1
|
Wareham LK, Baratta RO, Del Buono BJ, Schlumpf E, Calkins DJ. Collagen in the central nervous system: contributions to neurodegeneration and promise as a therapeutic target. Mol Neurodegener 2024; 19:11. [PMID: 38273335 PMCID: PMC10809576 DOI: 10.1186/s13024-024-00704-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
The extracellular matrix is a richly bioactive composition of substrates that provides biophysical stability, facilitates intercellular signaling, and both reflects and governs the physiological status of the local microenvironment. The matrix in the central nervous system (CNS) is far from simply an inert scaffold for mechanical support, instead conducting an active role in homeostasis and providing broad capacity for adaptation and remodeling in response to stress that otherwise would challenge equilibrium between neuronal, glial, and vascular elements. A major constituent is collagen, whose characteristic triple helical structure renders mechanical and biochemical stability to enable bidirectional crosstalk between matrix and resident cells. Multiple members of the collagen superfamily are critical to neuronal maturation and circuit formation, axon guidance, and synaptogenesis in the brain. In mature tissue, collagen interacts with other fibrous proteins and glycoproteins to sustain a three-dimensional medium through which complex networks of cells can communicate. While critical for matrix scaffolding, collagen in the CNS is also highly dynamic, with multiple binding sites for partnering matrix proteins, cell-surface receptors, and other ligands. These interactions are emerging as critical mediators of CNS disease and injury, particularly regarding changes in matrix stiffness, astrocyte recruitment and reactivity, and pro-inflammatory signaling in local microenvironments. Changes in the structure and/or deposition of collagen impact cellular signaling and tissue biomechanics in the brain, which in turn can alter cellular responses including antigenicity, angiogenesis, gliosis, and recruitment of immune-related cells. These factors, each involving matrix collagen, contribute to the limited capacity for regeneration of CNS tissue. Emerging therapeutics that attempt to rebuild the matrix using peptide fragments, including collagen-enriched scaffolds and mimetics, hold great potential to promote neural repair and regeneration. Recent evidence from our group and others indicates that repairing protease-degraded collagen helices with mimetic peptides helps restore CNS tissue and promote neuronal survival in a broad spectrum of degenerative conditions. Restoration likely involves bolstering matrix stiffness to reduce the potential for astrocyte reactivity and local inflammation as well as repairing inhibitory binding sites for immune-signaling ligands. Facilitating repair rather than endogenous replacement of collagen degraded by disease or injury may represent the next frontier in developing therapies based on protection, repair, and regeneration of neurons in the central nervous system.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute , Vanderbilt University Medical Center, 1161 21st Avenue S, 37232, Nashville, TN, USA
| | - Robert O Baratta
- Stuart Therapeutics, Inc., 411 SE Osceola St, 34994, Stuart, FL, USA
| | - Brian J Del Buono
- Stuart Therapeutics, Inc., 411 SE Osceola St, 34994, Stuart, FL, USA
| | - Eric Schlumpf
- Stuart Therapeutics, Inc., 411 SE Osceola St, 34994, Stuart, FL, USA
| | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute , Vanderbilt University Medical Center, 1161 21st Avenue S, 37232, Nashville, TN, USA
| |
Collapse
|
2
|
Liu Y, Zhang W, Hu C, Zheng C, Zhang F, Yang L, Li Z, Wang Y. A composite hydrogel improves the survival and differentiation of human iPSC-derived neural stem cells after ischemic stroke. COMPOSITES PART B: ENGINEERING 2023; 259:110711. [DOI: 10.1016/j.compositesb.2023.110711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Heide F, Koch M, Stetefeld J. Heparin Mimetics and Their Impact on Extracellular Matrix Protein Assemblies. Pharmaceuticals (Basel) 2023; 16:ph16030471. [PMID: 36986571 PMCID: PMC10059586 DOI: 10.3390/ph16030471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Heparan sulfate is a crucial extracellular matrix component that organizes structural features and functional protein processes. This occurs through the formation of protein-heparan sulfate assemblies around cell surfaces, which allow for the deliberate local and temporal control of cellular signaling. As such, heparin-mimicking drugs can directly affect these processes by competing with naturally occurring heparan sulfate and heparin chains that then disturb protein assemblies and decrease regulatory capacities. The high number of heparan-sulfate-binding proteins that are present in the extracellular matrix can cause obscure pathological effects that should be considered and examined in more detail, especially when developing novel mimetics for clinical use. The objective of this article is to investigate recent studies that present heparan-sulfate-mediated protein assemblies and the impact of heparin mimetics on the assembly and function of these protein complexes.
Collapse
Affiliation(s)
- Fabian Heide
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Manuel Koch
- Institute for Experimental Dental Research and Oral Musculoskeletal Biology, Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Jörg Stetefeld
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
4
|
Liang S, Su Y, Yao R. 3D Bioprinting of Induced Pluripotent Stem Cells and Disease Modeling. Handb Exp Pharmacol 2023; 281:29-56. [PMID: 36882603 DOI: 10.1007/164_2023_646] [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] [Indexed: 03/09/2023]
Abstract
Patient-derived induced pluripotent stem cells (iPSCs), carrying the genetic information of the disease and capable of differentiating into multilineages in vitro, are valuable for disease modeling. 3D bioprinting enables the assembly of the cell-laden hydrogel into hierarchically three-dimensional architectures that recapitulate the natural tissues and organs. Investigation of iPSC-derived physiological and pathological models constructed by 3D bioprinting is a fast-growing field still in its infancy. Distinctly from cell lines and adult stem cells, iPSCs and iPSC-derived cells are more susceptible to external stimuli which can disturb the differentiation, maturation, and organization of iPSCs and their progeny. Here we discuss the fitness of iPSCs and 3D bioprinting from the perspective of bioinks and printing technologies. We provide a timely review of the progress of 3D bioprinting iPSC-derived physiological and pathological models by exemplifying the relatively prosperous cardiac and neurological fields. We also discuss scientific rigors and highlight the remaining issues to offer a guiding framework for bioprinting-assisted personalized medicine.
Collapse
Affiliation(s)
- Shaojun Liang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China
| | - Yijun Su
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China
| | - Rui Yao
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering,, Tsinghua University, Beijing, China.
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, China.
| |
Collapse
|
5
|
Devel L, Guedeney N, Bregant S, Chowdhury A, Jean M, Legembre P. Role of metalloproteases in the CD95 signaling pathways. Front Immunol 2022; 13:1074099. [PMID: 36544756 PMCID: PMC9760969 DOI: 10.3389/fimmu.2022.1074099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/21/2022] [Indexed: 12/08/2022] Open
Abstract
CD95L (also known as FasL or CD178) is a member of the tumor necrosis family (TNF) superfamily. Although this transmembrane ligand has been mainly considered as a potent apoptotic inducer in CD95 (Fas)-expressing cells, more recent studies pointed out its role in the implementation of non-apoptotic signals. Accordingly, this ligand has been associated with the aggravation of inflammation in different auto-immune disorders and in the metastatic occurrence in different cancers. Although it remains to decipher all key factors involved in the ambivalent role of this ligand, accumulating clues suggest that while the membrane bound CD95L triggers apoptosis, its soluble counterpart generated by metalloprotease-driven cleavage is responsible for its non-apoptotic functions. Nonetheless, the metalloproteases (MMPs and ADAMs) involved in the CD95L shedding, the cleavage sites and the different stoichiometries and functions of the soluble CD95L remain to be elucidated. To better understand how soluble CD95L triggers signaling pathways from apoptosis to inflammation or cell migration, we propose herein to summarize the different metalloproteases that have been described to be able to shed CD95L, their cleavage sites and the biological functions associated with the released ligands. Based on these new findings, the development of CD95/CD95L-targeting therapeutics is also discussed.
Collapse
Affiliation(s)
- Laurent Devel
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Gif-sur-Yvette, France
| | - Nicolas Guedeney
- Université de Rennes 1, Institut des Sciences Chimiques de Rennes - UMR CNRS 6226 Equipe COrInt, Rennes, France
| | - Sarah Bregant
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, Gif-sur-Yvette, France
| | - Animesh Chowdhury
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Mickael Jean
- Université de Rennes 1, Institut des Sciences Chimiques de Rennes - UMR CNRS 6226 Equipe COrInt, Rennes, France
| | - Patrick Legembre
- CRIBL UMR CNRS 7276 INSERM 1262, Université de Limoges, Rue Marcland, Limoges, France,*Correspondence: Patrick Legembre,
| |
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
|
Stowers RS. Advances in Extracellular Matrix-Mimetic Hydrogels to Guide Stem Cell Fate. Cells Tissues Organs 2021; 211:703-720. [PMID: 34082418 DOI: 10.1159/000514851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 01/25/2023] Open
Abstract
In the fields of regenerative medicine and tissue engineering, stem cells offer vast potential for treating or replacing diseased and damaged tissue. Much progress has been made in understanding stem cell biology, yielding protocols for directing stem cell differentiation toward the cell type of interest for a specific application. One particularly interesting and powerful signaling cue is the extracellular matrix (ECM) surrounding stem cells, a network of biopolymers that, along with cells, makes up what we define as a tissue. The composition, structure, biochemical features, and mechanical properties of the ECM are varied in different tissues and developmental stages, and serve to instruct stem cells toward a specific lineage. By understanding and recapitulating some of these ECM signaling cues through engineered ECM-mimicking hydrogels, stem cell fate can be directed in vitro. In this review, we will summarize recent advances in material systems to guide stem cell fate, highlighting innovative methods to capture ECM functionalities and how these material systems can be used to provide basic insight into stem cell biology or make progress toward therapeutic objectives.
Collapse
Affiliation(s)
- Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
8
|
Garry MG, Kren S, Wenger JB, Garry DJ. Biologically Derived, Three-Dimensional, Embryonic Scaffolds for Long-Term Cardiomyocyte Culture. Stem Cells Dev 2021; 30:697-704. [PMID: 33843275 DOI: 10.1089/scd.2021.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The ability to maintain viable cultures of mature, primary cardiomyocytes is challenging. The lack of viable cardiomyocyte cultures severely limits in vitro biochemical assays, toxicology assays, drug screening assays, and other analyses. Here, we describe a novel three-dimensional (3D) embryonic scaffold, which supports the culture of postnatal day 7 murine cardiomyocytes within the embryonic heart for, at least, 28 days. We have observed that these cardiomyocytes display normal differentiation, protein expression, and function after extended culture. This novel culture system will allow for prolonged treatment of cardiomyocytes in a natural 3D orientation and has the potential for providing a superior tool for the screening of therapeutic compounds.
Collapse
Affiliation(s)
- Mary G Garry
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine (IEM), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stefan Kren
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph B Wenger
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel J Garry
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine (IEM), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
9
|
Sadeghi A, Khordad E, Ebrahimi V, Raoofi A, Alipour F, Ebrahimzadeh-Bideskan A. Neuroprotective effects of vitamin C and garlic on glycoconjugates changes of cerebellar cortex in lead-exposed rat offspring. J Chem Neuroanat 2021; 114:101948. [PMID: 33785394 DOI: 10.1016/j.jchemneu.2021.101948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022]
Abstract
The deteriorating effects of Lead (Pb) on central nervous system (CNS) such as cerebellum has been demonstrated in previous studies. Glycoconjugates with the important role in CNS development may be affected by Pb-exposure. Utilization of antioxidant agents and herbal plants has attracted a great deal of attention on attenuating neurotoxicants-induced damage. Thus, in this study the neuroprotective effects of vitamin C and garlic on content of glycoconjugates of cerebellar cortex in Pb-exposed animals were investigated. Wistar pregnant rats were divided into: control (C), Pb-exposed (Pb) (1500 ppm lead acetate in drinking water), Pb plus vitamin C (Pb + Vit C) (500 mg/kg) intraperitoneally, Pb plus garlic (Pb + G) (1 mL /100 g body weight fresh garlic juice via gavage), Pb plus vitamin C and garlic (Pb + Vit C + G), and sham groups (Sh). Finally, levels of Pb in blood were measured in both rats and offspring on postnatal day 50 (PND50). Also, the cerebellums were removed for measuring Pb-levels and performing lectin histochemistry. Blood and cerebellar Pb-levels were increased in Pb-exposed group compared to control group (P < 0.001), whereas they were decreased significantly in Pb + Vit C, Pb + G, and Pb + Vit C + G groups (P < 0.01). By using MPA, UEA-1, and WGA lectin histochemistry, Pb-exposed group showed weak staining intensity compared to other groups. Besides, significant decrease was observed in the density of lectin-positive neurons of Pb-exposed group compared to the control group (P < 0.001). Moreover, strong staining intensity and high lectin-positive neurons were found in Pb + Vit C, Pb + G and Pb + Vit C + G groups than Pb-exposed group (P < 0.001). The present study revealed that Pb-exposure can result in alteration in the cerebellar glycoconjugates contents and co-administration of vitamin C and garlic could attenuate the adverse effects of Pb. The findings of this study revealed the ameliorating effects of vitamin C and garlic against Pb, suggesting the potential use of vitamin C and garlic as preventive agents in Pb poisoning.
Collapse
Affiliation(s)
- Akram Sadeghi
- Institute of Neuroscience and Medicine (INM-10), Research Centre Jülich GmbH, Germany
| | - Elnaz Khordad
- Department of Physiology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Vahid Ebrahimi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Raoofi
- Leishmaniasis Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran; Department of Anatomy, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Fatemeh Alipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Development of Decellularized Oviductal Hydrogels as a Support for Rabbit Embryo Culture. Reprod Sci 2021; 28:1644-1658. [PMID: 33511539 DOI: 10.1007/s43032-020-00446-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/20/2020] [Indexed: 02/08/2023]
Abstract
The oviducts (fallopian tubes in mammals) function as the site of fertilization and provide necessary support for early embryonic development, mainly via embryonic exposure to the tubal microenvironment. The main objective of this study was to create an oviduct-specific extracellular matrix (oviECM) hydrogel rich in bioactive components that mimics the native environment, thus optimizing the developmental trajectories of cultured embryos. Rabbit oviducts were decellularized through SDS treatment and enzymatic digestion, and the acellular tissue was converted into oviductal pre-gel extracellular matrix (ECM) solutions. Incubation of these solutions at 37 °C resulted in stable hydrogels with a fibrous structure based on scanning electron microscopy. Histological staining, DNA quantification and colorimetric assays confirmed that the decellularized tissue and hydrogels contained no cellular or nuclear components but retained important components of the ECM, e.g. hyaluronic acid, glycoproteins and collagens. To evaluate the ability of oviECM hydrogels to maintain early embryonic development, two-cell rabbit embryos were cultured on oviECM-coated surfaces and compared to those cultured with standard techniques. Embryo development was similar in both conditions, with 95.9% and 98% of the embryos reaching the late morula/early blastocyst stage by 48 h under standard culture and oviECM conditions, respectively. Metabolomic analysis of culture media in the presence or absence of embryos, however, revealed that the oviECM coating may include signalling molecules and release compounds beneficial to embryo metabolism.
Collapse
|
11
|
Green DW, Watson JA, Watson GS, Stamboulis A. Sequenced Somatic Cell Reprogramming and Differentiation Inside Nested Hydrogel Droplets. ACTA ACUST UNITED AC 2020; 4:e2000071. [PMID: 32597033 DOI: 10.1002/adbi.202000071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/06/2020] [Indexed: 11/08/2022]
Abstract
The efficient genesis of pluripotent cells or therapeutic cells for regenerative medicine involves several external manipulations and conditioning protocols, which drives down clinical applicability. Automated programming of the genesis by microscale physical forces and chronological biochemistry can increase clinical success. The design and fabrication of nested polysaccharide droplets (millimeter-sized) with cell sustaining properties of natural tissues and intrinsic properties for time and space evolution of cell transformation signals between somatic cells, pluripotent cells and differentiated therapeutic cells in a swift and efficient manner without the need for laborious external manipulation are reported. Cells transform between phenotypic states by having single and double nested droplets constituted with extracellular matrix proteins and reprogramming, and differentiation factors infused chronologically across the droplet space. The cell transformation into germ layer cells and bone cells is successfully tested in vitro and in vivo and promotes the formation of new bone tissues. Thus, nested droplets with BMP-2 loaded guests synthesize mineralized bone tissue plates along the length of a cranial non-union bone defect at 4 weeks. The advantages of sequenced somatic cell reprogramming and differentiation inside an individual hydrogel module without external manipulation, promoted by formulating tissue mimetic physical, mechanical, and chemical microenvironments are shown.
Collapse
Affiliation(s)
- David W Green
- School of Metallurgy and Materials, Healthcare Technologies Institute, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jolanta A Watson
- School of Science and Engineering, University of the Sunshine Coast, Fraser Coast, Hervey Bay, QLD, 4655, Australia
| | - Gregory S Watson
- School of Science and Engineering, University of the Sunshine Coast, Fraser Coast, Hervey Bay, QLD, 4655, Australia
| | - Artemis Stamboulis
- School of Metallurgy and Materials, Healthcare Technologies Institute, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
12
|
Walter T, Gruenewald A, Detsch R, Boccaccini AR, Vogel N. Cell Interactions with Size-Controlled Colloidal Monolayers: Toward Improved Coatings in Bone Tissue Engineering. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:1793-1803. [PMID: 32017853 DOI: 10.1021/acs.langmuir.9b03308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The surface structure of biomaterials is of key importance to control its interactions with biological environments. Industrial fabrication and coating processes often introduce particulate nanostructures at implant surfaces. Understanding the cellular interaction with particle-based surface topologies and feature sizes in the colloidal length scale therefore offers the possibility to improve the biological response of synthetic biomaterials. Here, surfaces with controlled topography and regular feature sizes covering the relevant length scale of particulate coatings (100-1000 nm) are fabricated by colloidal templating. Using fluorescent microscopy, WST assay, and morphology analysis, results show that adhesion and attachment of bone-marrow derived murine stromal cells (ST2) are strongly influenced by the surface feature size while geometric details play an insignificant role. Quantitative analysis shows enhanced cell adhesion, spreading, viability, and activity when surface feature size decreases below 200 nm compared to flat surfaces, while larger feature sizes are detrimental to cell adhesion. Kinetic studies reveal that most cells on surfaces with larger features lose contact with the substrate over time. This study identifies colloidal templating as a simple method for creating highly defined model systems to investigate complex cell functions and provides design criteria for the choice of particulate coatings on commercial implant materials.
Collapse
Affiliation(s)
- Teresa Walter
- Institute of Particle Technology , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 4 , 91058 Erlangen , Germany
| | - Alina Gruenewald
- Institute of Biomaterials , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 6 , 91058 Erlangen , Germany
| | - Rainer Detsch
- Institute of Biomaterials , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 6 , 91058 Erlangen , Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 6 , 91058 Erlangen , Germany
| | - Nicolas Vogel
- Institute of Particle Technology , Friedrich-Alexander University Erlangen-Nürnberg , Cauerstrasse 4 , 91058 Erlangen , Germany
| |
Collapse
|
13
|
Gu Z, Guo J, Wang H, Wen Y, Gu Q. Bioengineered microenvironment to culture early embryos. Cell Prolif 2020; 53:e12754. [PMID: 31916359 PMCID: PMC7046478 DOI: 10.1111/cpr.12754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
The abnormalities of early post-implantation embryos can lead to early pregnancy loss and many other syndromes. However, it is hard to study embryos after implantation due to the limited accessibility. The success of embryo culture in vitro can avoid the challenges of embryonic development in vivo and provide a powerful research platform for research in developmental biology. The biophysical and chemical cues of the microenvironments impart significant spatiotemporal effects on embryonic development. Here, we summarize the main strategies which enable researchers to grow embryos outside of the body while overcoming the implantation barrier, highlight the roles of engineered microenvironments in regulating early embryonic development, and finally discuss the future challenges and new insights of early embryo culture.
Collapse
Affiliation(s)
- Zhen Gu
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijingChina
- CAS Key Laboratory of Bio‐inspired Materials and Interfacial ScienceTechnical Institute of Physics and ChemistryChinese Academy of SciencesBeijingChina
| | - Jia Guo
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
| | - Yongqiang Wen
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijingChina
| | - Qi Gu
- State Key Laboratory of Membrane BiologyInstitute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
14
|
Liang S, Zhou H, Yin N, Lu Y, Faiola F. Embryoid body-based RNA-seq analyses reveal a potential TBBPA multifaceted developmental toxicity. JOURNAL OF HAZARDOUS MATERIALS 2019; 376:223-232. [PMID: 31129320 DOI: 10.1016/j.jhazmat.2019.05.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 06/09/2023]
Abstract
The frequent detection of tetrabromobisphenol A (TBBPA) in the human body, especially in umbilical cord serum and breast milk, has raised concerns about TBBPA potential effects on embryonic development. The differentiation of embryonic stem cells (ESCs) in vitro can serve as a model for the early stages of embryonic development. In this study, we differentiated mouse ESCs via 3D aggregates called embryoid bodies in presence of environment and human relevant TBPPA concentrations for 28 days. We collected samples at different time points and analyzed TBBPA-dependent global gene expression changes by RNA-seq. Our analyses revealed a potential TBBPA multifaceted developmental toxicity with effects on the nervous and cardiac/skeletal muscle systems. Mechanistically, our findings suggest TBBPA endocrine disrupting activities in part via prolactin signaling.
Collapse
Affiliation(s)
- Shaojun Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hui Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanping Lu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Kumari S, Vermeulen S, van der Veer B, Carlier A, de Boer J, Subramanyam D. Shaping Cell Fate: Influence of Topographical Substratum Properties on Embryonic Stem Cells. TISSUE ENGINEERING. PART B, REVIEWS 2018; 24:255-266. [PMID: 29455619 PMCID: PMC7116060 DOI: 10.1089/ten.teb.2017.0468] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Development of multicellular organisms is a highly orchestrated process, with cells responding to factors and features present in the extracellular milieu. Changes in the surrounding environment help decide the fate of cells at various stages of development. This review highlights recent research that details the effects of mechanical properties of the surrounding environment and extracellular matrix and the underlying molecular mechanisms that regulate the behavior of embryonic stem cells (ESCs). In this study, we review the role of mechanical properties during embryogenesis and discuss the effect of engineered microtopographies on ESC pluripotency.
Collapse
Affiliation(s)
- Sarita Kumari
- National Center for Cell Science, SP Pune University, Pune, India
| | - Steven Vermeulen
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Ben van der Veer
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Aurélie Carlier
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | - Jan de Boer
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, Maastricht, The Netherlands
| | | |
Collapse
|
16
|
Rico-Varela J, Ho D, Wan LQ. In Vitro Microscale Models for Embryogenesis. ADVANCED BIOSYSTEMS 2018; 2:1700235. [PMID: 30533517 PMCID: PMC6286056 DOI: 10.1002/adbi.201700235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Indexed: 12/15/2022]
Abstract
Embryogenesis is a highly regulated developmental process requiring complex mechanical and biochemical microenvironments to give rise to a fully developed and functional embryo. Significant efforts have been taken to recapitulate specific features of embryogenesis by presenting the cells with developmentally relevant signals. The outcomes, however, are limited partly due to the complexity of this biological process. Microtechnologies such as micropatterned and microfluidic systems, along with new emerging embryonic stem cell-based models, could potentially serve as powerful tools to study embryogenesis. The aim of this article is to review major studies involving the culturing of pluripotent stem cells using different geometrical patterns, microfluidic platforms, and embryo/embryoid body-on-a-chip modalities. Indeed, new research opportunities have emerged for establishing in vitro culture for studying human embryogenesis and for high-throughput pharmacological testing platforms and disease models to prevent defects in early stages of human development.
Collapse
Affiliation(s)
- Jennifer Rico-Varela
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| | - Dominic Ho
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| |
Collapse
|
17
|
Silva R, Singh R, Sarker B, Papageorgiou DG, Juhasz-Bortuzzo JA, Roether JA, Cicha I, Kaschta J, Schubert DW, Chrissafis K, Detsch R, Boccaccini AR. Hydrogel matrices based on elastin and alginate for tissue engineering applications. Int J Biol Macromol 2018; 114:614-625. [PMID: 29572141 DOI: 10.1016/j.ijbiomac.2018.03.091] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/21/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Hydrogels from natural polymers are widely used in tissue engineering due to their unique properties, especially when regarding the cell environment and their morphological similarity to the extracellular matrix (ECM) of native tissues. In this study, we describe the production and characterization of novel hybrid hydrogels composed of alginate blended with elastin from bovine neck ligament. The properties of elastin as a component of the native ECM were combined with the excellent chemical and mechanical stability as well as biocompatibility of alginate to produce two hybrid hydrogels geometries, namely 2D films obtained using sonication treatment and 3D microcapsules produced by pressure-driven extrusion. The resulting blend hydrogels were submitted to an extensive physico-chemical characterization. Furthermore, the biological compatibility of these materials was assessed using normal human dermal fibroblasts, indicating the suitability of this blend for soft tissue engineering.
Collapse
Affiliation(s)
- Raquel Silva
- Institute of Briomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany.
| | - Raminder Singh
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology and Nanomedicine, ENT Department, University Hospital Erlangen, 91054 Erlangen, Germany; Laboratory of Molecular Cardiology, Medical Clinic 2, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Bapi Sarker
- Institute of Briomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Dimitrios G Papageorgiou
- Solid State Physics Section, Physics Department, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; School of Materials and National Graphene Institute, University of Manchester, Oxford Road, M13 9PL Manchester, United Kingdom
| | - Judith A Juhasz-Bortuzzo
- Institute of Briomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Judith A Roether
- Institute for Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Iwona Cicha
- Cardiovascular Nanomedicine Unit, Section of Experimental Oncology and Nanomedicine, ENT Department, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Joachim Kaschta
- Institute for Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Dirk W Schubert
- Institute for Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Konstantinos Chrissafis
- Solid State Physics Section, Physics Department, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Rainer Detsch
- Institute of Briomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Briomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany.
| |
Collapse
|
18
|
Kehr N, Motealleh A. Nanocomposite (Janus) paper as 3D cell culture system. Colloids Surf B Biointerfaces 2017; 156:236-242. [DOI: 10.1016/j.colsurfb.2017.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 04/20/2017] [Accepted: 05/05/2017] [Indexed: 12/15/2022]
|
19
|
Kandasamy M, Roll L, Langenstroth D, Brüstle O, Faissner A. Glycoconjugates reveal diversity of human neural stem cells (hNSCs) derived from human induced pluripotent stem cells (hiPSCs). Cell Tissue Res 2017; 368:531-549. [DOI: 10.1007/s00441-017-2594-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
|
20
|
Tan G, Liu Y, Wu Y, Ouyang K, Zhou L, Yu P, Liao J, Ning C. Electrically Reversible Redox-Switchable Polydopamine Films for Regulating Cell Behavior. Electrochim Acta 2017. [DOI: 10.1016/j.electacta.2016.12.189] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
21
|
Chen J, Tanguay RL, Xiao Y, Haggard DE, Ge X, Jia Y, Zheng Y, Dong Q, Huang C, Lin K. TBBPA exposure during a sensitive developmental window produces neurobehavioral changes in larval zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 216:53-63. [PMID: 27239688 DOI: 10.1016/j.envpol.2016.05.059] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/05/2016] [Accepted: 05/21/2016] [Indexed: 06/05/2023]
Abstract
Tetrabromobisphenol A (TBBPA), one of the most widely used brominated flame retardants (BFRs), is a ubiquitous contaminant in the environment and in the human body. This study demonstrated that zebrafish embryos exposed to TBBPA during a sensitive window of 8-48 h post-fertilization (hpf) displayed morphological malformations and mortality. Zebrafish exposed exclusively between 48 and 96 hpf were phenotypically normal. TBBPA was efficiently absorbed and accumulated in zebrafish embryos, but was eliminated quickly when the exposure solution was removed. Larval behavior assays conducted at 120 hpf indicated that exposure to 5 μM TBBPA from 8 to 48 hpf produced larvae with significantly lower average activity and speed of movement in the normal condition than in those exposed from 48 to 96 hpf. Specifically, 8-48 hpf-exposed larvae spent significantly less time in both activity bursts and gross movements compared to control or 48-96 hpf exposed larvae. Consistent with the motor deficits, TBBPA induced apoptotic cell death, delayed cranial motor neuron development, inhibited primary motor neuron development and loosed muscle fiber during the early developmental stages. To further explore TBBPA-induced developmental and neurobehavioral toxicity, RNA-Seq analysis was used to identify early transcriptional changes following TBBPA exposure. In total, 1969 transcripts were significantly differentially expressed (P < 0.05, FDR < 0.05, 1.5-FC) upon TBBPA exposure. Functional and pathway analysis of the TBBPA transcriptional profile identified biological processes involved in nerve development, muscle filament sliding and contraction, and extracellular matrix disassembly and organization changed significantly. In addition, TBBPA also led to an elevation in the expression of genes encoding uridine diphosphate glucuronyl transferases (ugt), which could affect thyroxine (T4) metabolism and subsequently lead to neurobehavioral changes. In summary, TBBPA exposure during a narrow, sensitive developmental window perturbs various molecular pathways and results in neurobehavioral deficits in zebrafish.
Collapse
Affiliation(s)
- Jiangfei Chen
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, No.130, Mei Long Road, Shanghai 200237, China; Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Robert L Tanguay
- Environmental and Molecular Toxicology, The Sinnhuber Aquatic Research Laboratory and the Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97333, USA
| | - Yanyan Xiao
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Derik E Haggard
- Environmental and Molecular Toxicology, The Sinnhuber Aquatic Research Laboratory and the Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97333, USA
| | - Xiaoqing Ge
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yinhang Jia
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Yi Zheng
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Qiaoxiang Dong
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Changjiang Huang
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou 325035, China.
| | - Kuangfei Lin
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resources and Environmental Engineering, East China University of Science and Technology, No.130, Mei Long Road, Shanghai 200237, China.
| |
Collapse
|
22
|
Lim KT, Seonwoo H, Choi KS, Jin H, Jang KJ, Kim J, Kim JW, Kim SY, Choung PH, Chung JH. Pulsed-Electromagnetic-Field-Assisted Reduced Graphene Oxide Substrates for Multidifferentiation of Human Mesenchymal Stem Cells. Adv Healthc Mater 2016; 5:2069-79. [PMID: 27332788 DOI: 10.1002/adhm.201600429] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Indexed: 12/12/2022]
Abstract
Electromagnetic fields (EMFs) can modulate cell proliferation, DNA replication, wound healing, cytokine expression, and the differentiation of mesenchymal stem cells (MSCs). Graphene, a 2D crystal of sp(2) -hybridized carbon atoms, has entered the spotlight in cell and tissue engineering research. However, a combination of graphene and EMFs has never been applied in tissue engineering. This study combines reduced graphene oxide (RGO) and pulsed EMFs (PEMFs) on the osteogenesis and neurogenesis of MSCs. First, the chemical properties of RGO are measured. After evaluation, the RGO is adsorbed onto glass, and its morphological and electrical properties are investigated. Next, an in vitro study is conducted using human alveolar bone marrow stem cells (hABMSCs). Their cell viability, cell adhesion, and extracellular matrix (ECM) formation are increased by RGO and PEMFs. The combination of RGO and PEMFs enhances osteogenic differentiation. Together, RGO and PEMFs enhance the neurogenic and adipogenic differentiation of hABMSCs. Moreover, in a DNA microarray analysis, the combination of RGO and PEMFs synergically increases ECM formation, membrane proteins, and metabolism. The combination of RGO and PEMFs is expected to be an efficient platform for stem cell and tissue engineering.
Collapse
Affiliation(s)
- Ki-Taek Lim
- Department of Biosystems Engineering; College of Agricultural and Life Sciences; Kangwon National University; Chuncheon 200-701 Republic of Korea
| | - Hoon Seonwoo
- Department of Biosystems and Biomaterials Science and Engineering; Seoul National University; Seoul 151-742 Republic of Korea
| | - Kyung Soon Choi
- Advanced Nano-Surface Research Group; Korea Basic Science Institute; Daejeon 305-333 Republic of Korea
| | - Hexiu Jin
- School of Stomatology; Capital Medical University; Beijing 10050 China
| | - Kyung-Je Jang
- Department of Biosystems and Biomaterials Science and Engineering; Seoul National University; Seoul 151-742 Republic of Korea
| | - Jangho Kim
- Department of Rural and Biosystems Engineering; Chonnam National University; Gwangju 500-757 Republic of Korea
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering; Institute for Nanoscience and Engineering; University of Arkansas; Fayetteville AR 72701 USA
| | - Soo Young Kim
- School of Chemical Engineering and Materials Science; Chung-Ang University; Seoul 151-742 Republic of Korea
| | - Pill-Hoon Choung
- Department of Oral and Maxillofacial Surgery and Dental Research Institute; School of Dentistry; Seoul National University; Seoul 110-774 Republic of Korea
| | - Jong Hoon Chung
- Department of Biosystems and Biomaterials Science and Engineering; Seoul National University; Seoul 151-742 Republic of Korea
- Research Institute of Agriculture and Life Sciences; Seoul National University; Seoul 151-742 Republic of Korea
| |
Collapse
|
23
|
Loganathan R, Rongish BJ, Smith CM, Filla MB, Czirok A, Bénazéraf B, Little CD. Extracellular matrix motion and early morphogenesis. Development 2016; 143:2056-65. [PMID: 27302396 PMCID: PMC4920166 DOI: 10.1242/dev.127886] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For over a century, embryologists who studied cellular motion in early amniotes generally assumed that morphogenetic movement reflected migration relative to a static extracellular matrix (ECM) scaffold. However, as we discuss in this Review, recent investigations reveal that the ECM is also moving during morphogenesis. Time-lapse studies show how convective tissue displacement patterns, as visualized by ECM markers, contribute to morphogenesis and organogenesis. Computational image analysis distinguishes between cell-autonomous (active) displacements and convection caused by large-scale (composite) tissue movements. Modern quantification of large-scale 'total' cellular motion and the accompanying ECM motion in the embryo demonstrates that a dynamic ECM is required for generation of the emergent motion patterns that drive amniote morphogenesis.
Collapse
Affiliation(s)
- Rajprasad Loganathan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brenda J Rongish
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Christopher M Smith
- Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA
| | - Michael B Filla
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Andras Czirok
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA Department of Biological Physics, Eotvos University, Budapest 1117, Hungary
| | - Bertrand Bénazéraf
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch Graffenstaden 67400, France
| | - Charles D Little
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
24
|
Seda Kehr N, Riehemann K. Controlled Cell Growth and Cell Migration in Periodic Mesoporous Organosilica/Alginate Nanocomposite Hydrogels. Adv Healthc Mater 2016; 5:193-7. [PMID: 26648333 DOI: 10.1002/adhm.201500638] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Indexed: 12/11/2022]
Abstract
Nanocomposite (NC) hydrogels with different periodic mesoporous organosilica (PMO) concentrations and a NC hydrogel bilayer with various PMO concentrations inside the layers of the hydrogel matrix are prepared. The effect of the PMO concentration on cell growth and migration of cells is reported. The cells migrate in the bilayer NC hydrogel towards higher PMO concentrations and from cell culture plates to NC hydrogel scaffolds.
Collapse
Affiliation(s)
- Nermin Seda Kehr
- Physikalisches Institut and CeNTech; Westfälische Wilhelms-Universität Münster; Heisenbergstraße 11 D-48149 Münster Germany
| | - Kristina Riehemann
- Physikalisches Institut and CeNTech; Westfälische Wilhelms-Universität Münster; Heisenbergstraße 11 D-48149 Münster Germany
| |
Collapse
|
25
|
Li Y, Kilian KA. Bridging the Gap: From 2D Cell Culture to 3D Microengineered Extracellular Matrices. Adv Healthc Mater 2015; 4:2780-96. [PMID: 26592366 PMCID: PMC4780579 DOI: 10.1002/adhm.201500427] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/05/2015] [Indexed: 12/20/2022]
Abstract
Historically the culture of mammalian cells in the laboratory has been performed on planar substrates with media cocktails that are optimized to maintain phenotype. However, it is becoming increasingly clear that much of biology discerned from 2D studies does not translate well to the 3D microenvironment. Over the last several decades, 2D and 3D microengineering approaches have been developed that better recapitulate the complex architecture and properties of in vivo tissue. Inspired by the infrastructure of the microelectronics industry, lithographic patterning approaches have taken center stage because of the ease in which cell-sized features can be engineered on surfaces and within a broad range of biocompatible materials. Patterning and templating techniques enable precise control over extracellular matrix properties including: composition, mechanics, geometry, cell-cell contact, and diffusion. In this review article we explore how the field of engineered extracellular matrices has evolved with the development of new hydrogel chemistry and the maturation of micro- and nano- fabrication. Guided by the spatiotemporal regulation of cell state in developing tissues, techniques for micropatterning in 2D, pseudo-3D systems, and patterning within 3D hydrogels will be discussed in the context of translating the information gained from 2D systems to synthetic engineered 3D tissues.
Collapse
Affiliation(s)
- Yanfen Li
- Department of Materials Science and Engineering, Department of Bioengineering, Institute for Genomic Biology, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana IL, 61801
| | - Kristopher A. Kilian
- Department of Materials Science and Engineering, Department of Bioengineering, Institute for Genomic Biology, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana IL, 61801
| |
Collapse
|
26
|
Rasmussen CH, Petersen DR, Moeller JB, Hansson M, Dufva M. Collagen Type I Improves the Differentiation of Human Embryonic Stem Cells towards Definitive Endoderm. PLoS One 2015; 10:e0145389. [PMID: 26713616 PMCID: PMC4694921 DOI: 10.1371/journal.pone.0145389] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 12/03/2015] [Indexed: 12/16/2022] Open
Abstract
Human embryonic stem cells have the ability to generate all cell types in the body and can potentially provide an unlimited source of cells for cell replacement therapy to treat degenerative diseases such as diabetes. Current differentiation protocols of human embryonic stem cells towards insulin producing beta cells focus on soluble molecules whereas the impact of cell-matrix interactions has been mainly unattended. In this study almost 500 different extracellular matrix protein combinations were screened to systemically identify extracellular matrix proteins that influence differentiation of human embryonic stem cells to the definitive endoderm lineage. The percentage of definitive endoderm cells after differentiation on collagen I and fibronectin was >85% and 65%, respectively. The cells on collagen I substrates displayed different morphology and gene expression during differentiation as assessed by time lapse studies compared to cells on the other tested substrates. Global gene expression analysis showed that cells differentiated on collagen I were largely similar to cells on fibronectin after completed differentiation. Collectively, the data suggest that collagen I induces a more rapid and consistent differentiation of stem cells to definitive endoderm. The results shed light on the importance of extracellular matrix proteins for differentiation and also points to a cost effective and easy method to improve differentiation.
Collapse
Affiliation(s)
| | | | | | | | - Martin Dufva
- DTU Nanotech, Technical University of Denmark, Kgs. Lyngby, Denmark
- * E-mail: (MH); (MD)
| |
Collapse
|
27
|
Costa AM, Mano JF. Extremely strong and tough hydrogels as prospective candidates for tissue repair – A review. Eur Polym J 2015. [DOI: 10.1016/j.eurpolymj.2015.07.053] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
28
|
Kehr NS, Atay S, Ergün B. Self-assembled Monolayers and Nanocomposite Hydrogels of Functional Nanomaterials for Tissue Engineering Applications. Macromol Biosci 2014; 15:445-63. [DOI: 10.1002/mabi.201400363] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nermin Seda Kehr
- Physikalisches Institut and Center for Nanotechnology; Westfälische Wilhelms-Universität Münster; Heisenbergstrasse 11 D-48149 Münster Germany
| | - Seda Atay
- Department of Nanotechnology and Nanomedicine; Hacettepe University; 06800 Ankara Turkey
| | - Bahar Ergün
- Department of Chemistry; Biochemistry Division; Hacettepe University; 06800 Ankara Turkey
| |
Collapse
|
29
|
Wei J, Han J, Zhao Y, Cui Y, Wang B, Xiao Z, Chen B, Dai J. The importance of three-dimensional scaffold structure on stemness maintenance of mouse embryonic stem cells. Biomaterials 2014; 35:7724-33. [PMID: 24930853 DOI: 10.1016/j.biomaterials.2014.05.060] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 05/21/2014] [Indexed: 12/26/2022]
Abstract
Revealing the mechanisms of cell fate regulation is important for scientific research and stem cell-based therapy. The traditional two-dimensional (2D) cultured mES cells are in a very different 2D niche from the in vivo equivalent-inner cell mass (ICM). Because the cell fate decision could be regulated by many cues which could be impacted by geometry, the traditional 2D culture system would hamper us from understanding the in vivo situations correctly. Three-dimensional (3D) scaffold was believed to provide a 3D environment closed to the in vivo one. In this work, three different scaffolds were prepared for cell culture. Several characters of mES cells were changed under 3D scaffolds culture compared to 2D, and these changes were mainly due to the alteration in geometry but not the matrix. The self-renewal of mES cells was promoted by the introducing of dimensionality. The stemness maintenance of mES was supported by all three 3D scaffolds without feeder cells in the long-time culture. Our findings demonstrated that the stemness maintenance of mES cells was promoted by the 3D geometry of scaffolds and this would provide a promising platform for ES cell research.
Collapse
Affiliation(s)
- Jianshu Wei
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Graduate School, ChineseAcademy of Sciences, Beijing 100190, China
| | - Jin Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Cui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bin Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
30
|
Faulk DM, Johnson SA, Zhang L, Badylak SF. Role of the Extracellular Matrix in Whole Organ Engineering. J Cell Physiol 2014; 229:984-9. [DOI: 10.1002/jcp.24532] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 01/07/2023]
Affiliation(s)
- Denver M. Faulk
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Bioengineering; University of Pittsburgh; Pittsburgh Pennsylvania
| | - Scott A. Johnson
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania
| | - Li Zhang
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Bioengineering; University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh; Pittsburgh Pennsylvania
| |
Collapse
|
31
|
Han YL, Wang S, Zhang X, Li Y, Huang G, Qi H, Pingguan-Murphy B, Li Y, Lu TJ, Xu F. Engineering physical microenvironment for stem cell based regenerative medicine. Drug Discov Today 2014; 19:763-73. [PMID: 24508818 DOI: 10.1016/j.drudis.2014.01.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/20/2014] [Accepted: 01/27/2014] [Indexed: 12/13/2022]
Abstract
Regenerative medicine has rapidly evolved over the past decade owing to its potential applications to improve human health. Targeted differentiations of stem cells promise to regenerate a variety of tissues and/or organs despite significant challenges. Recent studies have demonstrated the vital role of the physical microenvironment in regulating stem cell fate and improving differentiation efficiency. In this review, we summarize the main physical cues that are crucial for controlling stem cell differentiation. Recent advances in the technologies for the construction of physical microenvironment and their implications in controlling stem cell fate are also highlighted.
Collapse
Affiliation(s)
- Yu Long Han
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Shuqi Wang
- Brigham Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaohui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Hao Qi
- Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and training Center, Beijing, 100094, China
| | - Tian Jian Lu
- Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, 710049, China; Bioinspired Engineering & Biomechanics Center, Xi'an Jiaotong University, Shaanxi, 710049, China.
| |
Collapse
|
32
|
Zhou C, Qian SS, Li XJ, Yao F, Forsythe JS, Fu GD. Synthesis and characterization of well-defined PAA–PEG multi-responsive hydrogels by ATRP and click chemistry. RSC Adv 2014. [DOI: 10.1039/c4ra09438g] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Well-defined multi-responsive PAA–PEG hydrogels exhibit a unique swelling property at different pH and Ca2+ secondary crosslinking, and can potentially be used as stimuli responsive biomaterials.
Collapse
Affiliation(s)
- Chao Zhou
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing, China
| | - Shan-shan Qian
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing, China
| | - Xia-jun Li
- School of Public Health
- Southeast University
- Nanjing, China
| | - Fang Yao
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing, China
| | - John S. Forsythe
- Department of Materials Engineering
- Monash University
- Clayton, Australia
| | - Guo-dong Fu
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing, China
| |
Collapse
|
33
|
Sharma B, Ramus MD, Kirkwood CT, Sperry EE, Chu PH, Kao WW, Albig AR. Lumican exhibits anti-angiogenic activity in a context specific manner. CANCER MICROENVIRONMENT 2013; 6:263-71. [PMID: 23775523 DOI: 10.1007/s12307-013-0134-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/04/2013] [Indexed: 12/20/2022]
Abstract
A series of overexpression studies have shown that lumican suppresses angiogenesis in tumors produced from pancreatic adenocarcinoma, fibrosarcoma, and melanoma tumor cells. Despite lumican's anti-angiogenic activity, a clear correlation of differential expression of lumican in various cancers and cancer malignancy has failed to emerge. Therefore, we hypothesized that either 1.) endogenously expressed lumican is not anti-angiogenic or alternatively that 2.) lumican exhibits angiostatic activity only in limited microenvironments. Previously, lumican was shown to suppress tumor growth and angiogenesis in subcutaneously injected PanO2 pancreatic adenocarcinoma cells. Therefore, to determine if endogenously expressed lumican is anti-angiogenic we subcutaneously injected PanO2 cells into wild-type and lumican knockout mice and compared tumor growth and vascular densities of the resulting tumors. We found that tumors grown in lumican knockout animals were larger and contained significantly elevated vascular densities compared to those grown in wild-type mice. Interestingly however lumican knockout animals did not exhibit enhanced angiogenesis in aortic ring assays, matrigel plugs, or healing wound biopsies raising the possibility that lumican suppresses angiogenesis only in tumor microenvironments. To test this possibility, we sought a tumor model wherein lumican did not exhibit anti-angiogenic activity. Utilizing the 4T1 breast cancer model, we found that lumican suppressed 4T1 tumor growth and lung metastasis, but not angiogenesis. In conclusion, these results show that the angiostatic activity of lumican is dependent on currently undefined microenvironmental cues and therefore helps to understand why differential expression of lumican does not consistently correlate with human tumor malignancy.
Collapse
Affiliation(s)
- Bikram Sharma
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Suh HN, Han HJ. Fibronectin-induced VEGF receptor and calcium channel transactivation stimulate GLUT-1 synthesis and trafficking through PPARγ and TC10 in mouse embryonic stem cells. Stem Cell Res 2013; 10:371-86. [DOI: 10.1016/j.scr.2013.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 01/21/2013] [Accepted: 01/24/2013] [Indexed: 01/24/2023] Open
|
35
|
Prewitz M, Seib FP, Pompe T, Werner C. Polymeric biomaterials for stem cell bioengineering. Macromol Rapid Commun 2012; 33:1420-31. [PMID: 22887752 DOI: 10.1002/marc.201200382] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Indexed: 12/16/2022]
Abstract
This review covers the application of polymeric materials in stem cell bioengineering. Main emphasis is directed towards current material design concepts that mimic distinct exogenous signals of the stem cell microenvironment. Progress within the field of stem cell-specific biomaterials will be discussed, focusing on pluripotent, hematopoietic, mesenchymal and neural stem cells. The future role of biomaterials will be outlined with possible applications for cell reprogramming and engineering cancer cell microenvironments.
Collapse
Affiliation(s)
- Marina Prewitz
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Hohe Straße 6, 01069 Dresden, Germany
| | | | | | | |
Collapse
|
36
|
Titushkin I, Sun S, Cho M. Structure and Biology of the Cellular Environment: The Extracellular Matrix. NANOTECHNOLOGY FOR BIOLOGY AND MEDICINE 2012. [DOI: 10.1007/978-0-387-31296-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
37
|
He Q, Ao Q, Gong Y, Zhang X. Preparation of chitosan films using different neutralizing solutions to improve endothelial cell compatibility. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2011; 22:2791-2802. [PMID: 22042456 DOI: 10.1007/s10856-011-4444-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 09/05/2011] [Indexed: 05/31/2023]
Abstract
The development of chitosan-based constructs for application in large-size defects or highly vascularized tissues is still a challenging issue. The poor endothelial cell compatibility of chitosan hinders the colonization of vascular endothelial cells in the chitosan-based constructs, and retards the establishment of a functional microvascular network following implantation. The aim of the present study is to prepare chitosan films with different neutralization methods to improve their endothelial cell compatibility. Chitosan salt films were neutralized with either sodium hydroxide (NaOH) aqueous solution, NaOH ethanol solution, or ethanol solution without NaOH. The physicochemical properties and endothelial cell compatibility of the chitosan films were investigated. Results indicated that neutralization with different solutions affected the surface chemistry, swelling ratio, crystalline conformation, nanotopography, and mechanical properties of the chitosan films. The NaOH ethanol solution-neutralized chitosan film (Chi-NaOH/EtOH film) displayed a nanofiber-dominant surface, while the NaOH aqueous solution-neutralized film (Chi-NaOH/H(2)O film) and the ethanol solution-neutralized film (Chi-EtOH film) displayed nanoparticle-dominant surfaces. Moreover, the Chi-NaOH/EtOH films exhibited a higher stiffness as compared to the Chi-NaOH/H(2)O and Chi-EtOH films. Endothelial cell compatibility of the chitosan films was evaluated with a human microvascular endothelial cell line, HMEC-1. Compared with the Chi-NaOH/H(2)O and Chi-EtOH films, HMECs cultured on the Chi-NaOH/EtOH films fully spread and exhibited significantly higher levels of adhesion and proliferation, with retention of the endothelial phenotype and function. Our findings suggest that the surface nanotopography and mechanical properties contribute to determining the endothelial cell compatibility of chitosan films. The nature of the neutralizing solutions can affect the physicochemical properties and endothelial cell compatibility of chitosan films. Therefore, selection of suitable neutralization methods is highly important for the application of chitosan in tissue engineering.
Collapse
Affiliation(s)
- Qing He
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Tsinghua University, Beijing, China
| | | | | | | |
Collapse
|
38
|
Kraehenbuehl TP, Langer R, Ferreira LS. Three-dimensional biomaterials for the study of human pluripotent stem cells. Nat Methods 2011; 8:731-6. [PMID: 21878920 DOI: 10.1038/nmeth.1671] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The self-renewal and differentiation of human pluripotent stem cells (hPSCs) have typically been studied in flat, two-dimensional (2D) environments. In this Perspective, we argue that 3D model systems may be needed in addition, as they mimic the natural 3D tissue organization more closely. We survey methods that have used 3D biomaterials for expansion of undifferentiated hPSCs, directed differentiation of hPSCs and transplantation of differentiated hPSCs in vivo.
Collapse
Affiliation(s)
- Thomas P Kraehenbuehl
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | |
Collapse
|
39
|
Ma X, Tang Z, Wang N, Zhao S, Wang R, Tan L, Mu Y, Li K. Identification of extracellular matrix and cell adhesion molecule genes associated with muscle development in pigs. DNA Cell Biol 2011; 30:469-79. [PMID: 21476946 DOI: 10.1089/dna.2011.1218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Extracellular matrix (ECM) and cell adhesion molecule (CAM) genes are involved in the regulation of skeletal muscle development; however, their roles in skeletal muscle development in pigs are still poorly understood. 65 days postcopulation (dpc) is a critical time point in pig development. Therefore, we analyzed expression of ECM and CAM genes in the longissimus dorsi muscles at 65 dpc from Landrace (lean-type: L65), Tongcheng (obese-type: T65), and Wuzhishan pigs (miniature-type: W65) using microarray technology. A total of 35 genes were differently expressed between the breeds, and of them, 18, 18, and 20 genes, were observed in the comparisons of L65 versus T65, L65 versus W65, and T65 versus W65 (L65/T65, L65/W65, and T65/W65), respectively. In L65/T65, differently expressed genes were widely distributed, whereas in L65/W65 and T65/W65, they mostly focused on the genes encoding CAMs and ECMs proteins. Moreover, the largest number of up-regulated genes involved in skeletal muscle development was detected in L65, a moderate number in W65, and the smallest number was in T65. Cluster analysis suggested that T65 showed a more similar expression pattern to L65 than W65. In addition, we validated that five genes from microarray data were more highly expressed in the prenatal as compared to postnatal periods in Landrace and Tongcheng pigs and showed a greater range of high-level expression during gestation in Landrace than Tongcheng pigs. Our data indicated that ECM and CAM genes are differently expressed among the three breeds, and more complicated molecular events involving CAMs and ECMs were observed in Wuzhishan pigs. This study advances our knowledge of the molecular basis of phenotypic variation and provides a helpful resource for the identification of candidate genes associated with meat production traits in pigs.
Collapse
Affiliation(s)
- Xishan Ma
- Department of Gene and Cell Engineering, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Freytes DO, Wan LQ, Vunjak-Novakovic G. Geometry and force control of cell function. J Cell Biochem 2010; 108:1047-58. [PMID: 19795385 DOI: 10.1002/jcb.22355] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue engineering is becoming increasingly ambitious in its efforts to create functional human tissues, and to provide stem cell scientists with culture systems of high biological fidelity. Novel engineering designs are being guided by biological principles, in an attempt to recapitulate more faithfully the complexities of native cellular milieu. Three-dimensional (3D) scaffolds are being designed to mimic native-like cell environments and thereby elicit native-like cell responses. Also, the traditional focus on molecular regulatory factors is shifting towards the combined application of molecular and physical factors. Finally, methods are becoming available for the coordinated presentation of molecular and physical factors in the form of controllable spatial and temporal gradients. Taken together, these recent developments enable the interrogation of cellular behavior within dynamic culture settings designed to mimic some aspects of native tissue development, disease, or regeneration. We discuss here these advanced cell culture environments, with emphasis on the derivation of design principles from the development (the biomimetic paradigm) and the geometry-force control of cell function (the biophysical regulation paradigm).
Collapse
Affiliation(s)
- Donald O Freytes
- Department of Biomedical Engineering, Columbia University, New York, New York 10032, USA
| | | | | |
Collapse
|
41
|
Guimarães A, Martins A, Pinho ED, Faria S, Reis RL, Neves NM. Solving cell infiltration limitations of electrospun nanofiber meshes for tissue engineering applications. Nanomedicine (Lond) 2010; 5:539-54. [DOI: 10.2217/nnm.10.31] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Utilize the dual composition strategy to increase the pore size and solve the low cell infiltration capacity on random nanofiber meshes, an intrinsic limitation of electrospun scaffolds for tissue engineering applications. Materials & methods: Polycaprolactone and poly(ethylene oxide) solutions were electrospun simultaneously to obtain a dual composition nanofiber mesh. Selective dissolution of the poly(ethylene oxide) nanofiber fraction was performed. The biologic performance of these enhanced pore size nanofibrous structures was assessed with human osteoblastic cells. Results: The electrospun nanofiber meshes, after the poly(ethylene oxide) dissolution, showed statistically significant larger pore sizes when compared with polycaprolactone nanofiber meshes with a similar polycaprolactone volume fraction. This was also confirmed by interferometric optical profilometry. Using scanning electron microscopy and laser scanning confocal microscopy, it was observed that osteoblastic cells could penetrate into the nanofibrous structure and migrate into the opposite and unseeded side of the mesh. Conclusion: An electrospun mesh was created with sufficient pore size to allow cell infiltration into its structure, thus resulting in a fully populated construct appropriate for 3D tissue engineering applications.
Collapse
Affiliation(s)
- Ana Guimarães
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, Department Polymer Engineering, University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, S. Cláudio do Barco, 4806–909 Caldas das Taipas, Guimarães, Portugal
- IBB – Institute for Biotechnology & Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - Albino Martins
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, Department Polymer Engineering, University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, S. Cláudio do Barco, 4806–909 Caldas das Taipas, Guimarães, Portugal
- IBB – Institute for Biotechnology & Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - Elisabete D Pinho
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, Department Polymer Engineering, University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, S. Cláudio do Barco, 4806–909 Caldas das Taipas, Guimarães, Portugal
- IBB – Institute for Biotechnology & Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - Susana Faria
- Research Centre Officina Mathematica, Department of Mathematics for Science & Technology, University of Minho, Campus de Azurém, 4800–058 Guimarães, Portugal
| | - Rui L Reis
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, Department Polymer Engineering, University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, S. Cláudio do Barco, 4806–909 Caldas das Taipas, Guimarães, Portugal
- IBB – Institute for Biotechnology & Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - Nuno M Neves
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, Department Polymer Engineering, University of Minho; Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, S. Cláudio do Barco, 4806–909 Caldas das Taipas, Guimarães, Portugal
- IBB – Institute for Biotechnology & Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| |
Collapse
|
42
|
Self-assembling peptide nanofiber scaffolds, platelet-rich plasma, and mesenchymal stem cells for injectable bone regeneration with tissue engineering. J Craniofac Surg 2010; 20:1523-30. [PMID: 19816290 DOI: 10.1097/scs.0b013e3181b09b7e] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The purpose of this study was to investigate a capability of PuraMatrix (PM), which is a self-assembling peptide nanomaterial, as a scaffold for bone regeneration in combination with dog mesenchymal stem cells (dMSCs) and/or platelet-rich plasma (PRP) using tissue engineering and regenerative technology. Initially, teeth were extracted from an adult hybrid dog's mandible region. After 4 weeks, bone defects were prepared on both sides of the mandible with a trephine bar. The following graft materials were implanted into these defects: (1) control (defect only), (2) PM, (3) PM/PRP, (4) PM/dMSCs, and (5) PM/dMSCs/PRP. From scanning electron microscope images, PM had a three-dimensional nanostructure, and dMSCs attached on the surface of PM. At 2, 4, and 8 weeks after implantation, each sample was collected from the graft area with a trephine bar and assessed by histologic and histomorphometric analyses. It was observed that the bone regenerated by PM/dMSCs/PRP was of excellent quality, and mature bone had been formed. Histometrically, at 8 weeks, newly formed bone areas comprised 12.39 +/- 1.29% (control), 25.28 +/- 3.92% (PM), 27.72 +/- 3.15% (PM/PRP), 50.07 +/- 3.97% (PM/dMSCs), and 58.43 +/- 5.06% (PM/dMSCs/PRP). The PM/dMSCs and PM/dMSCs/PRP groups showed a significant increase at all weeks compared with the control, PM, or PM/PRP (P < 0.05 at 2, 4, and 8 weeks, analysis of variance). These results showed that MSCs might keep their own potential and promote new bone regeneration in the three-dimensional structure by PM scaffolds. Taken together, it is suggested that PM might be useful as a scaffold of bone regeneration in cell therapy, and these results might lead to an effective treatment method for bone defects.
Collapse
|
43
|
Espira L, Czubryt MP. Emerging concepts in cardiac matrix biologyThis article is one of a selection of papers published in a special issue on Advances in Cardiovascular Research. Can J Physiol Pharmacol 2009; 87:996-1008. [DOI: 10.1139/y09-105] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cardiac extracellular matrix, far from being merely a static support structure for the heart, is now recognized to play central roles in cardiac development, morphology, and cell signaling. Recent studies have better shaped our understanding of the tremendous complexity of this active and dynamic network. By activating intracellular signal cascades, the matrix transduces myocardial physical forces into responses by myocytes and fibroblasts, affecting their function and behavior. In turn, cardiac fibroblasts and myocytes play active roles in remodeling the matrix. Coupled with the ability of the matrix to act as a dynamic reservoir for growth factors and cytokines, this interplay between the support structure and embedded cells has the potential to exert dramatic effects on cardiac structure and function. One of the clearest examples of this occurs when cell–matrix interactions are altered inappropriately, contributing to pathological fibrosis and heart failure. This review will examine some of the recent concepts that have emerged regarding exactly how the cardiac matrix mediates these effects, how our collective vision of the matrix has changed as a result, and the current state of attempts to pharmacologically treat fibrosis.
Collapse
Affiliation(s)
- Leon Espira
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Michael P. Czubryt
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
44
|
Milleret V, Simonet M, Bittermann AG, Neuenschwander P, Hall H. Cyto- and hemocompatibility of a biodegradable 3D-scaffold material designed for medical applications. J Biomed Mater Res B Appl Biomater 2009; 91:109-21. [DOI: 10.1002/jbm.b.31379] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
45
|
Zheng Z, Zhang L, Kong L, Wang A, Gong Y, Zhang X. The behavior of MC3T3-E1 cells on chitosan/poly-L-lysine composite films: Effect of nanotopography, surface chemistry, and wettability. J Biomed Mater Res A 2009; 89:453-65. [DOI: 10.1002/jbm.a.31979] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
46
|
Chuai M, Weijer CJ. Who moves whom during primitive streak formation in the chick embryo. HFSP JOURNAL 2009; 3:71-6. [PMID: 19794819 DOI: 10.2976/1.3103933] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Accepted: 03/03/2009] [Indexed: 11/19/2022]
Abstract
Gastrulation is a critical stage in the development of all vertebrates. During gastrulation mesendoderm cells move inside the embryo to form the gut, muscles, and skeleton. In amniotes the mesendoderm cells move inside the embryo through a structure known as the primitive streak, extending from the posterior pole anterior through the midline of the embryo. Primitive streak formation involves large scale cell flows of a layer of highly polarized epithelial epiblast cells. The epiblast is separated from a lower layer of hypoblast cells through a well developed basal lamina. Recent experiments in which in vivo extracellular matrix dynamics was followed via labeling with fibronectin specific fluorescent antibodies and time-lapse microscopy have suggested that extracellular matrix dynamics essentially coincides with the observed epiblast cell displacements (Zamir et al., 2008, PLoS Biol 6, e247). These observations raise the important question of who moves whom and where do cells derive traction. We discuss these matters and their implications for our understanding of the mechanisms underlying cell flows during primitive streak formation in the chick embryo.
Collapse
Affiliation(s)
- Manli Chuai
- Division of Cell and Developmental Biology, Wellcome Trust Biocentre, College of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | | |
Collapse
|
47
|
Abstract
Artificial ECMs that not only closely mimic the hybrid nature of the natural ECM but also provide tunable material properties and enhanced biological functions are attractive candidates for tissue engineering applications. This review summarizes recent advances in developing multicomponent hybrid hydrogels by integrating modular and heterogeneous building blocks into well-defined, multifunctional hydrogel composites. The individual building blocks can be chemically, morphologically, and functionally diverse, and the hybridization can occur at molecular level or microscopic scale. The modular nature of the designs, combined with the potential synergistic effects of the hybrid systems, has resulted in novel hydrogel matrices with robust structure and defined functions.
Collapse
Affiliation(s)
- Xinqiao Jia
- Department of Materials Science and Engineering, Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19716, USA.
| | | |
Collapse
|
48
|
Srouji S, Kizhner T, Suss-Tobi E, Livne E, Zussman E. 3-D Nanofibrous electrospun multilayered construct is an alternative ECM mimicking scaffold. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2008; 19:1249-55. [PMID: 17701297 DOI: 10.1007/s10856-007-3218-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Accepted: 01/22/2007] [Indexed: 05/16/2023]
Abstract
UNLABELLED Extra cellular matrix (ECM) is a natural cell environment, possesses complicated nano- and macro- architecture. Mimicking this three-dimensional (3-D) web is a challenge in the modern tissue engineering. This study examined the application of a novel 3-D construct, produced by multilayered organization of electrospun nanofiber membranes, for human bone marrow-derived mesenchymal stem cells (hMSCs) support. The hMSCs were seeded on an electrospun scaffold composed of poly epsilon-caproloactone (PCL) and collagen (COL) (1:1), and cultured in a dynamic flow bioreactor prior to in vivo implantation. Cell viability after seeding was analyzed by AlamarBlue Assay. At the various stages of experiment, cell morphology was examined by histology, scanning electron microscopy (SEM) and confocal microscopy. RESULTS A porous 3-D network of randomly oriented nanofibers appeared to support cell attachment in a way similar to traditionally used tissue culture polysterene plate. The following 6 week culture process of the tested construct in the dynamic flow system led to massive cell proliferation with even distribution inside the scaffold. Subcutaneous implantation of the cultured construct into nude mice demonstrated good integration with the surrounding tissues and neovascularization. CONCLUSION The combination of electrospinning technology with multilayer technique resulted in the novel 3-D nanofiber multilayered construct, able to contain efficient cell mass necessary for a successful in vivo grafting. The success of this approach with undifferentiated cells implies the possibility of its application as a platform for development of constructs with cells directed into various tissue types.
Collapse
Affiliation(s)
- S Srouji
- Anatomy and Cell Biology, Faculty of Medicine, Technion-I.I.T, Haifa 32000, Israel.
| | | | | | | | | |
Collapse
|
49
|
Martins A, Araújo JV, Reis RL, Neves NM. Electrospun nanostructured scaffolds for tissue engineering applications. Nanomedicine (Lond) 2007; 2:929-42. [DOI: 10.2217/17435889.2.6.929] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Despite being known for decades (since 1934), electrospinning has emerged recently as a very widespread technology to produce synthetic nanofibrous structures. These structures have morphologies and fiber diameters in a range comparable with those found in the extracellular matrix of human tissues. Therefore, nanofibrous scaffolds are intended to provide improved environments for cell attachment, migration, proliferation and differentiation when compared with traditional scaffolds. In addition, the process versatility and the highly specific surface area of nanofiber meshes may facilitate their use as local drug-release systems. Common electrospun nanofiber meshes are characterized by a random orientation. However, in some special cases, aligned distributions of the fibers can be obtained, with an interconnected microporous structure. The characteristic pore sizes and the inherent planar structure of the meshes can be detrimental for the desired cell infiltration into the inner regions, and eventually compromise tissue regeneration. Several strategies can be followed to overcome these limitations, and are discussed in detail here.
Collapse
Affiliation(s)
- Albino Martins
- 3B’s Research Group – Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, Campus de Gualtar, University of Minho, 4710–057 Braga, Portugal
- Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - José V Araújo
- 3B’s Research Group – Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, Campus de Gualtar, University of Minho, 4710–057 Braga, Portugal
- Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - Rui L Reis
- 3B’s Research Group – Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, Campus de Gualtar, University of Minho, 4710–057 Braga, Portugal
- Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| | - Nuno M Neves
- 3B’s Research Group – Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, Campus de Gualtar, University of Minho, 4710–057 Braga, Portugal
- Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Braga, Portugal
| |
Collapse
|
50
|
GOLDBERG MICHAEL, LANGER ROBERT, JIA XINQIAO. Nanostructured materials for applications in drug delivery and tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2007; 18:241-68. [PMID: 17471764 PMCID: PMC3017754 DOI: 10.1163/156856207779996931] [Citation(s) in RCA: 592] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Research in the areas of drug delivery and tissue engineering has witnessed tremendous progress in recent years due to their unlimited potential to improve human health. Meanwhile, the development of nanotechnology provides opportunities to characterize, manipulate and organize matter systematically at the nanometer scale. Biomaterials with nano-scale organizations have been used as controlled release reservoirs for drug delivery and artificial matrices for tissue engineering. Drug-delivery systems can be synthesized with controlled composition, shape, size and morphology. Their surface properties can be manipulated to increase solubility, immunocompatibility and cellular uptake. The limitations of current drug delivery systems include suboptimal bioavailability, limited effective targeting and potential cytotoxicity. Promising and versatile nano-scale drug-delivery systems include nanoparticles, nanocapsules, nanotubes, nanogels and dendrimers. They can be used to deliver both small-molecule drugs and various classes of biomacromolecules, such as peptides, proteins, plasmid DNA and synthetic oligodeoxynucleotides. Whereas traditional tissue-engineering scaffolds were based on hydrolytically degradable macroporous materials, current approaches emphasize the control over cell behaviors and tissue formation by nano-scale topography that closely mimics the natural extracellular matrix (ECM). The understanding that the natural ECM is a multifunctional nanocomposite motivated researchers to develop nanofibrous scaffolds through electrospinning or self-assembly. Nanocomposites containing nanocrystals have been shown to elicit active bone growth. Drug delivery and tissue engineering are closely related fields. In fact, tissue engineering can be viewed as a special case of drug delivery where the goal is to accomplish controlled delivery of mammalian cells. Controlled release of therapeutic factors in turn will enhance the efficacy of tissue engineering. From a materials point of view, both the drug-delivery vehicles and tissue-engineering scaffolds need to be biocompatible and biodegradable. The biological functions of encapsulated drugs and cells can be dramatically enhanced by designing biomaterials with controlled organizations at the nanometer scale. This review summarizes the most recent development in utilizing nanostructured materials for applications in drug delivery and tissue engineering.
Collapse
Affiliation(s)
- MICHAEL GOLDBERG
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, E25-342, Cambridge, MA 02139, USA
| | - ROBERT LANGER
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, E25-342, Cambridge, MA 02139, USA
| | - XINQIAO JIA
- Department of Materials Science and Engineering, 201 DuPont Hall, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|