1
|
Nurul Alam AMM, Kim CJ, Kim SH, Kumari S, Lee EY, Hwang YH, Joo ST. Scaffolding fundamentals and recent advances in sustainable scaffolding techniques for cultured meat development. Food Res Int 2024; 189:114549. [PMID: 38876607 DOI: 10.1016/j.foodres.2024.114549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/26/2024] [Accepted: 05/25/2024] [Indexed: 06/16/2024]
Abstract
In cultured meat (CM) production, Scaffolding plays an important role by aiding cell adhesion, growth, differentiation, and alignment. The existence of fibrous microstructure in connective and muscle tissues has attracted considerable interest in the realm of tissue engineering and triggered the interest of researchers to implement scaffolding techniques. A wide array of research efforts is ongoing in scaffolding technologies for achieving the real meat structure on the principality of biomedical research and to replace serum free CM production. Scaffolds made of animal-derived biomaterials are found efficient in replicating the extracellular matrix (ECM), thus focus should be paid to utilize animal byproducts for this purpose. Proper identification and utilization of plant-derived scaffolding biomaterial could be helpful to add diversified options in addition to animal derived sources and reduce in cost of CM production through scaffolds. Furthermore, techniques like electrospinning, modified electrospinning and 3D bioprinting should be focused on to create 3D porous scaffolds to mimic the ECM of the muscle tissue and form real meat-like structures. This review discusses recent advances in cutting edge scaffolding techniques and edible biomaterials related to structured CM production.
Collapse
Affiliation(s)
- A M M Nurul Alam
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea.
| | - Chan-Jin Kim
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea.
| | - So-Hee Kim
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea
| | - Swati Kumari
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea
| | - Eun-Yeong Lee
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea
| | - Young-Hwa Hwang
- Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52852, Republic of Korea.
| | - Seon-Tea Joo
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52852, Republic of Korea; Institute of Agriculture & Life Science, Gyeongsang National University, Jinju 52852, Republic of Korea.
| |
Collapse
|
2
|
Liu Y, Guo Q, Zhang X, Wang Y, Mo X, Wu T. Progress in Electrospun Fibers for Manipulating Cell Behaviors. ADVANCED FIBER MATERIALS 2023; 5:1241-1272. [DOI: 10.1007/s42765-023-00281-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/08/2023] [Indexed: 01/06/2025]
|
3
|
Buchmann S, Enrico A, Holzreuter MA, Reid M, Zeglio E, Niklaus F, Stemme G, Herland A. Probabilistic cell seeding and non-autofluorescent 3D-printed structures as scalable approach for multi-level co-culture modeling. Mater Today Bio 2023; 21:100706. [PMID: 37435551 PMCID: PMC10331311 DOI: 10.1016/j.mtbio.2023.100706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/13/2023] Open
Abstract
To model complex biological tissue in vitro, a specific layout for the position and numbers of each cell type is necessary. Establishing such a layout requires manual cell placement in three dimensions (3D) with micrometric precision, which is complicated and time-consuming. Moreover, 3D printed materials used in compartmentalized microfluidic models are opaque or autofluorescent, hindering parallel optical readout and forcing serial characterization methods, such as patch-clamp probing. To address these limitations, we introduce a multi-level co-culture model realized using a parallel cell seeding strategy of human neurons and astrocytes on 3D structures printed with a commercially available non-autofluorescent resin at micrometer resolution. Using a two-step strategy based on probabilistic cell seeding, we demonstrate a human neuronal monoculture that forms networks on the 3D printed structure and can establish cell-projection contacts with an astrocytic-neuronal co-culture seeded on the glass substrate. The transparent and non-autofluorescent printed platform allows fluorescence-based immunocytochemistry and calcium imaging. This approach provides facile multi-level compartmentalization of different cell types and routes for pre-designed cell projection contacts, instrumental in studying complex tissue, such as the human brain.
Collapse
Affiliation(s)
- Sebastian Buchmann
- Division of Nanobiotechnology, KTH Royal Institute of Technology, Tomtebodavägen 23a, 171 65, Solna, Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Alessandro Enrico
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
- Synthetic Physiology lab, Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100, Pavia, Italy
| | - Muriel Alexandra Holzreuter
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
| | - Michael Reid
- Department of Fiber and Polymer Technology, Wallenberg Wood Science Centre, KTH Royal Institute of Technology, Teknikringen 56-58, 100 44, Stockholm, Sweden
| | - Erica Zeglio
- Division of Nanobiotechnology, KTH Royal Institute of Technology, Tomtebodavägen 23a, 171 65, Solna, Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Frank Niklaus
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
| | - Göran Stemme
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
| | - Anna Herland
- Division of Nanobiotechnology, KTH Royal Institute of Technology, Tomtebodavägen 23a, 171 65, Solna, Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| |
Collapse
|
4
|
Chauhan A, Bhatt AD. A review on design of scaffold for osteoinduction: Toward the unification of independent design variables. Biomech Model Mechanobiol 2023; 22:1-21. [PMID: 36121530 DOI: 10.1007/s10237-022-01635-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
Biophysical stimulus quantifies the osteoinductivity of the scaffold concerning the mechanoregulatory mathematical models of scaffold-assisted cellular differentiation. Consider a set of independent structural variables ($) that comprises bulk porosity levels ([Formula: see text]) and a set of morphological features of the micro-structure ([Formula: see text]) associated with scaffolds, i.e., [Formula: see text]. The literature suggests that biophysical stimulus ([Formula: see text]) is a function of independent structural variables ($). Limited understanding of the functional correlation between biophysical stimulus and structural features results in the lack of the desired osteoinductivity in a scaffold. Consequently, it limits their broad applicability to assist bone tissue regeneration for treating critical-sized bone fractures. The literature indicates the existence of multi-dimensional independent design variable space as a probable reason for the general lack of osteoinductivity in scaffolds. For instance, known morphological features are the size, shape, orientation, continuity, and connectivity of the porous regions in the scaffold. It implies that the number of independent variables ([Formula: see text]) is more than two, i.e., [Formula: see text], which interact and influence the magnitude of [Formula: see text] in a unified manner. The efficiency of standard engineering design procedures to analyze the correlation between dependent variable ([Formula: see text]) and independent variables ($) in 3D mutually orthogonal Cartesian coordinate system diminishes proportionally with the increase in the number of independent variables ([Formula: see text]) (Deb in Optimization for engineering design-algorithms and examples, PHI Learning Private Limited, New Delhi, 2012). Therefore, there is an immediate need to devise a framework that has the potential to quantify the micro-structural's morphological features in a unified manner to increase the prospects of scaffold-assisted bone tissue regeneration.
Collapse
Affiliation(s)
- Atul Chauhan
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India.
| | - Amba D Bhatt
- Department of Mechanical Engineering, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| |
Collapse
|
5
|
Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects. Acta Biomater 2021; 132:129-148. [PMID: 33813090 DOI: 10.1016/j.actbio.2021.03.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem cells (HSCs) have proven their clinical relevance in stem cell transplantation to cure patients with hematological disorders. Key to their regenerative potential is their natural microenvironment - their niche - in the bone marrow (BM). Developments in the field of biomaterials enable the recreation of such environments with increasing preciseness in the laboratory. Such artificial niches help to gain a fundamental understanding of the biophysical and biochemical processes underlying the interaction of HSCs with the materials in their environment and the disturbance of this interplay during diseases affecting the BM. Artificial niches also have the potential to multiply HSCs in vitro, to enable the targeted differentiation of HSCs into mature blood cells or to serve as drug-testing platforms. In this review, we will introduce the importance of artificial niches followed by the biology and biophysics of the natural archetype. We will outline how 2D biomaterials can be used to dissect the complexity of the natural niche into individual parameters for fundamental research and how 3D systems evolved from them. We will present commonly used biomaterials for HSC research and their applications. Finally, we will highlight two areas in the field of HSC research, which just started to unlock the possibilities provided by novel biomaterials, in vitro blood production and studying the pathophysiology of the niche in vitro. With these contents, the review aims to give a broad overview of the different biomaterials applied for HSC research and to discuss their potentials, challenges and future directions in the field. STATEMENT OF SIGNIFICANCE: Hematopoietic stem cells (HSCs) are multipotent cells responsible for maintaining the turnover of all blood cells. They are routinely applied to treat patients with hematological diseases. This high clinical relevance explains the necessity of multiplication or differentiation of HSCs in the laboratory, which is hampered by the missing natural microenvironment - the so called niche. Biomaterials offer the possibility to mimic the niche and thus overcome this hurdle. The review introduces the HSC niche in the bone marrow and discusses the utility of biomaterials in creating artificial niches. It outlines how 2D systems evolved into sophisticated 3D platforms, which opened the gateway to applications such as, expansion of clinically relevant HSCs, in vitro blood production, studying niche pathologies and drug testing.
Collapse
|
6
|
Singh A, Banerjee SL, Dhiman V, Bhadada SK, Sarkar P, Khamrai M, Kumari K, Kundu PP. Fabrication of calcium hydroxyapatite incorporated polyurethane-graphene oxide nanocomposite porous scaffolds from poly (ethylene terephthalate) waste: A green route toward bone tissue engineering. POLYMER 2020. [DOI: 10.1016/j.polymer.2020.122436] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
7
|
Xing F, Li L, Zhou C, Long C, Wu L, Lei H, Kong Q, Fan Y, Xiang Z, Zhang X. Regulation and Directing Stem Cell Fate by Tissue Engineering Functional Microenvironments: Scaffold Physical and Chemical Cues. Stem Cells Int 2019; 2019:2180925. [PMID: 31949436 PMCID: PMC6948329 DOI: 10.1155/2019/2180925] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/05/2019] [Indexed: 02/05/2023] Open
Abstract
It is well known that stem cells reside within tissue engineering functional microenvironments that physically localize them and direct their stem cell fate. Recent efforts in the development of more complex and engineered scaffold technologies, together with new understanding of stem cell behavior in vitro, have provided a new impetus to study regulation and directing stem cell fate. A variety of tissue engineering technologies have been developed to regulate the fate of stem cells. Traditional methods to change the fate of stem cells are adding growth factors or some signaling pathways. In recent years, many studies have revealed that the geometrical microenvironment played an essential role in regulating the fate of stem cells, and the physical factors of scaffolds including mechanical properties, pore sizes, porosity, surface stiffness, three-dimensional structures, and mechanical stimulation may affect the fate of stem cells. Chemical factors such as cell-adhesive ligands and exogenous growth factors would also regulate the fate of stem cells. Understanding how these physical and chemical cues affect the fate of stem cells is essential for building more complex and controlled scaffolds for directing stem cell fate.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041 Sichuan, China
| | - Lang Li
- Department of Pediatric Surgery, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041 Sichuan, China
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, Sichuan, China
| | - Cheng Long
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041 Sichuan, China
| | - Lina Wu
- National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, Sichuan, China
| | - Haoyuan Lei
- National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, Sichuan, China
| | - Qingquan Kong
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041 Sichuan, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, Sichuan, China
| | - Zhou Xiang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu, 610041 Sichuan, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 610064 Chengdu, Sichuan, China
| |
Collapse
|
8
|
Constructing Three-Dimensional Microenvironments Using Engineered Biomaterials for Hematopoietic Stem Cell Expansion. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:312-329. [DOI: 10.1089/ten.teb.2018.0286] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
9
|
Dwarshuis NJ, Parratt K, Santiago-Miranda A, Roy K. Cells as advanced therapeutics: State-of-the-art, challenges, and opportunities in large scale biomanufacturing of high-quality cells for adoptive immunotherapies. Adv Drug Deliv Rev 2017. [PMID: 28625827 DOI: 10.1016/j.addr.2017.06.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Therapeutic cells hold tremendous promise in treating currently incurable, chronic diseases since they perform multiple, integrated, complex functions in vivo compared to traditional small-molecule drugs or biologics. However, they also pose significant challenges as therapeutic products because (a) their complex mechanisms of actions are difficult to understand and (b) low-cost bioprocesses for large-scale, reproducible manufacturing of cells have yet to be developed. Immunotherapies using T cells and dendritic cells (DCs) have already shown great promise in treating several types of cancers, and human mesenchymal stromal cells (hMSCs) are now extensively being evaluated in clinical trials as immune-modulatory cells. Despite these exciting developments, the full potential of cell-based therapeutics cannot be realized unless new engineering technologies enable cost-effective, consistent manufacturing of high-quality therapeutic cells at large-scale. Here we review cell-based immunotherapy concepts focused on the state-of-the-art in manufacturing processes including cell sourcing, isolation, expansion, modification, quality control (QC), and culture media requirements. We also offer insights into how current technologies could be significantly improved and augmented by new technologies, and how disciplines must converge to meet the long-term needs for large-scale production of cell-based immunotherapies.
Collapse
Affiliation(s)
- Nate J Dwarshuis
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30332-0313, United States; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Kirsten Parratt
- The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States; Department of Material Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Adriana Santiago-Miranda
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30332-0313, United States; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Atlanta, GA 30332-0313, United States; The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, United States.
| |
Collapse
|
10
|
Regmi S, Jeong JH. Superiority of three-dimensional stem cell clusters over monolayer culture: An archetype to biological application. Macromol Res 2016. [DOI: 10.1007/s13233-016-4107-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
11
|
Dehdilani N, Shamsasenjan K, Movassaghpour A, Akbarzadehlaleh P, Amoughli Tabrizi B, Parsa H, Sabagi F. Improved Survival and Hematopoietic Differentiation of Murine Embryonic Stem Cells on Electrospun Polycaprolactone Nanofiber. CELL JOURNAL 2016; 17:629-38. [PMID: 26862522 PMCID: PMC4746413 DOI: 10.22074/cellj.2016.3835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 11/13/2014] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Three-dimensional (3D) biomimetic nanofiber scaffolds have widespread ap- plications in biomedical tissue engineering. They provide a suitable environment for cel- lular adhesion, survival, proliferation and differentiation, guide new tissue formation and development, and are one of the outstanding goals of tissue engineering. Electrospinning has recently emerged as a leading technique for producing biomimetic scaffolds with mi- cro to nanoscale topography and a high porosity similar to the natural extracellular matrix (ECM). These scaffolds are comprised of synthetic and natural polymers for tissue engi- neering applications. Several kinds of cells such as human embryonic stem cells (hESCs) and mouse ESCs (mESCs) have been cultured and differentiated on nanofiber scaffolds. mESCs can be induced to differentiate into a particular cell lineage when cultured as em- bryoid bodies (EBs) on nano-sized scaffolds. MATERIALS AND METHODS We cultured mESCs (2500 cells/100 µl) in 96-well plates with knockout Dulbecco's modified eagle medium (DMEM-KO) and Roswell Park Memorial Institute-1640 (RPMI-1640), both supplemented with 20% ESC grade fetal bovine serum (FBS) and essential factors in the presence of leukemia inhibitory factor (LIF). mESCs were seeded at a density of 2500 cells/100 µl onto electrospun polycaprolactone (PCL) nanofibers in 96-well plates. The control group comprised mESCs grown on tissue cul- ture plates (TCP) at a density of 2500 cells/100 µl. Differentiation of mESCs into mouse hematopoietic stem cells (mHSCs) was performed by stem cell factor (SCF), interleukin-3 (IL-3), IL-6 and Fms-related tyrosine kinase ligand (Flt3-L) cytokines for both the PCL and TCP groups. We performed an experimental study of mESCs differentiation. RESULTS PCL was compared to conventional TCP for survival and differentiation of mESCs to mHSCs. There were significantly more mESCs in the PCL group. Flowcyto- metric analysis revealed differences in hematopoietic differentiation between the PCL and TCP culture systems. There were more CD34+(Sca1+) and CD133+cells subpopulations in the PCL group compared to the conventional TCP culture system. CONCLUSION The nanofiber scaffold, as an effective surface, improves survival and differentiation of mESCs into mHSCs compared to gelatin coated TCP. More studies are necessary to understand how the topographical features of electrospun fibers af- fect cell growth and behavior. This can be achieved by designing biomimetic scaffolds for tissue engineering.
Collapse
Affiliation(s)
- Nima Dehdilani
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Aliakbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahram Amoughli Tabrizi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Hamed Parsa
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sabagi
- Iran Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
12
|
Choi JS, Mahadik BP, Harley BAC. Engineering the hematopoietic stem cell niche: Frontiers in biomaterial science. Biotechnol J 2015; 10:1529-45. [PMID: 26356030 PMCID: PMC4724421 DOI: 10.1002/biot.201400758] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/15/2015] [Accepted: 07/16/2015] [Indexed: 12/13/2022]
Abstract
Hematopoietic stem cells (HSCs) play a crucial role in the generation of the body's blood and immune cells. This process takes place primarily in the bone marrow in specialized 'niche' microenvironments, which provide signals responsible for maintaining a balance between HSC quiescence, self-renewal, and lineage specification required for life-long hematopoiesis. While our understanding of these signaling mechanisms continues to improve, our ability to engineer them in vitro for the expansion of clinically relevant HSC populations is still lacking. In this review, we focus on development of biomaterials-based culture platforms for in vitro study of interactions between HSCs and their local microenvironment. The tools and techniques used for both examining HSC-niche interactions as well as applying these findings towards controlled HSC expansion or directed differentiation in 2D and 3D platforms are discussed. These novel techniques hold the potential to push the existing boundaries of HSC cultures towards high-throughput, real-time, and single-cell level biomimetic approaches that enable a more nuanced understanding of HSC regulation and function. Their application in conjunction with innovative biomaterial platforms can pave the way for engineering artificial bone marrow niches for clinical applications as well as elucidating the pathology of blood-related cancers and disorders.
Collapse
Affiliation(s)
- Ji Sun Choi
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bhushan P Mahadik
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
13
|
Tresoldi C, Pellegata AF, Mantero S. Cells and stimuli in small-caliber blood vessel tissue engineering. Regen Med 2015; 10:505-27. [DOI: 10.2217/rme.15.19] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The absence of successful solutions in treatments of small-caliber vessel diseases led to the Vascular Tissue Engineering approach to develop functional nonimmunogenic tissue engineered blood vessels. In this context, the choice of cells to be seeded and the microenvironment conditioning are pivotal. Biochemical and biomechanical stimuli seem to activate physiological regulatory pathways that induce the production of molecules and proteins stimulating stem cell differentiation toward vascular lineage and reproducing natural cross-talks among vascular cells to improve the maturation of tissue engineered blood vessels. Thus, this review focuses on (1) available cell sources, and (2) biochemical and biomechanical stimuli, with the final aim to obtain the long-term stability of the endothelium and mechanical properties suitable for withstanding physiological load.
Collapse
Affiliation(s)
- Claudia Tresoldi
- Department of Chemistry, Materials & Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Alessandro Filippo Pellegata
- Department of Chemistry, Materials & Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Sara Mantero
- Department of Chemistry, Materials & Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milan, Italy
| |
Collapse
|
14
|
Ruedinger F, Lavrentieva A, Blume C, Pepelanova I, Scheper T. Hydrogels for 3D mammalian cell culture: a starting guide for laboratory practice. Appl Microbiol Biotechnol 2014; 99:623-36. [DOI: 10.1007/s00253-014-6253-y] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 12/21/2022]
|
15
|
Alessandri M, Lizzo G, Gualandi C, Mangano C, Giuliani A, Focarete ML, Calzà L. Influence of biological matrix and artificial electrospun scaffolds on proliferation, differentiation and trophic factor synthesis of rat embryonic stem cells. Matrix Biol 2013; 33:68-76. [PMID: 23954537 DOI: 10.1016/j.matbio.2013.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/02/2013] [Accepted: 08/02/2013] [Indexed: 01/06/2023]
Abstract
Two-dimensional vs three-dimensional culture conditions, such as the presence of extracellular matrix components, could deeply influence the cell fate and properties. In this paper we investigated proliferation, differentiation, survival, apoptosis, growth and neurotrophic factor synthesis of rat embryonic stem cells (RESCs) cultured in 2D and 3D conditions generated using Cultrex® Basement Membrane Extract (BME) and in poly-(L-lactic acid) (PLLA) electrospun sub-micrometric fibres. It is demonstrated that, in the absence of other instructive stimuli, growth, differentiation and paracrine activity of RESCs are directly affected by the different microenvironment provided by the scaffold. In particular, RESCs grown on an electrospun PLLA scaffolds coated or not with BME have a higher proliferation rate, higher production of bioactive nerve growth factor (NGF) and vascular endothelial growth factor (VEGF) compared to standard 2D conditions, lasting for at least 2 weeks. Due to the high mechanical flexibility of PLLA electrospun scaffolds, the PLLA/stem cell culture system offers an interesting potential for implantable neural repair devices.
Collapse
Affiliation(s)
- M Alessandri
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.
| | - G Lizzo
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy.
| | - C Gualandi
- Department of Chemistry "G. Ciamician" and National Consortium of Materials Science and Technology (INSTM, Bologna RU), University of Bologna, Bologna, Italy; Advanced Applications in Mechanical Engineering and Materials Technology. Interdepartmental Center for Industrial Research, University of Bologna, Bologna, Italy.
| | - C Mangano
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy
| | - A Giuliani
- Department of Veterinary Medical Science, University of Bologna, Bologna, Italy.
| | - M L Focarete
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy; Department of Chemistry "G. Ciamician" and National Consortium of Materials Science and Technology (INSTM, Bologna RU), University of Bologna, Bologna, Italy.
| | - L Calzà
- Health Sciences and Technologies-Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy; Department of Veterinary Medical Science, University of Bologna, Bologna, Italy.
| |
Collapse
|
16
|
Hosseinkhani M, Shirazi R, Rajaei F, Mahmoudi M, Mohammadi N, Abbasi M. Engineering of the embryonic and adult stem cell niches. IRANIAN RED CRESCENT MEDICAL JOURNAL 2013; 15:83-92. [PMID: 23682319 PMCID: PMC3652509 DOI: 10.5812/ircmj.7541] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 01/08/2013] [Indexed: 12/15/2022]
Abstract
CONTEXT Stem cells have the potential to generate a renewable source of cells for regenerative medicine due to their ability to self-renew and differentiate to various functional cell types of the adult organism. The extracellular microenvironment plays a pivotal role in controlling stem cell fate responses. Therefore, identification of appropriate environmental stimuli that supports cellular proliferation and lineage-specific differentiation is critical for the clinical application of the stem cell therapies. EVIDENCE ACQUISITION Traditional methods for stem cells culture offer limited manipulation and control of the extracellular microenvironment. Micro engineering approaches are emerging as powerful tools to control stem cell-microenvironment interactions and for performing high-throughput stem cell experiments. RESULTS In this review, we provided an overview of the application of technologies such as surface micropatterning, microfluidics, and engineered biomaterials for directing stem cell behavior and determining the molecular cues that regulate cell fate decisions. CONCLUSIONS Stem cells have enormous potential for therapeutic and pharmaceutical applications, because they can give rise to various cell types. Despite their therapeutic potential, many challenges, including the lack of control of the stem cell microenvironment remain. Thus, a greater understanding of stem cell biology that can be used to expand and differentiate embryonic and adult stem cells in a directed manner offers great potential for tissue repair and regenerative medicine.
Collapse
Affiliation(s)
- Mohsen Hosseinkhani
- Department of Anatomy, Qazvin University of Medical Science, Qazvin, IR Iran
- Corresponding author: Mohsen Hosseinkhani, Department of Anatomy, Qazvin University of Medical Science, Qazvin, IR Iran. Tel: +98-2188274683, Fax: +98-2188274683, E-mail:
| | - Reza Shirazi
- Department of Anatomy, Qazvin University of Medical Science, Qazvin, IR Iran
| | - Farzad Rajaei
- Department of Anatomy, Qazvin University of Medical Science, Qazvin, IR Iran
| | - Masoud Mahmoudi
- Department of Anatomy, Qazvin University of Medical Science, Qazvin, IR Iran
| | - Navid Mohammadi
- Department of Community Medicine, Tehran University of Medical Science, Tehran, IR Iran
| | - Mahnaz Abbasi
- Department of Rheumatology, Qazvin University of Medical Science, Qazvin, IR Iran
| |
Collapse
|
17
|
Fernandez I, Fridley KM, Arasappan D, Ambler RV, Tucker PW, Roy K. Gene expression profile and functionality of ESC-derived Lin-ckit+Sca-1+ cells are distinct from Lin-ckit+Sca-1+ cells isolated from fetal liver or bone marrow. PLoS One 2012; 7:e51944. [PMID: 23300581 PMCID: PMC3531429 DOI: 10.1371/journal.pone.0051944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 11/13/2012] [Indexed: 01/30/2023] Open
Abstract
In vitro bioreactor-based cultures are being extensively investigated for large-scale production of differentiated cells from embryonic stem cells (ESCs). However, it is unclear whether in vitro ESC-derived progenitors have similar gene expression profiles and functionalities as their in vivo counterparts. This is crucial in establishing the validity of ESC-derived cells as replacements for adult-isolated cells for clinical therapies. In this study, we compared the gene expression profiles of Lin-ckit+Sca-1+ (LKS) cells generated in vitro from mouse ESCs using either static or bioreactor-based cultures, with that of native LKS cells isolated from mouse fetal liver (FL) or bone marrow (BM). We found that in vitro-generated LKS cells were more similar to FL- than to BM LKS cells in gene expression. Further, when compared to cells derived from bioreactor cultures, static culture-derived LKS cells showed fewer differentially expressed genes relative to both in vivo LKS populations. Overall, the expression of hematopoietic genes was lower in ESC-derived LKS cells compared to cells from BM and FL, while the levels of non-hematopoietic genes were up-regulated. In order to determine if these molecular profiles correlated with functionality, we evaluated ESC-derived LKS cells for in vitro hematopoietic-differentiation and colony formation (CFU assay). Although static culture-generated cells failed to form any colonies, they did differentiate into CD11c+ and B220+ cells indicating some hematopoietic potential. In contrast, bioreactor-derived LKS cells, when differentiated under the same conditions failed to produce any B220+ or CD11c+ cells and did not form colonies, indicating that these cells are not hematopoietic progenitors. We conclude that in vitro culture conditions significantly affect the transcriptome and functionality of ESC-derived LKS cells and although in vitro differentiated LKS cells were lineage negative and expressed both ckit and Sca-1, these cells, especially those obtained from dynamic cultures, are significantly different from native cells of the same phenotype.
Collapse
Affiliation(s)
- Irina Fernandez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Krista M. Fridley
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Dhivya Arasappan
- Genome Sequencing and Analysis Facility, The University of Texas at Austin, Austin, Texas, United States of America
| | - Rosalind V. Ambler
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Philip W. Tucker
- Department of Molecular Genetics and Microbiology, The University of Texas at Austin, Austin, Texas, United States of America
- The Institute for Cell and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
| | - Krishnendu Roy
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
- The Institute for Cell and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
18
|
Higuchi S, Lin Q, Wang J, Lim TK, Joshi SB, Anand GS, Chung MCM, Sheetz MP, Fujita H. Heart extracellular matrix supports cardiomyocyte differentiation of mouse embryonic stem cells. J Biosci Bioeng 2012; 115:320-5. [PMID: 23168383 DOI: 10.1016/j.jbiosc.2012.10.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 10/01/2012] [Accepted: 10/04/2012] [Indexed: 01/18/2023]
Abstract
We have evaluated the effect of heart extracellular matrix (ECM) on the cardiomyocyte differentiation of mouse embryonic stem cells (ES cells) using de-cellularized heart tissue. Several lines of evidence indicate that ECM plays significant roles in cell proliferation, cell death and differentiation, but role of ECM possessing a 3D structure in differentiation has not been studied in detail. We found that there are substantial differences in the quantitative protein profiles of ECM in SDS-treated heart tissue compared to that of liver tissue, as assessed by iTRAQ™ quantitative proteomics analysis. When mouse ES cells were cultured on thin (60 μm) sections of de-cellularized tissue, the expression of cardiac myosin heavy chain (cMHC) and cardiac troponin I (cTnI) was high in ES cells cultured on heart ECM compared with those cultured on liver ECM. In addition, the protein expression of cMHC and cTnI was detected in cells on heart ECM after 2 weeks, which was not detectable in cells on liver ECM. These results indicate that heart ECM plays a critical role in the cardiomyocyte differentiation of ES cells. We propose that tissue-specific ECM induced cell lineage specification through mechano-transduction mediated by the structure, elasticity and components of ECM.
Collapse
Affiliation(s)
- Sayaka Higuchi
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kaivosoja E, Barreto G, Levón K, Virtanen S, Ainola M, Konttinen YT. Chemical and physical properties of regenerative medicine materials controlling stem cell fate. Ann Med 2012; 44:635-50. [PMID: 21568670 DOI: 10.3109/07853890.2011.573805] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Regenerative medicine is a multidisciplinary field utilizing the potential of stem cells and the regenerative capability of the body to restore, maintain, or enhance tissue and organ functions. Stem cells are unspecialized cells that can self-renew but also differentiate into several somatic cells when subjected the appropriate environmental cues. The ability to reliably direct stem cell fate would provide tremendous potential for basic research and clinical therapies. Proper tissue function and regeneration rely on the spatial and temporal control of biophysical and biochemical cues, including soluble molecules, cell-cell contacts, cell-extracellular matrix contacts, and physical forces. The mechanisms involved remain poorly understood. This review focuses on the stem cell-extracellular matrix interactions by summarizing the observations of the effects of material variables (such as overall architecture, surface topography, charge, ζ-potential, surface energy, and elastic modulus) on the stem cell fate. It also deals with the mechanisms underlying the effects of these extrinsic, material variables. Insight in the environmental interactions of the stem cells is crucial for the development of new material-based approaches for cell culture experiments and future experimental and clinical regenerative medicine applications.
Collapse
Affiliation(s)
- Emilia Kaivosoja
- Department of Medicine, Institute of Clinical Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
20
|
Tan JY, Chua CK, Leong KF. Fabrication of channeled scaffolds with ordered array of micro-pores through microsphere leaching and indirect Rapid Prototyping technique. Biomed Microdevices 2012; 15:83-96. [DOI: 10.1007/s10544-012-9690-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Lee BH, Li B, Guelcher SA. Gel microstructure regulates proliferation and differentiation of MC3T3-E1 cells encapsulated in alginate beads. Acta Biomater 2012; 8:1693-702. [PMID: 22306825 DOI: 10.1016/j.actbio.2012.01.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 12/27/2011] [Accepted: 01/10/2012] [Indexed: 12/17/2022]
Abstract
For cell transplantation into damaged tissues, viable cells must be delivered to the defect site in a suitable carrier. However, the hypoxic and nutrient-limited environment in the carrier can induce massive cell death. The aims of this study were to increase the viability and regulate the behavior of osteoprogenitor cells encapsulated in alginate hydrogels through control of the gel microstructure. Cell survivability in alginate beads was improved through the use of α-MEM as the solvent for alginic acid sodium salt, and by CaCl(2) solutions, which supplied additional nutrients for the cells compared to water or buffer. The mesh size and shear modulus of the hydrogel were hypothesized to regulate proliferation and differentiation of osteoprogenitor cells. MC3T3-E1 cells demonstrated enhanced osteoblast differentiation when encapsulated in high-density alginate with smaller mesh size and more rigid mechanical properties, as confirmed by increased alkaline phosphatase activity and osteocalcin secretion. However, MC3T3-E1 cells encapsulated in low-density alginate beads with a larger mesh size and more compliant mechanical properties exhibited increased proliferation. These results demonstrate that the microstructure of alginate hydrogels can regulate the behavior of osteoprogenitor cells, thus suggesting that the tuning the properties of the gel may be a useful approach for enhancing new bone formation.
Collapse
|
22
|
Asthana A, Kisaalita WS. Microtissue size and hypoxia in HTS with 3D cultures. Drug Discov Today 2012; 17:810-7. [PMID: 22484546 DOI: 10.1016/j.drudis.2012.03.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 01/21/2012] [Accepted: 03/12/2012] [Indexed: 01/16/2023]
Abstract
The three microenvironmental factors that characterize 3D cultures include: first, chemical and/or biochemical composition, second, spatial and temporal dimensions, and third, force and/or substrate physical properties. Although these factors have been studied individually, their interdependence and synergistic interactions have not been well appreciated. We make this case by illustrating how microtissue size (spatial) and hypoxia (chemical) can be used in the formation of physiologically more relevant constructs (or not) for cell-based high-throughput screening (HTS) in drug discovery. We further show how transcriptomic and/or proteomic results from heterogeneously sized microtissues and scaffold architectures that deliberately control hypoxia can misrepresent and represent in vivo conditions, respectively. We offer guidance, depending on HTS objectives, for rational 3D culture platform choice for better emulation of in vivo conditions.
Collapse
Affiliation(s)
- Amish Asthana
- Cellular Bioengineering Laboratory, Driftmier Engineering Center, University of Georgia, Athens, GA 30602, United States
| | | |
Collapse
|
23
|
Wang M, Pei H, Zhang L, Guan L, Zhang R, Jia Y, Li B, Yue W, Wang Y, Pei X. Hepatogenesis of adipose-derived stem cells on poly-lactide-co-glycolide scaffolds: in vitro and in vivo studies. Tissue Eng Part C Methods 2011; 16:1041-50. [PMID: 20064016 DOI: 10.1089/ten.tec.2009.0244] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human adipose-derived stem cells (hASCs) have been shown to be multipotent and could be induced into various cell types, which make them the ideal cell source for cell therapy or tissue engineering. However, differentiation of ASCs into hepatocytes on three-dimensional scaffold, an important part of tissue engineering, has not been reported. In this study, to investigate the hepatogenesis of ASCs on porous poly-lactide-co-glycolide (PLGA) scaffolds, we loaded hASCs on these scaffolds. The cell-scaffold complex was implanted into the peritoneal cavity of 70% hepatectomized rats with or without 14 days of induction in hepatic inducing medium. Our results indicated that hASCs cultured on the PLGA scaffolds in the hepatic inducing medium proliferated more efficiently and could be induced into cells with hepatocyte-like phenotypic and functional properties. In vivo studies showed that induced hASCs on PLGA scaffolds survived and maintained hepatic phenotype and function for at least 14 days after implantation; moreover, noninduced hASCs on PLGA scaffolds expressed human albumin 14 days after transplantation. Collectively, these results suggest that porous PLGA scaffolds are suitable for the hepatogenesis of hASCs. These findings might be helpful in the application of hASC-based tissue engineering for liver disease therapy.
Collapse
Affiliation(s)
- Min Wang
- Stem Cell and Regenerative Medicine Laboratory, Beijing Institute of Transfusion Medicine, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lin J, Nie H, Tucker PW, Roy K. Controlled major histocompatibility complex-T cell receptor signaling allows efficient generation of functional, antigen-specific CD8+ T cells from embryonic stem cells and thymic progenitors. Tissue Eng Part A 2010; 16:2709-20. [PMID: 20380488 DOI: 10.1089/ten.tea.2009.0707] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Generation of early T cells by coculturing stem cells on notch-ligand-expressing OP9 stromal cells (OP9-DL1) has been widely reported. However, further differentiation of these cells into mature, antigen-specific, functional T cells, without retroviral transduction of T cell receptors (TcRs), is yet to be achieved. In the thymic niche this differentiation is controlled by the interaction of developing TcRs with major histocompatibility (MHC) molecules on stromal cells. We hypothesized that by providing exogenous antigen-specific MHC/TcR signals, stem and progenitor cells could be engineered into functional, effector T cells specific for the same antigen. Here we demonstrate that both thymus-derived immature T cells (double positive [DP]: CD4+CD8+) and mouse embryonic stem cells can be efficiently differentiated into antigen-specific CD8+ T cells using either MHC tetramers or peptide-loaded stromal cells. DP cells, following MHC/TcR signaling, retained elevated recombination activating gene-1 levels, suggesting continuing TcR gene rearrangement. Both DP and embryonic stem-cell-derived CD8+ T cells showed significant cytotoxic T lymphocytes activity against antigen-loaded target cells, indicating that these cells are functional. Such directed differentiation strategy could provide an efficient method for generating functional, antigen-specific T cells from stem cells for potential use in adoptive T cell therapy.
Collapse
Affiliation(s)
- Jian Lin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | |
Collapse
|
25
|
Dickinson LE, Kusuma S, Gerecht S. Reconstructing the differentiation niche of embryonic stem cells using biomaterials. Macromol Biosci 2010; 11:36-49. [PMID: 20967797 DOI: 10.1002/mabi.201000245] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 07/30/2010] [Indexed: 01/14/2023]
Abstract
The biochemical cues and topographical architecture of the extracellular environment extensively influence ES cell fate. The microenvironment surrounding the developing embryo presents these instructive cues in a complex and interactive manner in order to guide cell fate decisions. Current stem cell research aims to reconstruct this multifaceted embryonic niche to recapitulate development in vitro. This review focuses on 2D and 3D differentiation niches created from natural and synthetic biomaterials to guide the differentiation of ES cells toward specific lineages. Biomaterials engineered to present specific physical constraints are also reviewed for their role in differentiation.
Collapse
Affiliation(s)
- Laura E Dickinson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, 3400 North Charles Street, Baltimore, MD 21210, USA
| | | | | |
Collapse
|
26
|
Di Maggio N, Piccinini E, Jaworski M, Trumpp A, Wendt DJ, Martin I. Toward modeling the bone marrow niche using scaffold-based 3D culture systems. Biomaterials 2010; 32:321-9. [PMID: 20952054 DOI: 10.1016/j.biomaterials.2010.09.041] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 09/19/2010] [Indexed: 01/12/2023]
Abstract
In the bone marrow, specialized microenvironments, called niches, regulate hematopoietic stem cell (HSC) maintenance and function through a complex crosstalk between different cell types. Although in vivo studies have been instrumental to elucidate some of the mechanisms by which niches exert their function, the establishment of an in vitro model that recapitulates the fundamental interactions of the niche components in a controlled setting would be of great benefit. We have previously shown that freshly harvested bone marrow- or adipose tissue-derived cells can be cultured under perfusion within porous scaffolds, allowing the formation of an organized 3D stromal tissue, composed by mesenchymal and endothelial progenitors and able to support hematopoiesis. Here we describe 3D scaffold-based perfusion systems as potential models to reconstruct ex vivo the bone marrow stem cell niche. We discuss how several culture parameters, including scaffold properties, cellular makeup and molecular signals, can be varied and controlled to investigate the role of specific cues in affecting HSC fate. We then provide a perspective of how the system could be exploited to improve stem cell-based therapies and how the model can be extended toward the engineering of other specialized stromal niches.
Collapse
Affiliation(s)
- Nunzia Di Maggio
- Departments of Surgery and of Biomedicine, Basel University Hospital, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
27
|
Fridley KM, Fernandez I, Li MTA, Kettlewell RB, Roy K. Unique differentiation profile of mouse embryonic stem cells in rotary and stirred tank bioreactors. Tissue Eng Part A 2010; 16:3285-98. [PMID: 20528675 DOI: 10.1089/ten.tea.2010.0166] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Embryonic stem (ES)-cell-derived lineage-specific stem cells, for example, hematopoietic stem cells, could provide a potentially unlimited source for transplantable cells, especially for cell-based therapies. However, reproducible methods must be developed to maximize and scale-up ES cell differentiation to produce clinically relevant numbers of therapeutic cells. Bioreactor-based dynamic culture conditions are amenable to large-scale cell production, but few studies have evaluated how various bioreactor types and culture parameters influence ES cell differentiation, especially hematopoiesis. Our results indicate that cell seeding density and bioreactor speed significantly affect embryoid body formation and subsequent generation of hematopoietic stem and progenitor cells in both stirred tank (spinner flask) and rotary microgravity (Synthecon™) type bioreactors. In general, high percentages of hematopoietic stem and progenitor cells were generated in both bioreactors, especially at high cell densities. In addition, Synthecon bioreactors produced more sca-1(+) progenitors and spinner flasks generated more c-Kit(+) progenitors, demonstrating their unique differentiation profiles. cDNA microarray analysis of genes involved in pluripotency, germ layer formation, and hematopoietic differentiation showed that on day 7 of differentiation, embryoid bodies from both bioreactors consisted of all three germ layers of embryonic development. However, unique gene expression profiles were observed in the two bioreactors; for example, expression of specific hematopoietic genes were significantly more upregulated in the Synthecon cultures than in spinner flasks. We conclude that bioreactor type and culture parameters can be used to control ES cell differentiation, enhance unique progenitor cell populations, and provide means for large-scale production of transplantable therapeutic cells.
Collapse
Affiliation(s)
- Krista M Fridley
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | | | |
Collapse
|
28
|
Forciniti L, Guimard NK, Lee S, Schmidt CE. Unique electrochemically synthesized polypyrrole:poly(lactic-co-glycolic acid) blends for biomedical applications. ACTA ACUST UNITED AC 2010. [DOI: 10.1039/c0jm01015d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
29
|
Wang L, Wang ZH, Shen CY, You ML, Xiao JF, Chen GQ. Differentiation of human bone marrow mesenchymal stem cells grown in terpolyesters of 3-hydroxyalkanoates scaffolds into nerve cells. Biomaterials 2009; 31:1691-8. [PMID: 19962755 DOI: 10.1016/j.biomaterials.2009.11.053] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 11/18/2009] [Indexed: 12/18/2022]
Abstract
Polyhydroxyalkanoates, abbreviated as PHA, have been studied for medical applications due to their suitable mechanical properties, blood and tissue tolerance and in vivo biodegradability. As a new member of PHA family, terpolyester of 3-hydroxybutyrate, 3-hydroxyvalerate and 3-hydroxyhexanoate, abbreviated as PHBVHHx, was compared with polylactic acid (PLA), copolyester of 3-hydroxybutyrate and 3-hydroxyhexanoate (PHBHHx) for their respective functions leading to differentiation of human bone marrow mesenchymal stem cell (hBMSC) into nerve cells. Results indicated that 3D scaffolds promoted the differentiation of hBMSC into nerve cells more intensively compared with 2D films. Smaller pore sizes of scaffolds increased differentiation of hBMSC into nerve cells, whereas decreased cell proliferation. PHBVHHx scaffolds with pore sizes of 30-60 microm could be used in nerve tissue engineering for treatment of nerve injury. The above results were supported by scanning electron microscope (SEM) and confocal microscopy observation on attachment and growth of hBMSCs on PLA, PHBHHx and PHBVHHx, and by CCK-8 evaluation of cell proliferation. In addition, expressions of nerve markers nestin, GFAP and beta-III tubulin of nerve cells differentiated from hBMSC grown in PHBVHHx scaffolds were confirmed by real-time PCR.
Collapse
Affiliation(s)
- Lei Wang
- Multidisciplinary Research Center, Shantou University, Shantou 515063, Guangdong, China
| | | | | | | | | | | |
Collapse
|
30
|
Ofek G, Willard VP, Koay EJ, Hu JC, Lin P, Athanasiou KA. Mechanical characterization of differentiated human embryonic stem cells. J Biomech Eng 2009; 131:061011. [PMID: 19449965 DOI: 10.1115/1.3127262] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human embryonic stem cells (hESCs) possess an immense potential in a variety of regenerative applications. A firm understanding of hESC mechanics, on the single cell level, may provide great insight into the role of biophysical forces in the maintenance of cellular phenotype and elucidate mechanical cues promoting differentiation along various mesenchymal lineages. Moreover, cellular biomechanics can provide an additional tool for characterizing stem cells as they follow certain differentiation lineages, and thus may aid in identifying differentiated hESCs, which are most suitable for tissue engineering. This study examined the viscoelastic properties of single undifferentiated hESCs, chondrogenically differentiated hESC subpopulations, mesenchymal stem cells (MSCs), and articular chondrocytes (ACs). hESC chondrogenesis was induced using either transforming growth factor-beta1 (TGF-beta1) or knock out serum replacer as differentiation agents, and the resulting cell populations were separated based on density. All cell groups were mechanically tested using unconfined creep cytocompression. Analyses of subpopulations from all differentiation regimens resulted in a spectrum of mechanical and morphological properties spanning the range of hESCs to MSCs to ACs. Density separation was further successful in isolating cellular subpopulations with distinct mechanical properties. The instantaneous and relaxed moduli of subpopulations from TGF-beta1 differentiation regimen were statistically greater than those of undifferentiated hESCs. In addition, two subpopulations from the TGF-beta1 group were identified, which were not statistically different from native articular chondrocytes in their instantaneous and relaxed moduli, as well as their apparent viscosity. Identification of a differentiated hESC subpopulation with similar mechanical properties as native chondrocytes may provide an excellent cell source for tissue engineering applications. These cells will need to withstand any mechanical stimulation regimen employed to augment the mechanical and biochemical characteristics of the neotissue. Density separation was effective at purifying distinct populations of cells. A differentiated hESC subpopulation was identified with both similar mechanical and morphological characteristics as ACs. Future research may utilize this cell source in cartilage regeneration efforts.
Collapse
Affiliation(s)
- Gidon Ofek
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | | | | | | | | | | |
Collapse
|
31
|
Placzek MR, Chung IM, Macedo HM, Ismail S, Mortera Blanco T, Lim M, Cha JM, Fauzi I, Kang Y, Yeo DCL, Ma CYJ, Polak JM, Panoskaltsis N, Mantalaris A. Stem cell bioprocessing: fundamentals and principles. J R Soc Interface 2009; 6:209-32. [PMID: 19033137 PMCID: PMC2659585 DOI: 10.1098/rsif.2008.0442] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In recent years, the potential of stem cell research for tissue engineering-based therapies and regenerative medicine clinical applications has become well established. In 2006, Chung pioneered the first entire organ transplant using adult stem cells and a scaffold for clinical evaluation. With this a new milestone was achieved, with seven patients with myelomeningocele receiving stem cell-derived bladder transplants resulting in substantial improvements in their quality of life. While a bladder is a relatively simple organ, the breakthrough highlights the incredible benefits that can be gained from the cross-disciplinary nature of tissue engineering and regenerative medicine (TERM) that encompasses stem cell research and stem cell bioprocessing. Unquestionably, the development of bioprocess technologies for the transfer of the current laboratory-based practice of stem cell tissue culture to the clinic as therapeutics necessitates the application of engineering principles and practices to achieve control, reproducibility, automation, validation and safety of the process and the product. The successful translation will require contributions from fundamental research (from developmental biology to the 'omics' technologies and advances in immunology) and from existing industrial practice (biologics), especially on automation, quality assurance and regulation. The timely development, integration and execution of various components will be critical-failures of the past (such as in the commercialization of skin equivalents) on marketing, pricing, production and advertising should not be repeated. This review aims to address the principles required for successful stem cell bioprocessing so that they can be applied deftly to clinical applications.
Collapse
Affiliation(s)
- Mark R Placzek
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
The engineering of patient-specific, anatomically shaped, digits. Biomaterials 2009; 30:2735-40. [PMID: 19203788 DOI: 10.1016/j.biomaterials.2009.01.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 01/19/2009] [Indexed: 11/20/2022]
Abstract
It is now recognized that geometric structures of scaffolds at several size levels have profound influences on cell adhesion, viability, proliferation and differentiation. This study aims to develop an integrated process to fabricate scaffolds with controllable geometric structures at nano-, micro- and macro-scales. A phase-separation method is used to prepare interconnected poly(L-lactide) (PLLA) nanofibrous (NF) scaffolds. The pore size of the NF scaffold at the scale of several hundred micrometers is controlled by the size of porogen, paraffin spheres. At millimeter scale and above, the overall shape of the scaffold is defined by a wax mold produced using a three-dimensional printer. The printer utilizes a stereo lithographic file generated from computed tomographic files retrieved from the National Library of Medicine's Visual Human Project. NF PLLA scaffolds with a human digit shape are successfully prepared using this process. Osteoblast cell line MC3T3-E1 cells are then seeded and cultured in the prepared scaffolds. Cell proliferation, differentiation and biomineralization are characterized to demonstrate the suitability of the scaffolds for the digit bone tissue engineering application.
Collapse
|
33
|
Nichols JE, Cortiella J, Lee J, Niles JA, Cuddihy M, Wang S, Bielitzki J, Cantu A, Mlcak R, Valdivia E, Yancy R, McClure ML, Kotov NA. In vitro analog of human bone marrow from 3D scaffolds with biomimetic inverted colloidal crystal geometry. Biomaterials 2008; 30:1071-9. [PMID: 19042018 DOI: 10.1016/j.biomaterials.2008.10.041] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2008] [Accepted: 10/22/2008] [Indexed: 11/28/2022]
Abstract
In vitro replicas of bone marrow can potentially provide a continuous source of blood cells for transplantation and serve as a laboratory model to examine human immune system dysfunctions and drug toxicology. Here we report the development of an in vitro artificial bone marrow based on a 3D scaffold with inverted colloidal crystal (ICC) geometry mimicking the structural topology of actual bone marrow matrix. To facilitate adhesion of cells, scaffolds were coated with a layer of transparent nanocomposite. After seeding with hematopoietic stem cells (HSCs), ICC scaffolds were capable of supporting expansion of CD34+ HSCs with B-lymphocyte differentiation. Three-dimensional organization was shown to be critical for production of B cells and antigen-specific antibodies. Functionality of bone marrow constructs was confirmed by implantation of matrices containing human CD34+ cells onto the backs of severe combined immunodeficiency (SCID) mice with subsequent generation of human immune cells.
Collapse
Affiliation(s)
- Joan E Nichols
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Neurite Outgrowth on a DNA Crosslinked Hydrogel with Tunable Stiffnesses. Ann Biomed Eng 2008; 36:1565-79. [DOI: 10.1007/s10439-008-9530-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Accepted: 06/19/2008] [Indexed: 01/25/2023]
|
35
|
Stevens B, Yang Y, Mohandas A, Stucker B, Nguyen KT. A review of materials, fabrication methods, and strategies used to enhance bone regeneration in engineered bone tissues. J Biomed Mater Res B Appl Biomater 2008; 85:573-82. [PMID: 17937408 DOI: 10.1002/jbm.b.30962] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Over the last decade, bone engineered tissues have been developed as alternatives to autografts and allografts to repair and reconstruct bone defects. This article provides a review of the current technologies in bone tissue engineering. Factors used for fabrication of three-dimensional bone scaffolds such as materials, cells, and biomolecular signals, as well as required properties for ideal bone scaffolds, are reviewed. In addition, current fabrication techniques including rapid prototyping are elaborated upon. Finally, this review article further discusses some effective strategies to enhance cell ingrowth in bone engineered tissues; for example, nanotopography, biomimetic materials, embedded growth factors, mineralization, and bioreactors. In doing so, it suggests that there is a possibility to develop bone substitutes that can repair bone defects and promote new bone formation for orthopedic applications.
Collapse
Affiliation(s)
- Brian Stevens
- Department of Biological and Irrigation Engineering, Utah State University, Logan, Utah, USA
| | | | | | | | | |
Collapse
|
36
|
Hwang DW, Jang SJ, Kim YH, Kim HJ, Shim IK, Jeong JM, Chung JK, Lee MC, Lee SJ, Kim SU, Kim S, Lee DS. Real-time in vivo monitoring of viable stem cells implanted on biocompatible scaffolds. Eur J Nucl Med Mol Imaging 2008; 35:1887-98. [PMID: 18437378 DOI: 10.1007/s00259-008-0751-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Accepted: 02/19/2008] [Indexed: 12/01/2022]
Abstract
PURPOSE Three-dimensional fibrous scaffolds provide an environment that enhances transplanted stem cell survival in vivo and facilitates imaging their localization, viability, and growth in vivo. To assess transplanted stem cell viability on biocompatible polymer scaffolds in vivo, we developed in vivo imaging systems for evaluation of implanted viable neural stem cells (NSC) and mesenchymal stem cells (MSC) on scaffolds using luciferase or sodium/iodide symporter (NIS) genes. METHODS Firefly luciferase stably expressing-C6 cell was established (C6-Fluc). The human neural stem cell, F3, was infected with adenoviral vector carrying luciferase gene (F3-Fluc) and MSC expressing NIS controlled by ubiquitin C promoter using lentiviral vector was established by treating blasticidine for 2 weeks (MSC-NIS). Chitosan and poly L-lactic acid (PLLA) scaffolds were used for in vivo image. In vivo expression of luciferase and human NIS was examined by bioluminescence image or (99m)Tc-pertechnetate gamma camera image, respectively. The cell/scaffold complex was implanted into subcutaneous or abdominal area of BALB/C nude mouse. For quantitative evaluation of cell viability, regions of interest were drawn on (99m)Tc-pertechnetate scintigraphy by manual. RESULTS The gradual increase of luciferase activity was observed in C6-Fluc seeded with chitosan according to the increase in the number of cells. C6-Fluc/chitosan complex subcutaneously implanted into nude mice showed longitudinal bioluminescence image until 34 days. Luciferase image of abdominal-injected C6-Fluc/PLLA complex was saturated in only 14 days, showing great cell growth due to abundant nutrients. F3 cells showed well-incorporated pattern with fibrous chitosan scaffold using scanning electron microscopy. F3 infected with Ad-Fluc showed >100-fold higher luciferase activity than luciferase activity in F3. Cell-number-dependent increase of luciferase activity was shown in F3-Fluc seeded on chitosan. F3-Fluc incorporation into chitosan after abdominal injection was clearly visible on bioluminescence image up to 11 days. Radionuclide imaging showed higher uptake by MSC-NIS on PLLA scaffolds than by MSC-NIS not seeded on a scaffold. Quantitative data showed significantly better survival of MSC-NIS on PLLA scaffolds than without scaffold at 72 h post-implantation, which concurred with histologic findings. CONCLUSION These results suggest that NSC-Fluc and MSC-NIS cells incorporated within polymer scaffolds can be monitored on a long-term basis by serial in vivo imaging. We believe that a biocompatible scaffold-based imaging system could be used to assess stem cell viabilities in a non-invasive way to aid the development of regenerative therapeutics.
Collapse
Affiliation(s)
- Do Won Hwang
- Programs in Neuroscience, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hanson AD, Wall ME, Pourdeyhimi B, Loboa EG. Effects of oxygen plasma treatment on adipose-derived human mesenchymal stem cell adherence to poly(L-lactic acid) scaffolds. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2008; 18:1387-400. [PMID: 17961322 DOI: 10.1163/156856207782246812] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Plasma treatment of substrate surfaces can be utilized to improve adhesion of cells to tissue-engineered scaffolds. The purpose of this study was to enhance cell adhesion to non-woven poly(L-lactic acid) (PLLA) scaffolds using oxygen plasma treatment to increase surface hydroxyl groups and thereby enhance substrate hydrophilicity. It was hypothesized that oxygen plasma treatment would increase the number of adipose-derived human mesenchymal stem cells (hMSCs) that adhered to melt-blown, non-woven PLLA scaffolds without affecting cell viability. The number of cells that adhered to the oxygen plasma-treated (10 min at 100 W) or untreated PLLA scaffolds was assessed at 2, 4, 8, 12, 24 and 48 h post-seeding via DNA analysis. Cell viability and morphology were also assessed at 2, 4, 8, 12 and 24 h post-seeding via a live/dead assay and hematoxylin staining, respectively. Oxygen plasma treatment decreased the contact angle of water from 75.6 degrees to 58.2 degrees , indicating an increase in the surface hydrophilicity of PLLA. The results of the DNA analysis indicated that there was an increased number of hMSCs on oxygen plasma treated scaffolds for two of the three donors. In addition, oxygen plasma treatment promoted a more even distribution of hMSCs throughout the scaffold and enhanced cell spreading at earlier time points without altering cell viability. This early induction of cell spreading and the uniform distribution of cells, in turn, may increase future proliferation and differentiation of hMSCs under conditions that simulate the microenvironment in vivo.
Collapse
Affiliation(s)
- Ariel D Hanson
- Joint Department of Biomedical Engineering, 2142 Burlington Nuclear Engineering Laboratories, Campus Box 7115, North Carolina State University, Raleigh, NC 27695, USA
| | | | | | | |
Collapse
|
38
|
Dawson E, Mapili G, Erickson K, Taqvi S, Roy K. Biomaterials for stem cell differentiation. Adv Drug Deliv Rev 2008; 60:215-28. [PMID: 17997187 DOI: 10.1016/j.addr.2007.08.037] [Citation(s) in RCA: 293] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Accepted: 08/11/2007] [Indexed: 12/13/2022]
Abstract
The promise of cellular therapy lies in the repair of damaged organs and tissues in vivo as well as generating tissue constructs in vitro for subsequent transplantation. Unfortunately, the lack of available donor cell sources limits its ultimate clinical applicability. Stem cells are a natural choice for cell therapy due to their pluripotent nature and self-renewal capacity. Creating reserves of undifferentiated stem cells and subsequently driving their differentiation to a lineage of choice in an efficient and scalable manner is critical for the ultimate clinical success of cellular therapeutics. In recent years, a variety of biomaterials have been incorporated in stem cell cultures, primarily to provide a conducive microenvironment for their growth and differentiation and to ultimately mimic the stem cell niche. In this review, we examine applications of natural and synthetic materials, their modifications as well as various culture conditions for maintenance and lineage-specific differentiation of embryonic and adult stem cells.
Collapse
Affiliation(s)
- Eileen Dawson
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | | | |
Collapse
|
39
|
Hwang NS, Varghese S, Elisseeff J. Controlled differentiation of stem cells. Adv Drug Deliv Rev 2008; 60:199-214. [PMID: 18006108 DOI: 10.1016/j.addr.2007.08.036] [Citation(s) in RCA: 227] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 08/18/2007] [Indexed: 12/13/2022]
Abstract
The extracellular microenvironment plays a significant role in controlling cellular behavior. Identification of appropriate biomaterials that support cellular attachment, proliferation and, most importantly in the case of human embryonic stem cells, lineage-specific differentiation is critical for tissue engineering and cellular therapy. In addition to growth factors and morphogenetic factors known to induce lineage commitment of stem cells, a number of scaffolding materials, including synthetic and naturally-derived biomaterials, have been utilized in tissue engineering approaches to direct differentiation. This review focuses on recent emerging findings and well-characterized differentiation models of human embryonic stem cells. Additionally, we also discuss about various strategies that have been used in stem cell expansion.
Collapse
|
40
|
Jell G, Verdejo R, Safinia L, Shaffer MSP, Stevens MM, Bismarck A. Carbon nanotube-enhanced polyurethane scaffolds fabricated by thermally induced phase separation. ACTA ACUST UNITED AC 2008. [DOI: 10.1039/b716109c] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
Mei Y, Goldberg M, Anderson D. The development of high-throughput screening approaches for stem cell engineering. Curr Opin Chem Biol 2007; 11:388-93. [PMID: 17702642 PMCID: PMC3017753 DOI: 10.1016/j.cbpa.2007.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2007] [Revised: 06/04/2007] [Accepted: 07/13/2007] [Indexed: 01/10/2023]
Abstract
It has become increasingly clear that both soluble factors, such as growth factors, and insoluble factors, including the surfaces on which cells grow, can have controlling effects on stem cell behavior and differentiation. While much progress has been made in biomaterial design and application, the rational design of biomaterial cues to direct stem cell behavior and differentiation remains challenging. Recent advances in automated, high-throughput methods for synthesizing and screening combinatorial biomaterial libraries and cellular microenvironments promise to accelerate the discovery of factors that control stem cell behavior. Specific examples include miniaturized, automated, combinatorial material synthesis and extracellular matrix screening methods as well microarrayed methods for creating local microenvironments of soluble factors, such as small molecules, siRNA, and other signaling molecules.
Collapse
Affiliation(s)
- Ying Mei
- Department of Chemical Engineering, MIT, 77 Massachusetts Avenue, E25-342, Cambridge, MA 02139, United States.
| | | | | |
Collapse
|
42
|
Pisu M, Concas A, Cao G. A novel simulation model for stem cells differentiation. J Biotechnol 2007; 130:171-82. [PMID: 17459507 DOI: 10.1016/j.jbiotec.2007.02.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 02/12/2007] [Accepted: 02/27/2007] [Indexed: 11/24/2022]
Abstract
A novel mathematical model to simulate mesenchymal stem cells differentiation into specialized cells is proposed. The model is based upon material balances for extracellular matrix compounds, growth factors and nutrients coupled with a mass-structured population balance describing cell growth, proliferation and differentiation. The proposed model is written in a general form and it may be used to simulate a generic cell differentiation pathway occurring in vivo or during in vitro cultivation when specific growth factors are used. Literature experimental data concerning the differentiation of mesenchymal stem cells into chondrocytes in terms of total DNA and glycosaminoglycan content are successfully compared with model results, thus demonstrating the validity of the proposed model as well as its predictive capability. A further test of the model capability is performed for the case of in vivo fracture healing during which mesenchymal stem cells differentiate into chondrocytes and osteoblasts. Considerations about the extension of the proposed model to different pathologies beside fracture healing are reported. Finally, sensitivity analysis of model parameters is also performed in order to clarify what mechanisms most strongly influence differentiation and the distribution of cell types.
Collapse
Affiliation(s)
- Massimo Pisu
- CRS4 (Center for Advanced Studies, Research and Development in Sardinia), Parco Scientifico e Tecnologico POLARIS, Edificio 1, 09010 Pula, Cagliari, Italy
| | | | | |
Collapse
|