracellular cues with intracellular signaling to molecularly dissect tissue development and further biomaterials development.
Collapse
Affiliation(s)
- Michael S Weiss
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208-3120, USA
| | | | | | | | | | | | | | | |
Collapse
|
1061
|
Shi J, Xing MMQ, Zhong W. Development of hydrogels and biomimetic regulators as tissue engineering scaffolds. MEMBRANES 2012; 2:70-90. [PMID: 24957963 PMCID: PMC4021879 DOI: 10.3390/membranes2010070] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 01/17/2012] [Accepted: 02/02/2012] [Indexed: 01/06/2023]
Abstract
This paper reviews major research and development issues relating to hydrogels as scaffolds for tissue engineering, the article starts with a brief introduction of tissue engineering and hydrogels as extracellular matrix mimics, followed by a description of the various types of hydrogels and preparation methods, before a discussion of the physical and chemical properties that are important to their application. There follows a short comment on the trends of future research and development. Throughout the discussion there is an emphasis on the genetic understanding of bone tissue engineering application.
Collapse
Affiliation(s)
- Junbin Shi
- Department of Textile Sciences, Faculty of Human Ecology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Malcolm M Q Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Wen Zhong
- Department of Textile Sciences, Faculty of Human Ecology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| |
Collapse
|
1062
|
Kniazeva E, Weidling JW, Singh R, Botvinick EL, Digman MA, Gratton E, Putnam AJ. Quantification of local matrix deformations and mechanical properties during capillary morphogenesis in 3D. Integr Biol (Camb) 2012; 4:431-9. [PMID: 22281872 DOI: 10.1039/c2ib00120a] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Reciprocal mechanical interactions between cells and the extracellular matrix (ECM) are thought to play important instructive roles in branching morphogenesis. However, most studies to date have failed to characterize these interactions on a length scale relevant to cells, especially in three-dimensional (3D) matrices. Here we utilized two complementary methods, spatio-temporal image correlation spectroscopy (STICS) and laser optical tweezers-based active microrheology (AMR), to quantify endothelial cell (EC)-mediated deformations of individual ECM elements and the local ECM mechanical properties, respectively, during the process of capillary morphogenesis in a 3D cell culture model. In experiments in which the ECM density was systematically varied, STICS revealed that the rate at which ECs deformed individual ECM fibers on the microscale positively correlated with capillary sprouting on the macroscale. ECs expressing constitutively active V14-RhoA displaced individual matrix fibers at significantly faster rates and displayed enhanced capillary sprouting relative to wild-type cells, while those expressing dominant-negative N19-RhoA behaved in an opposite fashion. In parallel, AMR revealed a local stiffening of the ECM proximal to the tips of sprouting ECs. By quantifying the dynamic physical properties of the cell-ECM interface in both space and time, we identified a correlation linking ECM deformation rates and local ECM stiffening at the microscale with capillary morphogenesis at the macroscale.
Collapse
Affiliation(s)
- Ekaterina Kniazeva
- Biomedical Engineering Department, Natural Sciences II, Room 3201, University of California, Irvine, Irvine, CA 92697-2715, USA
| | | | | | | | | | | | | |
Collapse
|
1063
|
Kabiri M, Kul B, Lott WB, Futrega K, Ghanavi P, Upton Z, Doran MR. 3D mesenchymal stem/stromal cell osteogenesis and autocrine signalling. Biochem Biophys Res Commun 2012; 419:142-7. [PMID: 22266317 DOI: 10.1016/j.bbrc.2012.01.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Accepted: 01/05/2012] [Indexed: 01/11/2023]
Abstract
Mesenchymal stem/stromal cells (MSC) are rapidly becoming a leading candidate for use in tissue regeneration, with first generation of therapies being approved for use in orthopaedic repair applications. Capturing the full potential of MSC will likely require the development of novel in vitro culture techniques and devices. Herein we describe the development of a straightforward surface modification of an existing commercial product to enable the efficient study of three dimensional (3D) human bone marrow-derived MSC osteogenic differentiation. Hundreds of 3D microaggregates, of either 42 or 168 cells each, were cultured in osteogenic induction medium and their differentiation was compared with that occurring in traditional two dimensional (2D) monolayer cultures. Osteogenic gene expression and matrix composition was significantly enhanced in the 3D microaggregate cultures. Additionally, BMP-2 gene expression was significantly up-regulated in 3D cultures at day 3 and 7 by approximately 25- and 30-fold, respectively. The difference in BMP-2 gene expression between 2D and 3D cultures was negligible in the more mature day 14 osteogenic cultures. These data support the notion that BMP-2 autocrine signalling is up-regulated in 3D MSC cultures, enhancing osteogenic differentiation. This study provides both mechanistic insight into MSC differentiation, as well as a platform for the efficient generation of microtissue units for further investigation or use in tissue engineering applications.
Collapse
Affiliation(s)
- Mahboubeh Kabiri
- Stem Cell Therapies Laboratory, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld., Australia
| | | | | | | | | | | | | |
Collapse
|
1064
|
Lee-Thedieck C, Rauch N, Fiammengo R, Klein G, Spatz JP. Impact of substrate elasticity on human hematopoietic stem and progenitor cell adhesion and motility. J Cell Sci 2012; 125:3765-75. [DOI: 10.1242/jcs.095596] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In the bone marrow, hematopoietic stem cells (HSCs) reside in endosteal and vascular niches. The interactions with the niches are essential for the maintenance of HSC number and properties. Although the molecular nature of these interactions is quite well understood, little is known about the role of physical parameters such as matrix elasticity. Osteoblasts, the major cellular component of the endosteal HSC niche, flatten during HSC mobilization. We show that this process is accompanied by osteoblast stiffening, demonstrating that not only biochemical signals but also mechanical properties of the niche are modulated. HSCs react to stiffer substrates with increased cell adhesion and migration, which could facilitate the exit of HSCs from the niche. These results indicate that matrix elasticity is an important factor in regulating the retention of HSCs in the endosteal niche and should be considered in attempts to propagate HSCs in vitro for clinical applications.
Collapse
|
1065
|
Shi J, Ouyang J, Li Q, Wang L, Wu J, Zhong W, Xing MMQ. Cell-compatible hydrogels based on a multifunctional crosslinker with tunable stiffness for tissue engineering. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c2jm34862d] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
1066
|
Sitasuwan P, Andrew Lee L, Bo P, Davis EN, Lin Y, Wang Q. A plant virus substrate induces early upregulation of BMP2 for rapid bone formation. Integr Biol (Camb) 2012; 4:651-60. [DOI: 10.1039/c2ib20041d] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
1067
|
Abstract
Alginate is a biomaterial that has found numerous applications in biomedical science and engineering due to its favorable properties, including biocompatibility and ease of gelation. Alginate hydrogels have been particularly attractive in wound healing, drug delivery, and tissue engineering applications to date, as these gels retain structural similarity to the extracellular matrices in tissues and can be manipulated to play several critical roles. This review will provide a comprehensive overview of general properties of alginate and its hydrogels, their biomedical applications, and suggest new perspectives for future studies with these polymers.
Collapse
|
1068
|
Aurand ER, Lampe KJ, Bjugstad KB. Defining and designing polymers and hydrogels for neural tissue engineering. Neurosci Res 2011; 72:199-213. [PMID: 22192467 DOI: 10.1016/j.neures.2011.12.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 11/07/2011] [Accepted: 12/07/2011] [Indexed: 12/16/2022]
Abstract
The use of biomaterials, such as hydrogels, as neural cell delivery devices is becoming more common in areas of research such as stroke, traumatic brain injury, and spinal cord injury. When reviewing the available research there is some ambiguity in the type of materials used and results are often at odds. This review aims to provide the neuroscience community who may not be familiar with fundamental concepts of hydrogel construction, with basic information that would pertain to neural tissue applications, and to describe the use of hydrogels as cell and drug delivery devices. We will illustrate some of the many tunable properties of hydrogels and the importance of these properties in obtaining reliable and consistent results. It is our hope that this review promotes creative ideas for ways that hydrogels could be adapted and employed for the treatment of a broad range of neurological disorders.
Collapse
Affiliation(s)
- Emily R Aurand
- Neuroscience Program, University of Colorado Denver - Anschutz Medical Campus, Aurora, CO 80045, USA.
| | | | | |
Collapse
|
1069
|
Epstein AK, Hochbaum AI, Kim P, Aizenberg J. Control of bacterial biofilm growth on surfaces by nanostructural mechanics and geometry. NANOTECHNOLOGY 2011; 22:494007. [PMID: 22101439 DOI: 10.1088/0957-4484/22/49/494007] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Surface-associated communities of bacteria, called biofilms, pervade natural and anthropogenic environments. Mature biofilms are resistant to a wide range of antimicrobial treatments and therefore pose persistent pathogenic threats. The use of surface chemistry to inhibit biofilm growth has been found to only transiently affect initial attachment. In this work, we investigate the tunable effects of physical surface properties, including high-aspect-ratio (HAR) surface nanostructure arrays recently reported to induce long-range spontaneous spatial patterning of bacteria on the surface. The functional parameters and length scale regimes that control such artificial patterning for the rod-shaped pathogenic species Pseudomonas aeruginosa are elucidated through a combinatorial approach. We further report a crossover regime of biofilm growth on a HAR nanostructured surface versus the nanostructure effective stiffness. When the 'softness' of the hair-like nanoarray is increased beyond a threshold value, biofilm growth is inhibited as compared to a flat control surface. This result is consistent with the mechanoselective adhesion of bacteria to surfaces. Therefore by combining nanoarray-induced bacterial patterning and modulating the effective stiffness of the nanoarray--thus mimicking an extremely compliant flat surface--bacterial mechanoselective adhesion can be exploited to control and inhibit biofilm growth.
Collapse
Affiliation(s)
- A K Epstein
- School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, MA 02138, USA
| | | | | | | |
Collapse
|
1070
|
Engineering approaches toward deconstructing and controlling the stem cell environment. Ann Biomed Eng 2011; 40:1301-15. [PMID: 22101755 DOI: 10.1007/s10439-011-0452-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 10/20/2011] [Indexed: 12/12/2022]
Abstract
Stem cell-based therapeutics have become a vital component in tissue engineering and regenerative medicine. The microenvironment within which stem cells reside, i.e., the niche, plays a crucial role in regulating stem cell self-renewal and differentiation. However, current biological techniques lack the means to recapitulate the complexity of this microenvironment. Nano- and microengineered materials offer innovative methods to (1) deconstruct the stem cell niche to understand the effects of individual elements; (2) construct complex tissue-like structures resembling the niche to better predict and control cellular processes; and (3) transplant stem cells or activate endogenous stem cell populations for regeneration of aged or diseased tissues. In this article, we highlight some of the latest advances in this field and discuss future applications and directions of the use of nano- and microtechnologies for stem cell engineering.
Collapse
|
1071
|
Gulotta LV, Wiznia D, Cunningham M, Fortier L, Maher S, Rodeo SA. What's new in orthopaedic research. J Bone Joint Surg Am 2011; 93:2136-41. [PMID: 22262389 DOI: 10.2106/jbjs.k.00981] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Lawrence V Gulotta
- Laboratory for Soft Tissue Research, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
1072
|
Hwang Y, Phadke A, Varghese S. Engineered microenvironments for self-renewal and musculoskeletal differentiation of stem cells. Regen Med 2011; 6:505-24. [PMID: 21749208 DOI: 10.2217/rme.11.38] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells hold great promise for therapies aimed at regenerating damaged tissue, drug screening and studying in vitro models of human disease. However, many challenges remain before these applications can become a reality. One such challenge is developing chemically defined and scalable culture conditions for derivation and expansion of clinically viable human pluripotent stem cells, as well as controlling their differentiation with high specificity. Interaction of stem cells with their extracellular microenvironment plays an important role in determining their differentiation commitment and functions. Regenerative medicine approaches integrating cell-matrix and cell-cell interactions, and soluble factors could lead to development of robust microenvironments to control various cellular responses. Indeed, several of these recent developments have provided significant insight into the design of microenvironments that can elicit the targeted cellular response. In this article, we will focus on some of these developments with an emphasis on matrix-mediated expansion of human pluripotent stem cells while maintaining their pluripotency. We will also discuss the role of matrix-based cues and cell-cell interactions in the form of soluble signals in directing stem cell differentiation into musculoskeletal lineages.
Collapse
Affiliation(s)
- Yongsung Hwang
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0412, USA
| | | | | |
Collapse
|
1073
|
Bratt-Leal AM, Kepple KL, Carpenedo RL, Cooke MT, McDevitt TC. Magnetic manipulation and spatial patterning of multi-cellular stem cell aggregates. Integr Biol (Camb) 2011; 3:1224-32. [PMID: 22076329 DOI: 10.1039/c1ib00064k] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The controlled assembly and organization of multi-cellular systems to mimic complex tissue structures is critical to the engineering of tissues for therapeutic and diagnostic applications. Recent advances in micro-scale technologies to control multi-cellular aggregate formation typically require chemical modification of the interface between cells and materials and lack multi-scale flexibility. Here we demonstrate that simple physical entrapment of magnetic microparticles within the extracellular space of stem cells spheroids during initial formation enables scaffold-free immobilization, translocation and directed assembly of multi-cellular aggregates across multiple length and time scales, even under dynamic suspension culture conditions. The response of aggregates to externally applied magnetic fields was a direct function of microparticle incorporation, allowing for rapid and transient control of the extracellular environment as well as separation of heterogeneous populations. In addition, spatial patterning of heterogeneous spheroid populations as well as individual multi-cellular aggregates was readily achieved by imposing temporary magnetic fields. Overall, this approach provides novel routes to examine stem cell differentiation and tissue morphogenesis with applications that encompass the creation of new model systems for developmental biology, scaffold-free tissue engineering strategies and scalable bioprocessing technologies.
Collapse
Affiliation(s)
- Andrés M Bratt-Leal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, USA
| | | | | | | | | |
Collapse
|
1074
|
Mahmoudifar N, Doran PM. Chondrogenesis and cartilage tissue engineering: the longer road to technology development. Trends Biotechnol 2011; 30:166-76. [PMID: 22071143 DOI: 10.1016/j.tibtech.2011.09.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 09/20/2011] [Accepted: 09/20/2011] [Indexed: 12/31/2022]
Abstract
Joint injury and disease are painful and debilitating conditions affecting a substantial proportion of the population. The idea that damaged cartilage in articulating joints might be replaced seamlessly with tissue-engineered cartilage is of obvious commercial interest because the market for such treatments is large. Recently, a wealth of new information about the complex biology of chondrogenesis and cartilage has emerged from stem cell research, including increasing evidence of the role of physical stimuli in directing differentiation. The challenge for the next generation of tissue engineers is to identify the key elements in this new body of knowledge that can be applied to overcome current limitations affecting cartilage synthesis in vitro. Here we review the status of cartilage tissue engineering and examine the contribution of stem cell research to technology development for cartilage production.
Collapse
Affiliation(s)
- Nastaran Mahmoudifar
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | | |
Collapse
|
1075
|
Nanostructuring PEG-fibrinogen hydrogels to control cellular morphogenesis. Biomaterials 2011; 32:7839-46. [DOI: 10.1016/j.biomaterials.2011.06.078] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 06/30/2011] [Indexed: 12/11/2022]
|
1076
|
Steinmetz NJ, Bryant SJ. The effects of intermittent dynamic loading on chondrogenic and osteogenic differentiation of human marrow stromal cells encapsulated in RGD-modified poly(ethylene glycol) hydrogels. Acta Biomater 2011; 7:3829-40. [PMID: 21742067 DOI: 10.1016/j.actbio.2011.06.031] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/28/2011] [Accepted: 06/21/2011] [Indexed: 10/18/2022]
Abstract
Biochemical and biomechanical cues are known to influence the differentiation of stem cells. Biomechanical cues arise from cellular interactions with their surrounding matrix and from applied forces. This study investigates the role of biomechanical cues in chondrogenic and osteogenic differentiation of human marrow stromal cells (hMSC) when encapsulated in synthetic hydrogels. Poly(ethylene glycol) hydrogels were fabricated with tethered cell adhesion moieties, RGD. Cell-laden hydrogels were subjected to 4 h daily intermittent dynamic compressive loading (0.3Hz, 15% amplitude strain) for up to 14 days and the cell response evaluated by gene expression and matrix deposition for chondrogenic and osteogenic markers. The three-dimensional hydrogel supported chondrogenesis and osteogenesis under free swelling conditions, as shown by the up-regulation of cartilage-related markers (SOX9, Col II, Col X, and aggrecan) and staining for type II collagen and aggrecan and osteogenically by up-regulation of ALP and staining for type I collagen and for mineralization. However, under dynamic loading the expression of cartilage-related markers SOX9, Col II, Col X, and aggrecan were down-regulated, along with reduced aggrecan staining and no positive staining for type II collagen. Additionally, the bone-related markers RUNX2, Col I, and ALP were down-regulated and positive staining for type I collagen and mineralization was reduced. In conclusion, the selected loading regime appears to have an inhibitory effect on chondrogenesis and osteogenesis of hMSC encapsulated in PEG-RGD hydrogels after 14 days in culture, potentially due to overloading of the differentiating hMSC before sufficient pericellular matrix is produced and/or due to large strains, particularly for osteogenically differentiating hMSC.
Collapse
|
1077
|
Wilson MJ, Liliensiek SJ, Murphy CJ, Murphy WL, Nealey PF. Hydrogels with well-defined peptide-hydrogel spacing and concentration: impact on epithelial cell behavior(). SOFT MATTER 2011; 8:390-398. [PMID: 23264803 PMCID: PMC3526380 DOI: 10.1039/c1sm06589k] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The spacing of peptides away from a hydrogel matrix dramatically impacts their availability and subsequent interactions with cells. Peptides were synthesized with monodisperse poly(ethylene glycol) spacers of different lengths that separate the peptide from the monomeric functionality which reacts during hydrogel polymerization. Specifically, bioactive RGD ligands were conjugated to PEG(5), PEG(11) or PEG(27) spacers via solid phase techniques and then functionalized with an acryloyl end group. These acryloyl-PEGx-RGD conjugates were then copolymerized with PEGDA to form an inert hydrogel network decorated with RGD ligands for cell interactions. As the PEG spacer length increases, the RGD concentration required to support cell attachment and spreading decreases. The competitive detachment of hTCEpi cells in the presence of soluble linear RGD also shows non-linear dependence on the PEG spacer length, as more cells remained attached and spread on gels functionalized with longer PEG-RGD conjugates in comparison to the shorter PEG-RGD conjugates. The strategy and synthetic techniques developed here allow for reproducible control over peptide-hydrogel spacing and peptide concentration, and may be extended for incorporation of multiple peptides and to other hydrogel platforms.
Collapse
Affiliation(s)
- Michelle J. Wilson
- Department of Chemical and Biological Engineering, The University of Wisconsin, Madison, WI, 53706, USA
| | - Sara J. Liliensiek
- Department of Chemical and Biological Engineering, The University of Wisconsin, Madison, WI, 53706, USA
| | - Christopher J. Murphy
- Department of Opthalmology and Vision Science, University of California, Davis, CA, 95616, USA
| | - William L. Murphy
- Department of Biomedical Engineering, The University of Wisconsin, Madison, WI, 53706, USA
| | - Paul F. Nealey
- Department of Chemical and Biological Engineering, The University of Wisconsin, Madison, WI, 53706, USA
| |
Collapse
|
1078
|
Gribova V, Crouzier T, Picart C. A material's point of view on recent developments of polymeric biomaterials: control of mechanical and biochemical properties. JOURNAL OF MATERIALS CHEMISTRY 2011; 21:14354-14366. [PMID: 25067892 PMCID: PMC4111539 DOI: 10.1039/c1jm11372k] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cells respond to a variety of stimuli, including biochemical, topographical and mechanical signals originating from their micro-environment. Cell responses to the mechanical properties of their substrates have been increasingly studied for about 14 years. To this end, several types of materials based on synthetic and natural polymers have been developed. Presentation of biochemical ligands to the cells is also important to provide additional functionalities or more selectivity in the details of cell/material interaction. In this review article, we will emphasize the development of synthetic and natural polymeric materials with well-characterized and tunable mechanical properties. We will also highlight how biochemical signals can be presented to the cells by combining them with these biomaterials. Such developments in materials science are not only important for fundamental biophysical studies on cell/material interactions but also for the design of a new generation of advanced and highly functional biomaterials.
Collapse
Affiliation(s)
- Varvara Gribova
- LMGP-MINATEC, Grenoble Institute of Technology, 3 parvis Louis Néel 38016 Grenoble, France
| | - Thomas Crouzier
- LMGP-MINATEC, Grenoble Institute of Technology, 3 parvis Louis Néel 38016 Grenoble, France
| | - Catherine Picart
- LMGP-MINATEC, Grenoble Institute of Technology, 3 parvis Louis Néel 38016 Grenoble, France
| |
Collapse
|
1079
|
Abstract
It is increasingly recognized that material surface topography is able to evoke specific cellular responses, endowing materials with instructive properties that were formerly reserved for growth factors. This opens the window to improve upon, in a cost-effective manner, biological performance of any surface used in the human body. Unfortunately, the interplay between surface topographies and cell behavior is complex and still incompletely understood. Rational approaches to search for bioactive surfaces will therefore omit previously unperceived interactions. Hence, in the present study, we use mathematical algorithms to design nonbiased, random surface features and produce chips of poly(lactic acid) with 2,176 different topographies. With human mesenchymal stromal cells (hMSCs) grown on the chips and using high-content imaging, we reveal unique, formerly unknown, surface topographies that are able to induce MSC proliferation or osteogenic differentiation. Moreover, we correlate parameters of the mathematical algorithms to cellular responses, which yield novel design criteria for these particular parameters. In conclusion, we demonstrate that randomized libraries of surface topographies can be broadly applied to unravel the interplay between cells and surface topography and to find improved material surfaces.
Collapse
|
1080
|
Cha C, Jeong JH, Shim J, Kong H. Tuning the dependency between stiffness and permeability of a cell encapsulating hydrogel with hydrophilic pendant chains. Acta Biomater 2011; 7:3719-28. [PMID: 21704737 DOI: 10.1016/j.actbio.2011.06.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 06/09/2011] [Accepted: 06/10/2011] [Indexed: 11/24/2022]
Abstract
The mechanical stiffness of a hydrogel plays a significant role in regulating the phenotype of cells that adhere to its surface. However, the effect of hydrogel stiffness on cells cultured within its matrix is not well understood, because of the intrinsic inverse dependency between the permeability and stiffness of hydrogels. This study therefore presents an advanced biomaterial design strategy to decrease the inverse dependency between permeability and stiffness of a cell encapsulating hydrogel. Hydrogels were made by cross-linking poly(ethylene glycol) diacrylate (PEGDA) and poly(ethylene glycol) monoacrylate (PEGMA), with PEGMA acting as a pendant polymer chain. Increasing the mass fraction of PEGMA while keeping the total polymer concentration constant led to a decrease in the elastic modulus (E) of the hydrogel, but caused a minimal increase in the swelling ratio (Q). The size and hydrophobicity of the end groups of pendant PEG chains further fine tuned the dependency between Q and E of the hydrogel. Pure PEGDA hydrogels with varying molecular weights, which show the same range of E but a much greater range of Q, were used as a control. Fibroblasts encapsulated in PEGDA-PEGMA hydrogels displayed more significant biphasic dependencies of cell viability and vascular endothelial growth factor (VEGF) expression on E than those encapsulated in pure PEGDA hydrogels, which were greatly influenced by Q. Overall, the hydrogel design strategy presented in this study will be highly useful to better regulate the phenotype and ultimately improve the therapeutic efficacy of a wide array of cells used in various biology studies and clinical settings.
Collapse
|
1081
|
Synergistic regulation of cell function by matrix rigidity and adhesive pattern. Biomaterials 2011; 32:9584-93. [PMID: 21955687 DOI: 10.1016/j.biomaterials.2011.09.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 09/01/2011] [Indexed: 11/23/2022]
Abstract
Cell-extracellular matrix (ECM) interactions play a critical role in regulating cellular behaviors. Recent studies of cell-ECM interactions have mainly focused on the actomyosin based and adhesion mediated mechanosensing pathways to understand how individual mechanical signals in the cell microenvironment, such as matrix rigidity and adhesive ECM pattern, are sensed by the cell and further trigger downstream intracellular signaling cascades and cellular responses. However, synergistic and collective regulation of cellular behaviors by matrix rigidity and adhesive ECM pattern are still elusive and largely uncharacterized. Here, we generated a library of microfabricated polydimethylsiloxane (PDMS) micropost arrays to study the synergistic and independent effects of matrix rigidity and adhesive ECM pattern on mechanoresponsive behaviors of both NIH/3T3 fibroblasts and human umbilical vein endothelial cells (HUVECs). We showed that both cell types were mechanosensitive and their cell spreading, FA formation, cytoskeletal contractility, and proliferation were all strongly dependent on both substrate rigidity and adhesive ECM pattern. We further showed that under the same substrate rigidity condition, smaller and closer adhesive ECM islands would cause both cells to spread out more, form more adhesion structures, and have a higher proliferation rate. The influence of adhesive ECM pattern on rigidity-mediated cytoskeletal contractility was cell type specific and was only significant for NIH/3T3. Morphometric analysis of cell populations revealed a strong correlation between focal adhesion and cell spreading, regardless of substrate rigidity and adhesive ECM pattern. We also observed a strong correlation between cellular traction force and cell spreading, with a substantially smaller independent effect of substrate rigidity on traction force. Our study here had determined key aspects of the biomechanical responses of adherent cells to independent and collective changes of substrate rigidity and adhesive ECM pattern.
Collapse
|
1082
|
An algorithm-based topographical biomaterials library to instruct cell fate. Proc Natl Acad Sci U S A 2011; 108:16565-70. [PMID: 21949368 DOI: 10.1073/pnas.1109861108] [Citation(s) in RCA: 274] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is increasingly recognized that material surface topography is able to evoke specific cellular responses, endowing materials with instructive properties that were formerly reserved for growth factors. This opens the window to improve upon, in a cost-effective manner, biological performance of any surface used in the human body. Unfortunately, the interplay between surface topographies and cell behavior is complex and still incompletely understood. Rational approaches to search for bioactive surfaces will therefore omit previously unperceived interactions. Hence, in the present study, we use mathematical algorithms to design nonbiased, random surface features and produce chips of poly(lactic acid) with 2,176 different topographies. With human mesenchymal stromal cells (hMSCs) grown on the chips and using high-content imaging, we reveal unique, formerly unknown, surface topographies that are able to induce MSC proliferation or osteogenic differentiation. Moreover, we correlate parameters of the mathematical algorithms to cellular responses, which yield novel design criteria for these particular parameters. In conclusion, we demonstrate that randomized libraries of surface topographies can be broadly applied to unravel the interplay between cells and surface topography and to find improved material surfaces.
Collapse
|
1083
|
Moraes C, Sun Y, Simmons CA. (Micro)managing the mechanical microenvironment. Integr Biol (Camb) 2011; 3:959-71. [PMID: 21931883 DOI: 10.1039/c1ib00056j] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mechanical forces are critical components of the cellular microenvironment and play a pivotal role in driving cellular processes in vivo. Dissecting cellular responses to mechanical forces is challenging, as even "simple" mechanical stimulation in vitro can cause multiple interdependent changes in the cellular microenvironment. These stimuli include solid deformation, fluid flows, altered physical and chemical surface features, and a complex transfer of loads between the various interacting components of a biological culture system. The active mechanical and biochemical responses of cells to these stimuli in generating internal forces, reorganizing cellular structures, and initiating intracellular signals that specify cell fate and remodel the surrounding environment further complicates cellular response to mechanical forces. Moreover, cells present a non-linear response to combinations of mechanical forces, materials, chemicals, surface features, matrix properties and other effectors. Microtechnology-based approaches to these challenges can yield key insights into the mechanical nature of cellular behaviour, by decoupling stimulation parameters; enabling multimodal control over combinations of stimuli; and increasing experimental throughput to systematically probe cellular response. In this critical review, we briefly discuss the complexities inherent in the mechanical stimulation of cells; survey and critically assess the applications of present microtechnologies in the field of experimental mechanobiology; and explore opportunities and possibilities to use these tools to obtain a deeper understanding of mechanical interactions between cells and their environment.
Collapse
Affiliation(s)
- Christopher Moraes
- Department of Mechanical & Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario M5S 3G8, Canada
| | | | | |
Collapse
|
1084
|
You JO, Rafat M, Ye GJC, Auguste DT. Nanoengineering the heart: conductive scaffolds enhance connexin 43 expression. NANO LETTERS 2011; 11:3643-8. [PMID: 21800912 DOI: 10.1021/nl201514a] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Scaffolds that couple electrical and elastic properties may be valuable for cardiac cell function. However, existing conductive materials do not mimic physiological properties. We prepared and characterized a tunable, hybrid hydrogel scaffold based on Au nanoparticles homogeneously synthesized throughout a polymer templated gel. Conductive gels had Young's moduli more similar to myocardium relative to polyaniline and polypyrrole, by 1-4 orders of magnitude. Neonatal rat cardiomyocytes exhibited increased expression of connexin 43 on hybrid scaffolds relative to HEMA with or without electrical stimulation.
Collapse
Affiliation(s)
- Jin-Oh You
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| | | | | | | |
Collapse
|
1085
|
Lopez JI, Kang I, You WK, McDonald DM, Weaver VM. In situ force mapping of mammary gland transformation. Integr Biol (Camb) 2011; 3:910-21. [PMID: 21842067 PMCID: PMC3564969 DOI: 10.1039/c1ib00043h] [Citation(s) in RCA: 211] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tumor progression is characterized by an incremental stiffening of the tissue. The importance of tissue rigidity to cancer is appreciated, yet the contribution of specific tissue elements to tumor stiffening and their physiological significance remains unclear. We performed high-resolution atomic force microscopy indentation in live and snap-frozen fluorescently labeled mammary tissues to explore the origin of the tissue stiffening associated with mammary tumor development in PyMT mice. The tumor epithelium, the tumor-associated vasculature and the extracellular matrix all contributed to mammary gland stiffening as it transitioned from normal to invasive carcinoma. Consistent with the concept that extracellular matrix stiffness modifies cell tension, we found that isolated transformed mammary epithelial cells were intrinsically stiffer than their normal counterparts but that the malignant epithelium in situ was far stiffer than isolated breast tumor cells. Moreover, using an in situ vitrification approach, we determined that the extracellular matrix adjacent to the epithelium progressively stiffened as tissue evolved from normal through benign to an invasive state. Importantly, we also noted that there was significant mechanical heterogeneity within the transformed tissue both in the epithelium and the tumor-associated neovasculature. The vascular bed within the tumor core was substantially stiffer than the large patent vessels at the invasive front that are surrounded by the stiffest extracellular matrix. These findings clarify the contribution of individual mammary gland tissue elements to the altered biomechanical landscape of cancerous tissues and emphasize the importance of studying cancer cell evolution under conditions that preserve native interactions.
Collapse
Affiliation(s)
- Jose I. Lopez
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, Department of Surgery, 513 Parnassus Ave, HSE 565, San Francisco, CA 94143-0456
| | - Inkyung Kang
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, Department of Surgery, 513 Parnassus Ave, HSE 565, San Francisco, CA 94143-0456
| | - Weon-Kyoo You
- Department of Anatomy, University of California at San Francisco, San Francisco, CA 94143
| | - Donald M. McDonald
- Department of Anatomy, University of California at San Francisco, San Francisco, CA 94143
| | - Valerie M. Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, Department of Surgery, 513 Parnassus Ave, HSE 565, San Francisco, CA 94143-0456
- Departments of Anatomy and Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
1086
|
Gojgini S, Tokatlian T, Segura T. Utilizing cell-matrix interactions to modulate gene transfer to stem cells inside hyaluronic acid hydrogels. Mol Pharm 2011; 8:1582-91. [PMID: 21823632 DOI: 10.1021/mp200171d] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The effective delivery of DNA locally would increase the applicability of gene therapy in tissue regeneration, where diseased tissue is to be repaired in situ. One promising approach is to use hydrogel scaffolds to encapsulate and deliver plasmid DNA in the form of nanoparticles to the diseased tissue, so that cells infiltrating the scaffold are transfected to induce regeneration. This study focuses on the design of a DNA nanoparticle-loaded hydrogel scaffold. In particular, this study focuses on understanding how cell-matrix interactions affect gene transfer to adult stem cells cultured inside matrix metalloproteinase (MMP) degradable hyaluronic acid (HA) hydrogel scaffolds. HA was cross-linked to form a hydrogel material using a MMP degradable peptide and Michael addition chemistry. Gene transfer inside these hydrogel materials was assessed as a function of polyplex nitrogen to phosphate ratio (N/P = 5 to 12), matrix stiffness (100-1700 Pa), RGD (Arg-Gly-Asp) concentration (10-400 μM), and RGD presentation (0.2-4.7 RGDs per HA molecule). All variables were found to affect gene transfer to mouse mensenchymal stem cells culture inside the DNA loaded hydrogels. As expected, higher N/P ratios lead to higher gene transfer efficiency but also higher toxicity; softer hydrogels resulted in higher transgene expression than stiffer hydrogels, and an intermediate RGD concentration and RGD clustering resulted in higher transgene expression. We believe that the knowledge gained through this in vitro model can be utilized to design better scaffold-mediated gene delivery for local gene therapy.
Collapse
Affiliation(s)
- Shiva Gojgini
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California 90095, United States
| | | | | |
Collapse
|
1087
|
Thein-Han W, Xu HHK. Collagen-calcium phosphate cement scaffolds seeded with umbilical cord stem cells for bone tissue engineering. Tissue Eng Part A 2011; 17:2943-54. [PMID: 21851269 DOI: 10.1089/ten.tea.2010.0674] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Human umbilical cord mesenchymal stem cells (hUCMSCs) avoid the invasive procedure required to harvest bone marrow MSCs. The addition of collagen fibers into self-setting calcium phosphate cement (CPC) may increase the scaffold strength, and enhance cell attachment and differentiation. The objectives of this study were to develop a novel class of collagen-CPC composite scaffolds, and to investigate hUCMSC attachment, proliferation, and osteogenic differentiation on collagen-CPC scaffolds for the first time. Collagen fibers in CPC improved the load-bearing capability. Flow cytometry showed that the hUCMSCs expressed cell surface markers characteristic of MSCs, and were negative for hematopoietic and endothelial cell markers. hUCMSCs proliferated rapidly in all CPC composite scaffolds, with cell number increasing by sevenfold in 8 days. Cellular function was enhanced with collagen fibers in CPC scaffolds. Cell density increased from (645±60) cells/mm(2) on CPC with 0% collagen, to (1056±65) cells/mm(2) on CPC with 8% collagen (p<0.05). The actin stress fibers inside the hUCMSCs were stained, and the fluorescence intensity was doubled when the collagen in CPC was increased by 0% to 8%. RT-PCR showed that hUCMSCs on CPC with collagen had higher osteogenic expression than those on CPC without collagen. Alizarin Red S staining revealed a great increase in mineralization by hUCMSCs on CPC with collagen than that without collagen. In conclusion, hUCMSCs showed excellent proliferation, differentiation, and synthesis of bone minerals in collagen-CPC composite scaffolds for the first time. The novel hUCMSC-seeded collagen-CPC construct with superior cell function and load-bearing capability is promising to enhance bone regeneration in a wide range of orthopedic and craniofacial applications.
Collapse
Affiliation(s)
- WahWah Thein-Han
- Biomaterials and Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | | |
Collapse
|
1088
|
Park JS, Chu JS, Tsou AD, Diop R, Tang Z, Wang A, Li S. The effect of matrix stiffness on the differentiation of mesenchymal stem cells in response to TGF-β. Biomaterials 2011; 32:3921-30. [PMID: 21397942 DOI: 10.1016/j.biomaterials.2011.02.019] [Citation(s) in RCA: 545] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 02/10/2011] [Indexed: 11/27/2022]
Abstract
Bone marrow mesenchymal stem cells (MSCs) are a valuable cell source for tissue engineering and regenerative medicine. Transforming growth factor β (TGF-β) can promote MSC differentiation into either smooth muscle cells (SMCs) or chondrogenic cells. Here we showed that the stiffness of cell adhesion substrates modulated these differential effects. MSCs on soft substrates had less spreading, fewer stress fibers and lower proliferation rate than MSCs on stiff substrates. MSCs on stiff substrates had higher expression of SMC markers α-actin and calponin-1; in contrast, MSCs on soft substrates had a higher expression of chondrogenic marker collagen-II and adipogenic marker lipoprotein lipase (LPL). TGF-β increased SMC marker expression on stiff substrates. However, TGF-β increased chondrogenic marker expression and suppressed adipogenic marker expression on soft substrates, while adipogenic medium and soft substrates induced adipogenic differentiation effectively. Rho GTPase was involved in the expression of all aforementioned lineage markers, but did not account for the differential effects of substrate stiffness. In addition, soft substrates did not significantly affect Rho activity, but inhibited Rho-induced stress fiber formation and α-actin assembly. Further analysis showed that MSCs on soft substrates had weaker cell adhesion, and that the suppression of cell adhesion strength mimicked the effects of soft substrates on the lineage marker expression. These results provide insights of how substrate stiffness differentially regulates stem cell differentiation, and have significant implications for the design of biomaterials with appropriate mechanical property for tissue regeneration.
Collapse
Affiliation(s)
- Jennifer S Park
- Department of Bioengineering, University of California, Berkeley, B108A Stanley Hall, Berkeley, CA 94720-1762, USA
| | | | | | | | | | | | | |
Collapse
|
1089
|
Ayala R, Zhang C, Yang D, Hwang Y, Aung A, Shroff SS, Arce FT, Lal R, Arya G, Varghese S. Engineering the cell-material interface for controlling stem cell adhesion, migration, and differentiation. Biomaterials 2011; 32:3700-11. [PMID: 21396708 DOI: 10.1016/j.biomaterials.2011.02.004] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 02/01/2011] [Indexed: 12/12/2022]
Abstract
The effective utilization of stem cells in regenerative medicine critically relies upon our understanding of the intricate interactions between cells and their extracellular environment. While bulk mechanical and chemical properties of the matrix have been shown to influence various cellular functions, the role of matrix interfacial properties on stem cell behavior is unclear. Here, we report the striking effect of matrix interfacial hydrophobicity on stem cell adhesion, motility, cytoskeletal organization, and differentiation. This is achieved through the development of tunable, synthetic matrices with control over their hydrophobicity without altering the chemical and mechanical properties of the matrix. The observed cellular responses are explained in terms of hydrophobicity-driven conformational changes of the pendant side chains at the interface leading to differential binding of proteins. These results demonstrate that the hydrophobicity of the extracellular matrix could play a considerably larger role in dictating cellular behaviors than previously anticipated. Additionally, these tunable matrices, which introduce a new control feature for regulating various cellular functions offer a platform for studying proliferation and differentiation of stem cells in a controlled manner and would have applications in regenerative medicine.
Collapse
Affiliation(s)
- Ramses Ayala
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, MC 0412, La Jolla, CA 92093, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
1090
|
Li D, Zhou J, Chowdhury F, Cheng J, Wang N, Wang F. Role of mechanical factors in fate decisions of stem cells. Regen Med 2011; 6:229-40. [PMID: 21391856 DOI: 10.2217/rme.11.2] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Stem cells derived from adult tissues or from the inner cell mass of blastocyst-stage embryos can self-renew in culture and have the remarkable potential to undergo lineage-specific differentiation. Extensive studies have been devoted to achieving a better understanding of the soluble factors and the mechanism(s) by which they regulate the fate decisions of these cells, but it is only recently that a critical role has been revealed for physical and mechanical factors in controlling self-renewal and lineage specification. This review summarizes selected aspects of current work on stem cell mechanics with an emphasis on the influence of matrix stiffness, surface topography, cell shape and mechanical forces on the fate determination of mesenchymal stem cells and embryonic stem cells.
Collapse
Affiliation(s)
- Dong Li
- Department of Cell & Developmental Biology & Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | | | | | |
Collapse
|
1091
|
D'Angelo F, Tiribuzi R, Armentano I, Kenny JM, Martino S, Orlacchio A. Mechanotransduction: tuning stem cells fate. J Funct Biomater 2011; 2:67-87. [PMID: 24956164 PMCID: PMC4030896 DOI: 10.3390/jfb2020067] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/07/2011] [Accepted: 06/17/2011] [Indexed: 01/10/2023] Open
Abstract
It is a general concern that the success of regenerative medicine-based applications is based on the ability to recapitulate the molecular events that allow stem cells to repair the damaged tissue/organ. To this end biomaterials are designed to display properties that, in a precise and physiological-like fashion, could drive stem cell fate both in vitro and in vivo. The rationale is that stem cells are highly sensitive to forces and that they may convert mechanical stimuli into a chemical response. In this review, we describe novelties on stem cells and biomaterials interactions with more focus on the implication of the mechanical stimulation named mechanotransduction.
Collapse
Affiliation(s)
- Francesco D'Angelo
- Department of Experimental Medicine and Biochemical Science, Section of Biochemistry and Molecular Biology, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Roberto Tiribuzi
- Department of Experimental Medicine and Biochemical Science, Section of Biochemistry and Molecular Biology, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Ilaria Armentano
- Materials Engineering Centre, UdR INSTM, NIPLAB, University of Perugia, Strada di Pentima 4, 05100 Terni, Italy.
| | - Josè Maria Kenny
- Materials Engineering Centre, UdR INSTM, NIPLAB, University of Perugia, Strada di Pentima 4, 05100 Terni, Italy.
| | - Sabata Martino
- Department of Experimental Medicine and Biochemical Science, Section of Biochemistry and Molecular Biology, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Aldo Orlacchio
- Department of Experimental Medicine and Biochemical Science, Section of Biochemistry and Molecular Biology, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| |
Collapse
|
1092
|
Abstract
The hallmarks of calcific aortic valve disease (CAVD) are the significant changes that occur in the organization, composition, and mechanical properties of the extracellular matrix (ECM), ultimately resulting in stiffened stenotic leaflets that obstruct flow and compromise cardiac function. Increasing evidence suggests that ECM maladaptations are not simply a result of valve cell dysfunction; they also contribute to CAVD progression by altering cellular and molecular signaling. In this review, we summarize the ECM changes that occur in CAVD. We also discuss examples of how the ECM influences cellular processes by signaling through adhesion receptors (matricellular signaling), by regulating the presentation and availability of growth factors and cytokines to cells (matricrine signaling), and by transducing externally applied forces and resisting cell-generated tractional forces (mechanical signaling) to regulate a wide range of pathological processes, including differentiation, fibrosis, calcification, and angiogenesis. Finally, we suggest areas for future research that should lead to new insights into bidirectional cell–ECM interactions in the aortic valve, their contributions to homeostasis and pathobiology, and possible targets to slow or prevent the progression of CAVD.
Collapse
Affiliation(s)
- Jan-Hung Chen
- From the Institute of Biomaterials and Biomedical Engineering (J.H.C., C.A.S.), Department of Mechanical and Industrial Engineering (J.H.C., C.A.S.), and Faculty of Dentistry (C.A.S.), University of Toronto, Toronto, Ontario, Canada
| | - Craig A. Simmons
- From the Institute of Biomaterials and Biomedical Engineering (J.H.C., C.A.S.), Department of Mechanical and Industrial Engineering (J.H.C., C.A.S.), and Faculty of Dentistry (C.A.S.), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
1093
|
Malmström J, Lovmand J, Kristensen S, Sundh M, Duch M, Sutherland DS. Focal complex maturation and bridging on 200 nm vitronectin but not fibronectin patches reveal different mechanisms of focal adhesion formation. NANO LETTERS 2011; 11:2264-2271. [PMID: 21598955 DOI: 10.1021/nl200447q] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The effects of protein type and pattern size on cell adhesion, spreading, and focal adhesion development are studied. Fibronectin and vitronectin patterns from 0.1 to 3 μm produced by colloidal lithography reveal important differences in how cells adhere to and bridge focal adhesions across protein nanopatterns versus micropatterns. Vinculin and zyxin in focal adhesions but not integrins are seen to bridge ligand gaps. Differences in protein mechanical properties are implicated as important factors in focal adhesion development.
Collapse
Affiliation(s)
- Jenny Malmström
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, DK-8000 Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
1094
|
Wheeldon I, Farhadi A, Bick AG, Jabbari E, Khademhosseini A. Nanoscale tissue engineering: spatial control over cell-materials interactions. NANOTECHNOLOGY 2011; 22:212001. [PMID: 21451238 PMCID: PMC3155808 DOI: 10.1088/0957-4484/22/21/212001] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cells interact with the surrounding environment by making tens to hundreds of thousands of nanoscale interactions with extracellular signals and features. The goal of nanoscale tissue engineering is to harness these interactions through nanoscale biomaterials engineering in order to study and direct cellular behavior. Here, we review two- and three-dimensional (2- and 3D) nanoscale tissue engineering technologies, and provide a holistic overview of the field. Techniques that can control the average spacing and clustering of cell adhesion ligands are well established and have been highly successful in describing cell adhesion and migration in 2D. Extension of these engineering tools to 3D biomaterials has created many new hydrogel and nanofiber scaffold technologies that are being used to design in vitro experiments with more physiologically relevant conditions. Researchers are beginning to study complex cell functions in 3D. However, there is a need for biomaterials systems that provide fine control over the nanoscale presentation of bioactive ligands in 3D. Additionally, there is a need for 2- and 3D techniques that can control the nanoscale presentation of multiple bioactive ligands and that can control the temporal changes in the cellular microenvironment.
Collapse
Affiliation(s)
- Ian Wheeldon
- Department of Medicine, Center for Biomedical Engineering, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arash Farhadi
- Department of Medicine, Center for Biomedical Engineering, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Nanotechnology Engineering Program, University of Waterloo, Waterloo, ON, N2L 3G1 Canada
| | - Alexander G. Bick
- Department of Medicine, Center for Biomedical Engineering, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Esmaiel Jabbari
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, 29208
| | - Ali Khademhosseini
- Department of Medicine, Center for Biomedical Engineering, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
1095
|
Concentration independent modulation of local micromechanics in a fibrin gel. PLoS One 2011; 6:e20201. [PMID: 21629793 PMCID: PMC3100350 DOI: 10.1371/journal.pone.0020201] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 04/27/2011] [Indexed: 11/19/2022] Open
Abstract
Methods for tuning extracellular matrix (ECM) mechanics in 3D cell culture that rely on increasing the concentration of either protein or cross-linking molecules fail to control important parameters such as pore size, ligand density, and molecular diffusivity. Alternatively, ECM stiffness can be modulated independently from protein concentration by mechanically loading the ECM. We have developed a novel device for generating stiffness gradients in naturally derived ECMs, where stiffness is tuned by inducing strain, while local mechanical properties are directly determined by laser tweezers based active microrheology (AMR). Hydrogel substrates polymerized within 35 mm diameter Petri dishes are strained non-uniformly by the precise rotation of an embedded cylindrical post, and exhibit a position-dependent stiffness with little to no modulation of local mesh geometry. Here we present the device in the context of fibrin hydrogels. First AMR is used to directly measure local micromechanics in unstrained hydrogels of increasing fibrin concentration. Changes in stiffness are then mapped within our device, where fibrin concentration is held constant. Fluorescence confocal imaging and orbital particle tracking are used to quantify structural changes in fibrin on the micro and nano levels respectively. The micromechanical strain stiffening measured by microrheology is not accompanied by ECM microstructural changes under our applied loads, as measured by confocal microscopy. However, super-resolution orbital tracking reveals nanostructural straightening, lengthening, and reduced movement of fibrin fibers. Furthermore, we show that aortic smooth muscle cells cultured within our device are morphologically sensitive to the induced mechanical gradient. Our results demonstrate a powerful cell culture tool that can be used in the study of mechanical effects on cellular physiology in naturally derived 3D ECM tissues.
Collapse
|
1096
|
Integrin activation and internalization on soft ECM as a mechanism of induction of stem cell differentiation by ECM elasticity. Proc Natl Acad Sci U S A 2011; 108:9466-71. [PMID: 21593411 DOI: 10.1073/pnas.1106467108] [Citation(s) in RCA: 252] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The mechanism by which ECM elasticity induces lineage specification of stem cells has not been clearly understood. Integrins are well-documented mechanosensors that are positioned at the beginning of the sensing pathway. By using an antibody specifically recognizing the active conformation of β1 integrin, we observed that β1 integrin activation in bone marrow mesenchymal stem cells (BMMSCs) was induced by soft substrate to a significantly greater degree than by stiff substrate. In contrast, however, the level of cell surface integrin on soft substrate was significantly lower than that on stiff substrate. Soft substrate markedly enhanced the internalization of integrin, and this internalization was mediated mainly through caveolae/raft-dependent endocytosis. The inhibition of integrin internalization blocked the neural lineage specification of BMMSCs on soft substrate. Furthermore, soft substrate also repressed the bone morphogenetic protein (BMP)/Smad pathway at least partially through integrin-regulated BMP receptor endocytosis. A theoretical analysis based on atomic force microscopy (AFM) data indicated that integrin-ligand complexes are more easily ruptured on soft substrate; this outcome may contribute to the enhancement of integrin internalization on soft substrate. Taken together, our results suggest that ECM elasticity affects integrin activity and trafficking to modulate integrin BMP receptor internalization, thus contributing to stem cell lineage specification.
Collapse
|
1097
|
Lessons from (patho)physiological tissue stiffness and their implications for drug screening, drug delivery and regenerative medicine. Adv Drug Deliv Rev 2011; 63:269-76. [PMID: 21241759 DOI: 10.1016/j.addr.2011.01.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Revised: 12/27/2010] [Accepted: 01/05/2011] [Indexed: 02/07/2023]
Abstract
Diseased tissues are noted for their compromised mechanical properties, which contribute to organ failure; regeneration entails restoration of tissue structure and thereby functions. Thus, the physical signature of a tissue is closely associated with its biological function. In this review, we consider a mechanics-centric view of disease and regeneration by drawing parallels between in vivo tissue-level observations and corroborative cellular evidence in vitro to demonstrate the importance of the mechanical stiffness of the extracellular matrix in these processes. This is not intended to devalue the importance of biochemical signaling; in fact, as we discuss, many mechanical stiffness-driven processes not only require cooperation with biochemical cues, but they ultimately converge at common signaling cascades to influence cell and tissue function in an integrative manner. The study of how physical and biochemical signals collectively modulate cell function not only brings forth a more holistic understanding of cell (patho)biology, but it also creates opportunities to control material properties to improve culture platforms for research and drug screening and aid in the rationale design of biomaterials for molecular therapy and tissue engineering applications.
Collapse
|
1098
|
Bron JL, Vonk LA, Smit TH, Koenderink GH. Engineering alginate for intervertebral disc repair. J Mech Behav Biomed Mater 2011; 4:1196-205. [PMID: 21783128 DOI: 10.1016/j.jmbbm.2011.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 04/02/2011] [Accepted: 04/04/2011] [Indexed: 10/18/2022]
Abstract
Alginate is frequently studied as a scaffold for intervertebral disc (IVD) repair, since it closely mimics mechanical and cell-adhesive properties of the nucleus pulposus (NP) of the IVD. The aim of this study was to assess the relation between alginate concentration and scaffold stiffness and find preparation conditions where the viscoelastic behaviour mimics that of the NP. In addition, we measured the effect of variations in scaffold stiffness on the expression of extracellular matrix molecules specific to the NP (proteoglycans and collagen) by native NP cells. We prepared sample discs of different concentrations of alginate (1%-6%) by two different methods, diffusion and in situ gelation. The stiffness increased with increasing alginate concentration, while the loss tangent (dissipative behaviour) remained constant. The diffusion samples were ten-fold stiffer than samples prepared by in situ gelation. Sample discs prepared from 2% alginate by diffusion closely matched the stiffness and loss tangent of the NP. The stiffness of all samples declined upon prolonged incubation in medium, especially for samples prepared by diffusion. The biosynthetic phenotype of native cells isolated from NPs was preserved in alginate matrices up to 4 weeks of culturing. Gene expression levels of extracellular matrix components were insensitive to alginate concentration and corresponding matrix stiffness, likely due to the poor adhesiveness of the cells to alginate. In conclusion, alginate can mimic the viscoelastic properties of the NP and preserve the biosynthetic phenotype of NP cells but certain limitations like long-term stability still have to be addressed.
Collapse
Affiliation(s)
- Johannes L Bron
- Department of Orthopaedic Surgery, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
1099
|
Bencherif SA, Guillemot F, Huebsch N, Edwards DA, Mooney DJ. [Cell-traction mediated configuration of the cell/extracellular-matrix interface plays a key role in stem cell fate]. Med Sci (Paris) 2011; 27:19-21. [PMID: 21299954 DOI: 10.1051/medsci/201127119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
1100
|
Chew SY, Low WC. Scaffold-based approach to direct stem cell neural and cardiovascular differentiation: An analysis of physical and biochemical effects. J Biomed Mater Res A 2011; 97:355-74. [DOI: 10.1002/jbm.a.33064] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/11/2011] [Accepted: 01/24/2011] [Indexed: 01/12/2023]
|