1
|
van Sprang JF, Aarts JGM, Arts B, Brouns JEP, Komil MI, Bartels PAA, Dankers PYW. Supramolecular Additive Screening to Engineer Microfibrous Rafts for Expansion of Pluripotent Stem Cells in Dynamic Suspension. Adv Healthc Mater 2025; 14:e2404186. [PMID: 40059619 PMCID: PMC12023819 DOI: 10.1002/adhm.202404186] [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: 10/27/2024] [Revised: 02/02/2025] [Indexed: 04/26/2025]
Abstract
Human induced pluripotent stem cells (hiPSCs) hold the potential to generate any human tissue for transplantation in regenerative therapies. These complex cell therapies require billions of cells, which is challenging to acquire in planar adherent cultures. Transitioning hiPSCs to 3D suspension culture on microcarrier materials, often bead-shaped, improves the total surface area accessible to cells, thereby enabling culture scale-up. However, bead-shaped microcarriers do not have the optimal shape configuration, because it is the lowest surface-to-volume ratio of all geometrical shapes, and it also induces uncontrolled cell clumping. Application of synthetic, microfibrous rafts as a replacement for bead-shaped microcarriers potentially solves these issues. Here, microfibrous rafts are engineered by first screening a supramolecular biomaterial library composed of bisurea (BU)-peptide conjugate additives for its ability to induce hiPSC adhesion and maintenance of its pluripotent state, followed by electrospinning the screening-hit into raft-like structures. The resulting rafts contain cylinder-like microfibers, which have a higher surface-to-volume ratio compared to conventional bead-shaped microcarriers, and the flat configuration of the rafts prevents clumping.
Collapse
Affiliation(s)
- Johnick F. van Sprang
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Jasper G. M. Aarts
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Boris Arts
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Joyce E. P. Brouns
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Muhabbat I. Komil
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Paul A. A. Bartels
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| |
Collapse
|
2
|
Redondo-Gómez C, Parreira P, Martins MCL, Azevedo HS. Peptide-based self-assembled monolayers (SAMs): what peptides can do for SAMs and vice versa. Chem Soc Rev 2024; 53:3714-3773. [PMID: 38456490 DOI: 10.1039/d3cs00921a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Self-assembled monolayers (SAMs) represent highly ordered molecular materials with versatile biochemical features and multidisciplinary applications. Research on SAMs has made much progress since the early begginings of Au substrates and alkanethiols, and numerous examples of peptide-displaying SAMs can be found in the literature. Peptides, presenting increasing structural complexity, stimuli-responsiveness, and biological relevance, represent versatile functional components in SAMs-based platforms. This review examines the major findings and progress made on the use of peptide building blocks displayed as part of SAMs with specific functions, such as selective cell adhesion, migration and differentiation, biomolecular binding, advanced biosensing, molecular electronics, antimicrobial, osteointegrative and antifouling surfaces, among others. Peptide selection and design, functionalisation strategies, as well as structural and functional characteristics from selected examples are discussed. Additionally, advanced fabrication methods for dynamic peptide spatiotemporal presentation are presented, as well as a number of characterisation techniques. All together, these features and approaches enable the preparation and use of increasingly complex peptide-based SAMs to mimic and study biological processes, and provide convergent platforms for high throughput screening discovery and validation of promising therapeutics and technologies.
Collapse
Affiliation(s)
- Carlos Redondo-Gómez
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - Paula Parreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| | - M Cristina L Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Helena S Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
| |
Collapse
|
3
|
Abdal Dayem A, Lee SB, Lim KM, Kim A, Shin HJ, Vellingiri B, Kim YB, Cho SG. Bioactive peptides for boosting stem cell culture platform: Methods and applications. Biomed Pharmacother 2023; 160:114376. [PMID: 36764131 DOI: 10.1016/j.biopha.2023.114376] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Peptides, short protein fragments, can emulate the functions of their full-length native counterparts. Peptides are considered potent recombinant protein alternatives due to their specificity, high stability, low production cost, and ability to be easily tailored and immobilized. Stem cell proliferation and differentiation processes are orchestrated by an intricate interaction between numerous growth factors and proteins and their target receptors and ligands. Various growth factors, functional proteins, and cellular matrix-derived peptides efficiently enhance stem cell adhesion, proliferation, and directed differentiation. For that, peptides can be immobilized on a culture plate or conjugated to scaffolds, such as hydrogels or synthetic matrices. In this review, we assess the applications of a variety of peptides in stem cell adhesion, culture, organoid assembly, proliferation, and differentiation, describing the shortcomings of recombinant proteins and their full-length counterparts. Furthermore, we discuss the challenges of peptide applications in stem cell culture and materials design, as well as provide a brief outlook on future directions to advance peptide applications in boosting stem cell quality and scalability for clinical applications in tissue regeneration.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Hyun Jin Shin
- Department of Ophthalmology, Research Institute of Medical Science, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute, Konkuk University, Seoul 05029, Republic of Korea; R&D Team, StemExOne co., ltd. 303, Life Science Bldg, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
4
|
Eve A. Transitions in development - an interview with Samira Musah. Development 2023; 150:dev201539. [PMID: 36637122 DOI: 10.1242/dev.201539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Samira Musah is an Assistant Professor in the Departments of Biomedical Engineering and Medicine at Duke University, USA. Samira's research focuses on leveraging pluripotent stem cells, bioengineering and organ-on-a-chip technologies to understand more about human kidney development, disease and therapy. We met with Samira over Microsoft Teams to hear more about her path to independence, mentors and her love of yoga.
Collapse
|
5
|
Cable J, Lutolf MP, Fu J, Park SE, Apostolou A, Chen S, Song CJ, Spence JR, Liberali P, Lancaster M, Meier AB, Pek NMQ, Wells JM, Capeling MM, Uzquiano A, Musah S, Huch M, Gouti M, Hombrink P, Quadrato G, Urenda JP. Organoids as tools for fundamental discovery and translation-a Keystone Symposia report. Ann N Y Acad Sci 2022; 1518:196-208. [PMID: 36177906 PMCID: PMC11293861 DOI: 10.1111/nyas.14874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Complex three-dimensional in vitro organ-like models, or organoids, offer a unique biological tool with distinct advantages over two-dimensional cell culture systems, which can be too simplistic, and animal models, which can be too complex and may fail to recapitulate human physiology and pathology. Significant progress has been made in driving stem cells to differentiate into different organoid types, though several challenges remain. For example, many organoid models suffer from high heterogeneity, and it can be difficult to fully incorporate the complexity of in vivo tissue and organ development to faithfully reproduce human biology. Successfully addressing such limitations would increase the viability of organoids as models for drug development and preclinical testing. On April 3-6, 2022, experts in organoid development and biology convened at the Keystone Symposium "Organoids as Tools for Fundamental Discovery and Translation" to discuss recent advances and insights from this relatively new model system into human development and disease.
Collapse
Affiliation(s)
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Roche Institute for Translational Bioengineering (ITB), Pharma Research and Early Development (pRED), F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sunghee Estelle Park
- Department of Bioengineering and NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Athanasia Apostolou
- Emulate Inc, Boston, Massachusetts, USA
- Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York City, New York, USA
| | - Cheng Jack Song
- Keck Medicine of University of Southern California, Los Angeles, California, USA
| | - Jason R Spence
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI) and University of Basel, Basel, Switzerland
| | | | - Anna B Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nicole Min Qian Pek
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, Ohio, USA
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - James M Wells
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, Ohio, USA
- Division of Developmental Biology and Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Ana Uzquiano
- Department of Stem Cell and Regenerative Biology, Harvard University
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samira Musah
- Developmental and Stem Cell Biology Program and Division of Nephrology, Department of Medicine and Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Biomolecular and Tissue Engineering, Durham, North Carolina, USA
- Department of Biomedical Engineering, Pratt School of Engineering, Durham, North Carolina, USA
- Duke Regeneration Center, Duke University, Durham, North Carolina, USA
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Mina Gouti
- Stem Cell Modelling of Development & Disease Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Pleun Hombrink
- University Medical Center Utrecht and HUB Organoids, Utrecht, Netherlands
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine and Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, USA
| | - Jean-Paul Urenda
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine and Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Ramasubramanian A, Muckom R, Sugnaux C, Fuentes C, Ekerdt BL, Clark DS, Healy KE, Schaffer DV. High-Throughput Discovery of Targeted, Minimally Complex Peptide Surfaces for Human Pluripotent Stem Cell Culture. ACS Biomater Sci Eng 2021; 7:1344-1360. [PMID: 33750112 DOI: 10.1021/acsbiomaterials.0c01462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human pluripotent stem cells harbor an unlimited capacity to generate therapeutically relevant cells for applications in regenerative medicine. However, to utilize these cells in the clinic, scalable culture systems that activate defined receptors and signaling pathways to sustain stem cell self-renewal are required; and synthetic materials offer considerable promise to meet these needs. De novo development of materials that target novel pathways has been stymied by a limited understanding of critical receptor interactions maintaining pluripotency. Here, we identify peptide agonists for the human pluripotent stem cell (hPSC) laminin receptor and pluripotency regulator, α6-integrin, through unbiased, library-based panning strategies. Biophysical characterization of adhesion suggests that identified peptides bind hPSCs through α6-integrin with sub-μM dissociation constants similar to laminin. By harnessing a high-throughput microculture platform, we developed predictive guidelines for presenting these integrin-targeting peptides alongside canonical binding motifs at optimal stoichiometries to generate nascent culture surfaces. Finally, when presented as self-assembled monolayers, predicted peptide combinations supported hPSC expansion, highlighting how unbiased screens can accelerate the discovery of targeted biomaterials.
Collapse
Affiliation(s)
- Anusuya Ramasubramanian
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Riya Muckom
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Caroline Sugnaux
- Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Christina Fuentes
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Barbara L Ekerdt
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Douglas S Clark
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - David V Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
7
|
Engineering Biophysical Cues for Controlled 3D Differentiation of Endoderm Derivatives. Methods Mol Biol 2021. [PMID: 33340355 DOI: 10.1007/978-1-0716-1174-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Biophysical cues synergize with biochemical cues to drive differentiation of pluripotent stem cells through specific phenotypic trajectory. Tools to manipulate the cell biophysical environment and identify the influence of specific environment perturbation in the presence of combinatorial inputs will be critical to control the development trajectory. Here we describe the procedure to perturb biophysical environment of pluripotent stem cells while maintaining them in 3D culture configuration. We also discuss a high-throughput platform for combinatorial perturbation of the cell microenvironment, and detail a statistical procedure to extract dominant environmental influences.
Collapse
|
8
|
Kulkarni A, Mochnáčová E, Majerova P, Čurlík J, Bhide K, Mertinková P, Bhide M. Single Domain Antibodies Targeting Receptor Binding Pockets of NadA Restrain Adhesion of Neisseria meningitidis to Human Brain Microvascular Endothelial Cells. Front Mol Biosci 2020; 7:573281. [PMID: 33425985 PMCID: PMC7785856 DOI: 10.3389/fmolb.2020.573281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/03/2020] [Indexed: 01/02/2023] Open
Abstract
Neisseria adhesin A (NadA), one of the surface adhesins of Neisseria meningitides (NM), interacts with several cell types including human brain microvascular endothelial cells (hBMECs) and play important role in the pathogenesis. Receptor binding pockets of NadA are localized on the globular head domain (A33 to K69) and the first coiled-coil domain (L121 to K158). Here, the phage display was used to develop a variable heavy chain domain (VHH) that can block receptor binding sites of recombinant NadA (rec-NadA). A phage library displaying VHH was panned against synthetic peptides (NadA-gdA33−K69 or NadA-ccL121−K158), gene encoding VHH was amplified from bound phages and re-cloned in the expression vector, and the soluble VHHs containing disulfide bonds were overexpressed in the SHuffle E. coli. From the repertoire of 96 clones, two VHHs (VHHF3–binding NadA-gdA33−K69 and VHHG9–binding NadA-ccL121−K158) were finally selected as they abrogated the interaction between rec-NadA and the cell receptor. Preincubation of NM with VHHF3 and VHHG9 significantly reduced the adhesion of NM on hBMECs in situ and hindered the traversal of NM across the in-vitro BBB model. The work presents a phage display pipeline with a single-round of panning to select receptor blocking VHHs. It also demonstrates the production of soluble and functional VHHs, which blocked the interaction between NadA and its receptor, decreased adhesion of NM on hBMECs, and reduced translocation of NM across BBB in-vitro. The selected NadA blocking VHHs could be promising molecules for therapeutic translation.
Collapse
Affiliation(s)
- Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia.,Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Evelína Mochnáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Petra Majerova
- Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ján Čurlík
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Patrícia Mertinková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy, Kosice, Slovakia.,Institute of Neuroimmunology of Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
9
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
10
|
Khalil AS, Xie AW, Johnson HJ, Murphy WL. Sustained release and protein stabilization reduce the growth factor dosage required for human pluripotent stem cell expansion. Biomaterials 2020; 248:120007. [PMID: 32302801 PMCID: PMC8445021 DOI: 10.1016/j.biomaterials.2020.120007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 02/24/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
Translation of human pluripotent stem cell (hPSC)-derived therapies to the clinic demands scalable, cost-effective methods for cell expansion. Culture media currently used for hPSC expansion rely on high concentrations and frequent supplementation of recombinant growth factors due to their short half-life at physiological temperatures. Here, we developed a biomaterial strategy using mineral-coated microparticles (MCMs) to sustain delivery of basic fibroblast growth factor (bFGF), a thermolabile protein critical for hPSC pluripotency and proliferation. We show that the MCMs stabilize bFGF against thermally induced activity loss and provide more efficient sustained release of active growth factor compared to polymeric carriers commonly used for growth factor delivery. Using a statistically driven optimization approach called Design of Experiments, we generated a bFGF-loaded MCM formulation that supported hPSC expansion over 25 passages without the need for additional bFGF supplementation to the media, resulting in greater than 80% reduction in bFGF usage compared to standard approaches. This materials-based strategy to stabilize and sustain delivery of a thermolabile growth factor has broad potential to reduce costs associated with recombinant protein supplements in scalable biomanufacturing of emerging cell therapies.
Collapse
Affiliation(s)
- Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Hunter J Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
11
|
Ireland RG, Kibschull M, Audet J, Ezzo M, Hinz B, Lye SJ, Simmons CA. Combinatorial extracellular matrix microarray identifies novel bioengineered substrates for xeno-free culture of human pluripotent stem cells. Biomaterials 2020; 248:120017. [PMID: 32283392 DOI: 10.1016/j.biomaterials.2020.120017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/09/2020] [Accepted: 03/27/2020] [Indexed: 11/24/2022]
Abstract
Stem cells in their microenvironment are exposed to a plethora of biochemical signals and biophysical forces. Interrogating the role of each factor in the cell microenvironment, however, remains difficult due to the inability to study microenvironmental cues and tease apart their interactions in high throughput. To address this need, we developed an extracellular matrix (ECM) microarray screening platform capable of tightly controlling substrate stiffness and ECM protein composition to screen the effects of these cues and their interactions on cell fate. We combined this platform with a design of experiments screening strategy to identify optimal conditions that can maintain human pluripotent stem cell (hPSC) pluripotency in chemically defined, xeno-free conditions. Combinations of ECM proteins (fibronectin, vitronectin, laminin-521, and collagen IV) were deposited on polydimethylsiloxane substrates with elastic moduli ranging from ~1 to 60 kPa using a high throughput protein plotter. Through our screening approach, we identified several non-intuitive protein-protein and protein-stiffness interactions and developed three novel culture substrates. hPSCs grown on these novel culture substrates displayed higher proliferation rates and pluripotency marker expression than current gold-standard culture substrates Geltrex- and vitronectin-coated plastic. This ECM microarray and screening approach is not limited to the factors studied here and can be broadly applied to other cell types to systematically screen microenvironmental conditions to optimally guide cell phenotype.
Collapse
Affiliation(s)
- Ronald G Ireland
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Mark Kibschull
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Julie Audet
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Maya Ezzo
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Boris Hinz
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Stephen J Lye
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Departments of Obstetrics & Gynaecology, Physiology, and Medicine, University of Toronto, Toronto, ON, Canada; Alliance for Human Development, Sinai Health System, Toronto, ON, Canada
| | - Craig A Simmons
- Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Kupfer ME, Lin WH, Ravikumar V, Qiu K, Wang L, Gao L, Bhuiyan DB, Lenz M, Ai J, Mahutga RR, Townsend D, Zhang J, McAlpine MC, Tolkacheva EG, Ogle BM. In Situ Expansion, Differentiation, and Electromechanical Coupling of Human Cardiac Muscle in a 3D Bioprinted, Chambered Organoid. Circ Res 2020; 127:207-224. [PMID: 32228120 DOI: 10.1161/circresaha.119.316155] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
RATIONALE One goal of cardiac tissue engineering is the generation of a living, human pump in vitro that could replace animal models and eventually serve as an in vivo therapeutic. Models that replicate the geometrically complex structure of the heart, harboring chambers and large vessels with soft biomaterials, can be achieved using 3-dimensional bioprinting. Yet, inclusion of contiguous, living muscle to support pump function has not been achieved. This is largely due to the challenge of attaining high densities of cardiomyocytes-a notoriously nonproliferative cell type. An alternative strategy is to print with human induced pluripotent stem cells, which can proliferate to high densities and fill tissue spaces, and subsequently differentiate them into cardiomyocytes in situ. OBJECTIVE To develop a bioink capable of promoting human induced pluripotent stem cell proliferation and cardiomyocyte differentiation to 3-dimensionally print electromechanically functional, chambered organoids composed of contiguous cardiac muscle. METHODS AND RESULTS We optimized a photo-crosslinkable formulation of native ECM (extracellular matrix) proteins and used this bioink to 3-dimensionally print human induced pluripotent stem cell-laden structures with 2 chambers and a vessel inlet and outlet. After human induced pluripotent stem cells proliferated to a sufficient density, we differentiated the cells within the structure and demonstrated function of the resultant human chambered muscle pump. Human chambered muscle pumps demonstrated macroscale beating and continuous action potential propagation with responsiveness to drugs and pacing. The connected chambers allowed for perfusion and enabled replication of pressure/volume relationships fundamental to the study of heart function and remodeling with health and disease. CONCLUSIONS This advance represents a critical step toward generating macroscale tissues, akin to aggregate-based organoids, but with the critical advantage of harboring geometric structures essential to the pump function of cardiac muscle. Looking forward, human chambered organoids of this type might also serve as a test bed for cardiac medical devices and eventually lead to therapeutic tissue grafting.
Collapse
Affiliation(s)
- Molly E Kupfer
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Stem Cell Institute (M.E.K., W.-H.L., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Wei-Han Lin
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Stem Cell Institute (M.E.K., W.-H.L., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Vasanth Ravikumar
- Department of Electrical Engineering (V.R.), University of Minnesota-Twin Cities, Minneapolis
| | - Kaiyan Qiu
- Department of Mechanical Engineering (K.Q., M.C.M.), University of Minnesota-Twin Cities, Minneapolis
| | - Lu Wang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.W., L.G., J.Z.)
| | - Ling Gao
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.W., L.G., J.Z.)
| | - Didarul B Bhuiyan
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Megan Lenz
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Jeffrey Ai
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Ryan R Mahutga
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - DeWayne Townsend
- Lillehei Heart Institute (D.T., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Department of Integrative Biology and Physiology (D.T.), University of Minnesota-Twin Cities, Minneapolis
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.W., L.G., J.Z.)
| | - Michael C McAlpine
- Department of Mechanical Engineering (K.Q., M.C.M.), University of Minnesota-Twin Cities, Minneapolis
| | - Elena G Tolkacheva
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Lillehei Heart Institute (D.T., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Institute for Engineering in Medicine (E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Brenda M Ogle
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Stem Cell Institute (M.E.K., W.-H.L., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Lillehei Heart Institute (D.T., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Institute for Engineering in Medicine (E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Masonic Cancer Center (B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| |
Collapse
|
13
|
Pang X, O'Malley C, Borges J, Rahman MM, Collis DWP, Mano JF, Mackenzie IC, S. Azevedo H. Supramolecular Presentation of Hyaluronan onto Model Surfaces for Studying the Behavior of Cancer Stem Cells. ACTA ACUST UNITED AC 2019; 3:e1900017. [DOI: 10.1002/adbi.201900017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/15/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Xinqing Pang
- School of Engineering and Materials ScienceInstitute of BioengineeringQueen Mary University of London E1 4NS UK
| | - Clare O'Malley
- School of Engineering and Materials ScienceInstitute of BioengineeringQueen Mary University of London E1 4NS UK
| | - João Borges
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of Aveiro 3810‐193 Aveiro Portugal
| | - Muhammad M. Rahman
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University of London E1 2AT UK
| | - Dominic W. P. Collis
- School of Engineering and Materials ScienceInstitute of BioengineeringQueen Mary University of London E1 4NS UK
| | - João F. Mano
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of Aveiro 3810‐193 Aveiro Portugal
| | - Ian C. Mackenzie
- Blizard InstituteBarts and The London School of Medicine and DentistryQueen Mary University of London E1 2AT UK
| | - Helena S. Azevedo
- School of Engineering and Materials ScienceInstitute of BioengineeringQueen Mary University of London E1 4NS UK
| |
Collapse
|
14
|
Cao B, Li Y, Yang T, Bao Q, Yang M, Mao C. Bacteriophage-based biomaterials for tissue regeneration. Adv Drug Deliv Rev 2019; 145:73-95. [PMID: 30452949 PMCID: PMC6522342 DOI: 10.1016/j.addr.2018.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 07/24/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
Abstract
Bacteriophage, also called phage, is a human-safe bacteria-specific virus. It is a monodisperse biological nanostructure made of proteins (forming the outside surface) and nucleic acids (encased in the protein capsid). Among different types of phages, filamentous phages have received great attention in tissue regeneration research due to their unique nanofiber-like morphology. They can be produced in an error-free format, self-assemble into ordered scaffolds, display multiple signaling peptides site-specifically, and serve as a platform for identifying novel signaling or homing peptides. They can direct stem cell differentiation into specific cell types when they are organized into proper patterns or display suitable peptides. These unusual features have allowed scientists to employ them to regenerate a variety of tissues, including bone, nerves, cartilage, skin, and heart. This review will summarize the progress in the field of phage-based tissue regeneration and the future directions in this field.
Collapse
Affiliation(s)
- Binrui Cao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States
| | - Yan Li
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States
| | - Tao Yang
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Qing Bao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Zhejiang, Hangzhou 310058, China.
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019, United States; School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China.
| |
Collapse
|
15
|
Musah S, Dimitrakakis N, Camacho DM, Church GM, Ingber DE. Directed differentiation of human induced pluripotent stem cells into mature kidney podocytes and establishment of a Glomerulus Chip. Nat Protoc 2019; 13:1662-1685. [PMID: 29995874 DOI: 10.1038/s41596-018-0007-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Protocols have been established to direct the differentiation of human induced pluripotent stem (iPS) cells into nephron progenitor cells and organoids containing many types of kidney cells, but it has been difficult to direct the differentiation of iPS cells to form specific types of mature human kidney cells with high yield. Here, we describe a detailed protocol for the directed differentiation of human iPS cells into mature, post-mitotic kidney glomerular podocytes with high (>90%) efficiency within 26 d and under chemically defined conditions, without genetic manipulations or subpopulation selection. We also describe how these iPS cell-derived podocytes may be induced to form within a microfluidic organ-on-a-chip (Organ Chip) culture device to build a human kidney Glomerulus Chip that mimics the structure and function of the kidney glomerular capillary wall in vitro within 35 d (starting with undifferentiated iPS cells). The podocyte differentiation protocol requires skills for culturing iPS cells, and the development of a Glomerulus Chip requires some experience with building and operating microfluidic cell culture systems. This method could be useful for applications in nephrotoxicity screening, therapeutic development, and regenerative medicine, as well as mechanistic study of kidney development and disease.
Collapse
Affiliation(s)
- Samira Musah
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Nikolaos Dimitrakakis
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA. .,Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
16
|
Hall ML, Ogle BM. Cardiac Extracellular Matrix Modification as a Therapeutic Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1098:131-150. [PMID: 30238369 PMCID: PMC6584040 DOI: 10.1007/978-3-319-97421-7_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The cardiac extracellular matrix (cECM) is comprised of proteins and polysaccharides secreted by cardiac cell types, which provide structural and biochemical support to cardiovascular tissue. The roles of cECM proteins and the associated family of cell surface receptor, integrins, have been explored in vivo via the generation of knockout experimental animal models. However, the complexity of tissues makes it difficult to isolate the effects of individual cECM proteins on a particular cell process or disease state. The desire to further dissect the role of cECM has led to the development of a variety of in vitro model systems, which are now being used not only for basic studies but also for testing drug efficacy and toxicity and for generating therapeutic scaffolds. These systems began with 2D coatings of cECM derived from tissue and have developed to include recombinant ECM proteins, ECM fragments, and ECM mimics. Most recently 3D model systems have emerged, made possible by several developing technologies including, and most notably, 3D bioprinting. This chapter will attempt to track the evolution of our understanding of the relationship between cECM and cell behavior from in vivo model to in vitro control systems. We end the chapter with a summary of how basic studies such as these have informed the use of cECM as a direct therapy.
Collapse
Affiliation(s)
- Mikayla L Hall
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Lillehei Heart Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Institute for Engineering in Medicine, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
17
|
Pang X, Li W, Landwehr E, Yuan Y, Wang W, Azevedo HS. Mimicking the endothelial glycocalyx through the supramolecular presentation of hyaluronan on patterned surfaces. Faraday Discuss 2019; 219:168-182. [DOI: 10.1039/c9fd00015a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Self-assembled monolayers of hyaluronan (HA)-binding peptide allow immobilization of HA for studying the function of the endothelial glycocalyx.
Collapse
Affiliation(s)
- Xinqing Pang
- School of Engineering and Materials Science
- Queen Mary University of London
- London E1 4NS
- UK
- Institute of Bioengineering
| | - Weiqi Li
- School of Engineering and Materials Science
- Queen Mary University of London
- London E1 4NS
- UK
- Institute of Bioengineering
| | - Eliane Landwehr
- Department of Chemistry
- University of Konstanz
- Konstanz 78464
- Germany
| | - Yichen Yuan
- School of Engineering and Materials Science
- Queen Mary University of London
- London E1 4NS
- UK
- Institute of Bioengineering
| | - Wen Wang
- School of Engineering and Materials Science
- Queen Mary University of London
- London E1 4NS
- UK
- Institute of Bioengineering
| | - Helena S. Azevedo
- School of Engineering and Materials Science
- Queen Mary University of London
- London E1 4NS
- UK
- Institute of Bioengineering
| |
Collapse
|
18
|
Corbin BA, Basal LA, White SA, Shen Y, Haacke EM, Fishbein KW, Allen MJ. Screening of ligands for redox-active europium using magnetic resonance imaging. Bioorg Med Chem 2018; 26:5274-5279. [PMID: 29653832 DOI: 10.1016/j.bmc.2018.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/27/2018] [Accepted: 04/01/2018] [Indexed: 12/31/2022]
Abstract
We report a screening procedure to predict ligand coordination to EuII and EuIII using magnetic resonance imaging in which bright images indicate complexation and dark images indicate no complexation. Here, paramagnetic GdIII is used as a surrogate for EuIII in the screening procedure to enable detection with magnetic resonance imaging. The screening procedure was tested using a set of eight ligands with known coordination to EuII and EuIII, and results were found to be consistent with expected binding. Validation of the screening procedure with known coordination chemistry enables use with new ligands in the future.
Collapse
Affiliation(s)
- Brooke A Corbin
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, United States
| | - Lina A Basal
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, United States
| | - Susan A White
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, United States
| | - Yimin Shen
- Department of Radiology, Wayne State University School of Medicine, Detroit, MI 48201, United States
| | - E Mark Haacke
- Department of Radiology, Wayne State University School of Medicine, Detroit, MI 48201, United States; Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, United States
| | - Kenneth W Fishbein
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, United States
| | - Matthew J Allen
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, United States; Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, United States.
| |
Collapse
|
19
|
Richardson TC, Mathew S, Candiello JE, Goh SK, Kumta PN, Banerjee I. Development of an Alginate Array Platform to Decouple the Effect of Multiparametric Perturbations on Human Pluripotent Stem Cells During Pancreatic Differentiation. Biotechnol J 2018; 13. [DOI: 10.1002/biot.201700099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/09/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Thomas C. Richardson
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| | - Shibin Mathew
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| | | | - Saik K. Goh
- Department of Bioengineering; University of Pittsburgh; Pittsburgh USA
| | - Prashant N. Kumta
- Department of Bioengineering, Chemical and Petroleum Engineering; Mechanical Engineering and Materials Science; University of Pittsburgh; Pittsburgh USA
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering; University of Pittsburgh; Pittsburgh USA
| |
Collapse
|
20
|
Long-Term Maintenance of Human Pluripotent Stem Cells on cRGDfK-Presenting Synthetic Surfaces. Sci Rep 2018; 8:701. [PMID: 29335618 PMCID: PMC5768753 DOI: 10.1038/s41598-018-19209-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
Synthetic human pluripotent stem cell (hPSC) culture surfaces with defined physical and chemical properties will facilitate improved research and therapeutic applications of hPSCs. In this study, synthetic surfaces for hPSC culture in E8 medium were produced for screening by modifying two polymer brush coatings [poly(acrylamide-co-acrylic acid) (PAAA) and poly(acrylamide-co-propargyl acrylamide) (PAPA)] to present single peptides. Adhesion of hPSC colonies was more consistently observed on surfaces modified with cRGDfK compared to surfaces modified with other peptide sequences tested. PAPA-coated polystyrene flasks with coupled cRGDfK (cRGDfK-PAPA) were then used for long-term studies of three hPSC lines (H9, hiPS-NHF1.3, Genea-02). Cell lines maintained for ten passages on cRGDfK-PAPA were assessed for colony morphology, proliferation rate, maintenance of OCT4 expression, cell viability at harvest, teratoma formation potential, and global gene expression as assessed by the PluriTest™ assay. cRGDfK-PAPA and control cultures maintained on Geltrex™ produced comparable results in most assays. No karyotypic abnormalities were detected in cultures maintained on cRGDfK-PAPA, while abnormalities were detected in cultures maintained on Geltrex™, StemAdhere™ or Synthemax™. This is the first report of long term maintenance of hPSC cultures on the scalable, stable, and cost-effective cRGDfK-PAPA coating.
Collapse
|
21
|
Chen X, Harkness L, Jia Z, Prowse A, Monteiro MJ, Gray PP. Methods for Expansion of Three-Dimensional Cultures of Human Embryonic Stem Cells Using a Thermoresponsive Polymer. Tissue Eng Part C Methods 2017; 24:146-157. [PMID: 29239281 DOI: 10.1089/ten.tec.2017.0331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are viewed as promising candidates for applications in regenerative medicine and therapy due to their proliferative and pluripotent properties. However, obtaining clinically significant numbers of hPSCs remains a limiting factor and impedes their use in therapeutic applications. Conventionally, hPSCs are cultured on two-dimensional surfaces coated with a suitable substrate, such as Matrigel™. This method, however, requires a large surface area to generate sufficient cell numbers to meet clinical needs and is therefore impractical as a manufacturing platform for cell expansion. In addition, the use of enzymes for cell detachment and small molecule inhibitors to increase plating efficiency may impact future cell behavior when used for routine subculturing. In this study, we describe a protocol to generate and maintain hPSC aggregates in a three-dimensional suspension culture by utilizing thermoresponsive nanobridges. The property of the polymer used in the nanobridges enables passaging and expansion through a temperature change in combination with mechanically applied shear to dissociate aggregates; thus, we eliminate the need of enzymes or small molecules for cell dissociation and viability, respectively. Utilizing this platform, maintenance of human embryonic stem cells for three continuous passages demonstrated high expression levels in key pluripotent markers.
Collapse
Affiliation(s)
- Xiaoli Chen
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Linda Harkness
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Zhongfan Jia
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Andrew Prowse
- 2 The Garvan Institute of Medical Research , Sydney, Australia
| | - Michael J Monteiro
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| | - Peter P Gray
- 1 Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland , Brisbane, Australia
| |
Collapse
|
22
|
Zhang D, Lee J, Kilian KA. Synthetic Biomaterials to Rival Nature's Complexity-a Path Forward with Combinatorics, High-Throughput Discovery, and High-Content Analysis. Adv Healthc Mater 2017; 6. [PMID: 28841770 DOI: 10.1002/adhm.201700535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/08/2017] [Indexed: 12/18/2022]
Abstract
Cells in tissue receive a host of soluble and insoluble signals in a context-dependent fashion, where integration of these cues through a complex network of signal transduction cascades will define a particular outcome. Biomaterials scientists and engineers are tasked with designing materials that can at least partially recreate this complex signaling milieu towards new materials for biomedical applications. In this progress report, recent advances in high throughput techniques and high content imaging approaches that are facilitating the discovery of efficacious biomaterials are described. From microarrays of synthetic polymers, peptides and full-length proteins, to designer cell culture systems that present multiple biophysical and biochemical cues in tandem, it is discussed how the integration of combinatorics with high content imaging and analysis is essential to extracting biologically meaningful information from large scale cellular screens to inform the design of next generation biomaterials.
Collapse
Affiliation(s)
- Douglas Zhang
- Department of Materials Science and Engineering; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
| | - Junmin Lee
- Department of Materials Science and Engineering; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
| | - Kristopher A. Kilian
- Department of Materials Science and Engineering; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
- Department of Bioengineering; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
| |
Collapse
|
23
|
Efficient feeder cells preparation system for large-scale preparation and application of induced pluripotent stem cells. Sci Rep 2017; 7:12266. [PMID: 28947775 PMCID: PMC5612988 DOI: 10.1038/s41598-017-10428-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/09/2017] [Indexed: 02/03/2023] Open
Abstract
Despite recent progress in the preparation of feeder cells for human induced pluripotent stem cells (hiPSCs), there remain issues which limit the acquisition of feeder cells in large scale. Approaches for obtaining feeder cells quickly on a large scale are in immediate need. To reach this goal, we established suspension-adhesion method (SAM) and three-dimensional (3D) suspension method (3DSM). In SAM, mouse embryonic fibroblast (MEF) growth were fully inhibited by 10 μg/ml mitomycin-C (MMC) in 0.5 hours, and the feeder cells generated display higher adherent and recovery rates as well as longer survival time compared to conventional method (CM). 3DSM, an optimized method of SAM in which MEFs were cultured and MMC treated in suspension, was developed to lower the costs and workload using CELLSPIN System. The yield of feeder cells is several times the yield of SAM while the adherent and recovery rates and the capacity of supporting hiPSCs growth were not sacrificed. Hence, 3DSM is an economical and easy way to generate large-scale feeder cells for hiPSCs cultures.
Collapse
|
24
|
Zade HM, Keshavarz R, Shekarabi HSZ, Bakhshinejad B. Biased selection of propagation-related TUPs from phage display peptide libraries. Amino Acids 2017; 49:1293-1308. [DOI: 10.1007/s00726-017-2452-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/09/2017] [Indexed: 10/19/2022]
|
25
|
Zhang D, Lee J, Sun MB, Pei Y, Chu J, Gillette MU, Fan TM, Kilian KA. Combinatorial Discovery of Defined Substrates That Promote a Stem Cell State in Malignant Melanoma. ACS CENTRAL SCIENCE 2017; 3:381-393. [PMID: 28573199 PMCID: PMC5445527 DOI: 10.1021/acscentsci.6b00329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Indexed: 06/07/2023]
Abstract
The tumor microenvironment is implicated in orchestrating cancer cell transformation and metastasis. However, specific cell-ligand interactions between cancer cells and the extracellular matrix are difficult to decipher due to a dynamic and multivariate presentation of many signaling molecules. Here we report a versatile peptide microarray platform that is capable of screening for cancer cell phenotypic changes in response to ligand-receptor interactions. Using a screen of 78 peptide combinations derived from proteins present in the melanoma microenvironment, we identify a proteoglycan binding and bone morphogenic protein 7 (BMP7) derived sequence that selectively promotes the expression of several putative melanoma initiating cell markers. We characterize signaling associated with each of these peptides in the activation of melanoma pro-tumorigenic signaling and reveal a role for proteoglycan mediated adhesion and signaling through Smad 2/3. A defined substratum that controls the state of malignant melanoma may prove useful in spatially normalizing a heterogeneous population of tumor cells for discovery of therapeutics that target a specific state and for identifying new drug targets and reagents for intervention.
Collapse
Affiliation(s)
- Douglas Zhang
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Junmin Lee
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Michael B. Sun
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Yi Pei
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - James Chu
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Martha U. Gillette
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Kristopher A. Kilian
- Department of Materials Science and Engineering, Department of Cell and Developmental
Biology, Department
of Veterinary Clinical Medicine, and Department of Bioengineering, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
26
|
Screening Mammalian Cells on a Hydrogel: Functionalized Small Molecule Microarray. Methods Mol Biol 2016. [PMID: 27873211 DOI: 10.1007/978-1-4939-6584-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Mammalian cell-based microarray technology has gained wide attention, for its plethora of promising applications. The platform is able to provide simultaneous information on multiple parameters for a given target, or even multiple target proteins, in a complex biological system. Here we describe the preparation of mammalian cell-based microarrays using selectively captured of human prostate cancer cells (PC-3). This platform was then used in controlled drug release and measuring the associated drug effects on these cancer cells.
Collapse
|
27
|
Martins IM, Reis RL, Azevedo HS. Phage Display Technology in Biomaterials Engineering: Progress and Opportunities for Applications in Regenerative Medicine. ACS Chem Biol 2016; 11:2962-2980. [PMID: 27661443 DOI: 10.1021/acschembio.5b00717] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The field of regenerative medicine has been gaining momentum steadily over the past few years. The emphasis in regenerative medicine is to use various in vitro and in vivo approaches that leverage the intrinsic healing mechanisms of the body to treat patients with disabling injuries and chronic diseases such as diabetes, osteoarthritis, and degenerative disorders of the cardiovascular and central nervous system. Phage display has been successfully employed to identify peptide ligands for a wide variety of targets, ranging from relatively small molecules (enzymes, cell receptors) to inorganic, organic, and biological (tissues) materials. Over the past two decades, phage display technology has advanced tremendously and has become a powerful tool in the most varied fields of research, including biotechnology, materials science, cell biology, pharmacology, and diagnostics. The growing interest in and success of phage display libraries is largely due to its incredible versatility and practical use. This review discusses the potential of phage display technology in biomaterials engineering for applications in regenerative medicine.
Collapse
Affiliation(s)
- Ivone M. Martins
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- CEB − Centre of Biological Engineering, University of Minho, 4710-057, Braga, Portugal
| | - Rui L. Reis
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helena S. Azevedo
- 3B’s Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of
the European Institute of Excellence on Tissue Engineering and Regenerative
Medicine, AvePark, 4805-717 Barco, Guimarães, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, United Kingdom
- Institute
of Bioengineering, Queen Mary University of London, London E1 4NS, United Kingdom
| |
Collapse
|
28
|
Rosch JC, Hollmann EK, Lippmann ES. In vitro selection technologies to enhance biomaterial functionality. Exp Biol Med (Maywood) 2016; 241:962-71. [PMID: 27188514 DOI: 10.1177/1535370216647182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cells make decisions and fate choices based in part on cues they receive from their external environment. Factors that affect the interpretation of these cues include the soluble proteins that are present at any given time, the cell surface receptors that are available to bind these proteins, and the relative affinities of the soluble proteins for their cognate receptors. Researchers have identified many of the biological motifs responsible for the high-affinity interactions between proteins and their receptors, and subsequently incorporated these motifs into biomaterials to elicit control over cell behavior. Common modes of control include localized sequestration of proteins to improve bioavailability and direct inhibition or activation of a receptor by an immobilized peptide or protein. However, naturally occurring biological motifs often possess promiscuous affinity for multiple proteins and receptors or lack programmable actuation in response to dynamic stimuli, thereby limiting the amount of control they can exert over cellular decisions. These natural motifs only represent a small fraction of the biological diversity that can be assayed by in vitro selection strategies, and the discovery of "artificial" motifs with varying affinity, specificity, and functionality could greatly expand the repertoire of engineered biomaterial properties. This minireview provides a brief summary of classical and emerging techniques in peptide phage display and nucleic acid aptamer selections and discusses prospective applications in the areas of cell adhesion, angiogenesis, neural regeneration, and immune modulation.
Collapse
Affiliation(s)
- Jonah C Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Emma K Hollmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
29
|
Kim HD, Lee EA, Choi YH, An YH, Koh RH, Kim SL, Hwang NS. High throughput approaches for controlled stem cell differentiation. Acta Biomater 2016; 34:21-29. [PMID: 26884279 DOI: 10.1016/j.actbio.2016.02.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 02/13/2016] [Accepted: 02/13/2016] [Indexed: 12/19/2022]
Abstract
Stem cells have unique ability to undergo self-renewal indefinitely in culture and potential to differentiate into almost all cell types in the human body. However, the developing a method for efficiently differentiating or manipulating these stem cells for therapeutic purposes remains a challenging problem. Pluripotent stem cells, as well as adult stem cells, require biological cues for their proliferation and differentiation. These cues are largely controlled by cell-cell, cell-insoluble factors (such as extracellular matrix), and cell-soluble factors (such as cytokine or growth factors) interactions. In this review, we describe a state of research on various stem cell-based tissue engineering applications and high throughput strategies for developing synthetic or biosynthetic microenvironments to allow efficient commitments in stem cells. STATEMENT OF SIGNIFICANCE Nowadays, pluripotency of stem cells have received much attention to use therapeutic purpose. However, a major difficulty with stem cell therapy is to control its differentiation through desired cells or tissues. In other words, various microenvironment factors are involved during stem cell differentiation, including dimensionality, growth factors, cell junctions, nutritional status, matrix stiffness, matrix composition, mechanical stress, and cell-matrix adhesion. Therefore, researchers have engineered a variety of platforms to enable controlling and monitoring bioactive factors to induce stem cell commitment. In this review, we report on recent advancements in a novel technology based on high-throughput strategies for stem cell-based tissue engineering applications.
Collapse
|
30
|
Lin E, Sikand A, Wickware J, Hao Y, Derda R. Peptide microarray patterning for controlling and monitoring cell growth. Acta Biomater 2016; 34:53-59. [PMID: 26805426 DOI: 10.1016/j.actbio.2016.01.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 01/09/2016] [Accepted: 01/20/2016] [Indexed: 02/08/2023]
Abstract
The fate of cells is influenced by their microenvironment and many cell types undergo differentiation when stimulated by extracellular cues, such as soluble growth factors and the insoluble extracellular matrix (ECM). Stimulating differentiation by insoluble or "immobilized" cues is a particularly attractive method because it allows for the induction of differentiation in a spatially-defined cohort of cells within a larger subpopulation. To improve the design of de novo screening of such insoluble factors, we describe a methodology for producing high-density peptide microarrays suitable for extended cell culture and fluorescence microscopy. As a model, we used a murine mammary gland cell line (NMuMG) that undergoes epithelial to mesenchymal transition (EMT) in response to soluble transforming growth factor beta (TGF-β) and surface-immobilized peptides that target TGF-β receptors (TGFβRI/II). We repurposed a well-established DNA microarray printing technique to produce arrays of micropatterned surfaces that displayed TGFβRI/II-binding peptides and integrin binding peptides. Upon long-term culture on these arrays, only NMuMG cells residing on EMT-stimulating areas exhibited growth arrest and decreased E-cadherin expression. We believe that the methodology created in this report will aid the development of peptide-decorated surfaces that can locally stimulate defined cell surface receptors and control EMT and other well-characterized differentiation events. STATEMENT OF SIGNIFICANCE Scope of work: This manuscript aims to accelerate the development of instructive biomaterials decorated with specific ligands that target cell-surface receptors and induce specific differentiation of cells upon contact. These materials can be used for practical applications, such as fabricating synthetic materials for large scale, stem cell culture, or investigating differentiation and asymmetric division in stem cells. Specifically, in this manuscript, we repurposed a DNA microarray printer to produce microarrays of peptide-terminated self-assembled monolayers (SAMs). To demonstrate the utility of these arrays in phenotypic assays with mammalian cells, we monitored the induction of epithelial to mesenchymal transition (EMT) in murine mammary epithelial cells using specific peptide ligands printed on these arrays. Novelty: We, and others, have published several strategies for producing peptide-based arrays suitable for long-term phenotypic assays. Many reports relied on patterning steps that made adaptation difficult. The use of a DNA microarray printer as the sole production tool simplified the production of peptide microarrays and increased the throughput of this technology. We confirmed that simplification in production did not compromise the performance of the array; it is still possible to study short-term adhesion, long-term growth, and complex phenotypic responses, such as EMT, in the cells. EMT was studied using immunofluorescent staining after four days of culture. IMPACT This methodology will serve as a foundation for future screening of instructive biomaterials in our research group. As DNA printers are broadly available in academic institutions, we foresee rapid adaptation of this approach by academic researchers.
Collapse
|
31
|
Ivanovska IL, Shin JW, Swift J, Discher DE. Stem cell mechanobiology: diverse lessons from bone marrow. Trends Cell Biol 2015; 25:523-32. [PMID: 26045259 PMCID: PMC4555184 DOI: 10.1016/j.tcb.2015.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 12/19/2022]
Abstract
A stem cell niche is defined by various chemical and physical features that influence whether a stem cell remains quiescent, divides, or differentiates. We review mechanical determinants that affect cell fate through actomyosin forces, nucleoskeleton remodeling, and mechanosensitive translocation of transcription factors. Current methods for physical characterization of tissue microenvironments are summarized together with efforts to recapitulate niche mechanics in culture. We focus on mesenchymal stem cells, particularly in osteogenesis and adipogenesis, and on blood stem cells - both of which reside in mechanically diverse marrow microenvironments. Given the explosion of efforts with pluripotent stem cells, the evident mechanosensitivity of clinically relevant, multipotent marrow cells underscores an increasing need to examine and understand in vivo and in vitro physical properties on length scales that cells sense.
Collapse
Affiliation(s)
- Irena L Ivanovska
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jae-Won Shin
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joe Swift
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Lou YR, Kanninen L, Kaehr B, Townson JL, Niklander J, Harjumäki R, Jeffrey Brinker C, Yliperttula M. Silica bioreplication preserves three-dimensional spheroid structures of human pluripotent stem cells and HepG2 cells. Sci Rep 2015; 5:13635. [PMID: 26323570 PMCID: PMC4555166 DOI: 10.1038/srep13635] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/30/2015] [Indexed: 11/19/2022] Open
Abstract
Three-dimensional (3D) cell cultures produce more in vivo-like multicellular structures such as spheroids that cannot be obtained in two-dimensional (2D) cell cultures. Thus, they are increasingly employed as models for cancer and drug research, as well as tissue engineering. It has proven challenging to stabilize spheroid architectures for detailed morphological examination. Here we overcome this issue using a silica bioreplication (SBR) process employed on spheroids formed from human pluripotent stem cells (hPSCs) and hepatocellular carcinoma HepG2 cells cultured in the nanofibrillar cellulose (NFC) hydrogel. The cells in the spheroids are more round and tightly interacting with each other than those in 2D cultures, and they develop microvilli-like structures on the cell membranes as seen in 2D cultures. Furthermore, SBR preserves extracellular matrix-like materials and cellular proteins. These findings provide the first evidence of intact hPSC spheroid architectures and similar fine structures to 2D-cultured cells, providing a pathway to enable our understanding of morphogenesis in 3D cultures.
Collapse
Affiliation(s)
- Yan-Ru Lou
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, the University of Helsinki, Helsinki 00014, Finland
| | - Liisa Kanninen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, the University of Helsinki, Helsinki 00014, Finland
| | - Bryan Kaehr
- Advanced Materials Laboratory, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA.,Department of Chemical and Biomolecular Engineering, the University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Jason L Townson
- Division of Molecular Medicine, Department of Internal Medicine, the University of New Mexico, Albuquerque, New Mexico 87131, USA.,Center for Micro-Engineered Materials, the University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Johanna Niklander
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, the University of Helsinki, Helsinki 00014, Finland
| | - Riina Harjumäki
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, the University of Helsinki, Helsinki 00014, Finland
| | - C Jeffrey Brinker
- Advanced Materials Laboratory, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA.,Department of Chemical and Biomolecular Engineering, the University of New Mexico, Albuquerque, New Mexico 87131, USA
| | - Marjo Yliperttula
- Centre for Drug Research, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, the University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
33
|
Nagaoka M, Kobayashi M, Kawai C, Mallanna SK, Duncan SA. Design of a Vitronectin-Based Recombinant Protein as a Defined Substrate for Differentiation of Human Pluripotent Stem Cells into Hepatocyte-Like Cells. PLoS One 2015; 10:e0136350. [PMID: 26308339 PMCID: PMC4550348 DOI: 10.1371/journal.pone.0136350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/01/2015] [Indexed: 01/05/2023] Open
Abstract
Maintenance and differentiation of human pluripotent stem cells (hPSCs) usually requires culture on a substrate for cell adhesion. A commonly used substratum is Matrigel purified from Engelbreth—Holm—Swarm sarcoma cells, and consists of a complex mixture of extracellular matrix proteins, proteoglycans, and growth factors. Several studies have successfully induced differentiation of hepatocyte-like cells from hPSCs. However, most of these studies have used Matrigel as a cell adhesion substrate, which is not a defined culture condition. In an attempt to generate a substratum that supports undifferentiated properties and differentiation into hepatic lineage cells, we designed novel substrates consisting of vitronectin fragments fused to the IgG Fc domain. hPSCs adhered to these substrates via interactions between integrins and the RGD (Arg-Gly-Asp) motif, and the cells maintained their undifferentiated phenotypes. Using a previously established differentiation protocol, hPSCs were efficiently differentiated into mesendodermal and hepatic lineage cells on a vitronectin fragment-containing substrate. We found that full-length vitronectin did not support stable cell adhesion during the specification stage. Furthermore, the vitronectin fragment with the minimal RGD-containing domain was sufficient for differentiation of human induced pluripotent stem cells into hepatic lineage cells under completely defined conditions that facilitate the clinical application of cells differentiated from hPSCs.
Collapse
Affiliation(s)
- Masato Nagaoka
- Tenure-track Program for Innovative Research, University of Fukui, Yoshida-gun, Fukui, Japan
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| | - Motohiro Kobayashi
- Division of Tumor Pathology, Department of Pathological Sciences, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui, Japan
| | - Chie Kawai
- Tenure-track Program for Innovative Research, University of Fukui, Yoshida-gun, Fukui, Japan
| | - Sunil K. Mallanna
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Stephen A. Duncan
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
34
|
Zhu B, Jiang B, Na Z, Yao SQ. Controlled proliferation and screening of mammalian cells on a hydrogel-functionalized small molecule microarray. Chem Commun (Camb) 2015; 51:10431-4. [PMID: 26028192 DOI: 10.1039/c5cc03278d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A hydrogel-functionalized small molecule microarray has been developed, on which PC-3 cancer cells were selectively grown. Subsequent controlled release of immobilized bioactive compounds enabled cell-based screening to be directly carried out on this platform.
Collapse
Affiliation(s)
- Biwei Zhu
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore117543.
| | | | | | | |
Collapse
|
35
|
Benam KH, Dauth S, Hassell B, Herland A, Jain A, Jang KJ, Karalis K, Kim HJ, MacQueen L, Mahmoodian R, Musah S, Torisawa YS, van der Meer AD, Villenave R, Yadid M, Parker KK, Ingber DE. Engineered in vitro disease models. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2015; 10:195-262. [PMID: 25621660 DOI: 10.1146/annurev-pathol-012414-040418] [Citation(s) in RCA: 380] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ultimate goal of most biomedical research is to gain greater insight into mechanisms of human disease or to develop new and improved therapies or diagnostics. Although great advances have been made in terms of developing disease models in animals, such as transgenic mice, many of these models fail to faithfully recapitulate the human condition. In addition, it is difficult to identify critical cellular and molecular contributors to disease or to vary them independently in whole-animal models. This challenge has attracted the interest of engineers, who have begun to collaborate with biologists to leverage recent advances in tissue engineering and microfabrication to develop novel in vitro models of disease. As these models are synthetic systems, specific molecular factors and individual cell types, including parenchymal cells, vascular cells, and immune cells, can be varied independently while simultaneously measuring system-level responses in real time. In this article, we provide some examples of these efforts, including engineered models of diseases of the heart, lung, intestine, liver, kidney, cartilage, skin and vascular, endocrine, musculoskeletal, and nervous systems, as well as models of infectious diseases and cancer. We also describe how engineered in vitro models can be combined with human inducible pluripotent stem cells to enable new insights into a broad variety of disease mechanisms, as well as provide a test bed for screening new therapies.
Collapse
Affiliation(s)
- Kambez H Benam
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115;
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Komura T, Kato K, Konagaya S, Nakaji-Hirabayashi T, Iwata H. Optimization of surface-immobilized extracellular matrices for the proliferation of neural progenitor cells derived from induced pluripotent stem cells. Biotechnol Bioeng 2015; 112:2388-96. [DOI: 10.1002/bit.25636] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 04/17/2015] [Accepted: 04/27/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Takashi Komura
- Institute for Frontier Medical Sciences; Kyoto University; 53 Kawahara-cho, Shogoin, Sakyo-ku Kyoto 606-8507 Japan
| | - Koichi Kato
- Institute for Frontier Medical Sciences; Kyoto University; 53 Kawahara-cho, Shogoin, Sakyo-ku Kyoto 606-8507 Japan
| | - Shuhei Konagaya
- Institute for Frontier Medical Sciences; Kyoto University; 53 Kawahara-cho, Shogoin, Sakyo-ku Kyoto 606-8507 Japan
| | - Tadashi Nakaji-Hirabayashi
- Institute for Frontier Medical Sciences; Kyoto University; 53 Kawahara-cho, Shogoin, Sakyo-ku Kyoto 606-8507 Japan
| | - Hiroo Iwata
- Institute for Frontier Medical Sciences; Kyoto University; 53 Kawahara-cho, Shogoin, Sakyo-ku Kyoto 606-8507 Japan
| |
Collapse
|
37
|
Tjhung KF, Deiss F, Tran J, Chou Y, Derda R. Intra-domain phage display (ID-PhD) of peptides and protein mini-domains censored from canonical pIII phage display. Front Microbiol 2015; 6:340. [PMID: 25972845 PMCID: PMC4412080 DOI: 10.3389/fmicb.2015.00340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/06/2015] [Indexed: 12/01/2022] Open
Abstract
In this paper, we describe multivalent display of peptide and protein sequences typically censored from traditional N-terminal display on protein pIII of filamentous bacteriophage M13. Using site-directed mutagenesis of commercially available M13KE phage cloning vector, we introduced sites that permit efficient cloning using restriction enzymes between domains N1 and N2 of the pIII protein. As infectivity of phage is directly linked to the integrity of the connection between N1 and N2 domains, intra-domain phage display (ID-PhD) allows for simple quality control of the display and the natural variations in the displayed sequences. Additionally, direct linkage to phage propagation allows efficient monitoring of sequence cleavage, providing a convenient system for selection and evolution of protease-susceptible or protease-resistant sequences. As an example of the benefits of such an ID-PhD system, we displayed a negatively charged FLAG sequence, which is known to be post-translationally excised from pIII when displayed on the N-terminus, as well as positively charged sequences which suppress production of phage when displayed on the N-terminus. ID-PhD of FLAG exhibited sub-nanomolar apparent Kd suggesting multivalent nature of the display. A TEV-protease recognition sequence (TEVrs) co-expressed in tandem with FLAG, allowed us to demonstrate that 99.9997% of the phage displayed the FLAG-TEVrs tandem and can be recognized and cleaved by TEV-protease. The residual 0.0003% consisted of phage clones that have excised the insert from their genome. ID-PhD is also amenable to display of protein mini-domains, such as the 33-residue minimized Z-domain of protein A. We show that it is thus possible to use ID-PhD for multivalent display and selection of mini-domain proteins (Affibodies, scFv, etc.).
Collapse
Affiliation(s)
- Katrina F Tjhung
- Department of Chemistry, Alberta Glycomics Centre, University of Alberta Edmonton, AB, Canada
| | - Frédérique Deiss
- Department of Chemistry, Alberta Glycomics Centre, University of Alberta Edmonton, AB, Canada
| | - Jessica Tran
- Department of Chemistry, Alberta Glycomics Centre, University of Alberta Edmonton, AB, Canada
| | - Ying Chou
- Department of Chemistry, Alberta Glycomics Centre, University of Alberta Edmonton, AB, Canada
| | - Ratmir Derda
- Department of Chemistry, Alberta Glycomics Centre, University of Alberta Edmonton, AB, Canada
| |
Collapse
|
38
|
Enam S, Jin S. Substrates for clinical applicability of stem cells. World J Stem Cells 2015; 7:243-252. [PMID: 25815112 PMCID: PMC4369484 DOI: 10.4252/wjsc.v7.i2.243] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/23/2014] [Accepted: 12/19/2014] [Indexed: 02/06/2023] Open
Abstract
The capability of human pluripotent stem cells (hPSCs) to differentiate into a variety of cells in the human body holds great promise for regenerative medicine. Many substrates exist on which hPSCs can be self-renewed, maintained and expanded to further the goal of clinical application of stem cells. In this review, we highlight numerous extracellular matrix proteins, peptide and polymer based substrates, scaffolds and hydrogels that have been pioneered. We discuss their benefits and shortcomings and offer future directions as well as emphasize commercially available synthetic peptides as a type of substrate that can bring the benefits of regenerative medicine to clinical settings.
Collapse
|
39
|
Signals from the surface modulate differentiation of human pluripotent stem cells through glycosaminoglycans and integrins. Proc Natl Acad Sci U S A 2014; 111:18126-31. [PMID: 25422477 DOI: 10.1073/pnas.1409525111] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The fate decisions of human pluripotent stem (hPS) cells are governed by soluble and insoluble signals from the microenvironment. Many hPS cell differentiation protocols use Matrigel, a complex and undefined substrate that engages multiple adhesion and signaling receptors. Using defined surfaces programmed to engage specific cell-surface ligands (i.e., glycosaminoglycans and integrins), the contribution of specific matrix signals can be dissected. For ectoderm and motor neuron differentiation, peptide-modified surfaces that can engage both glycosaminoglycans and integrins are effective. In contrast, surfaces that interact selectively with glycosaminoglycans are superior to Matrigel in promoting hPS cell differentiation to definitive endoderm and mesoderm. The modular surfaces were used to elucidate the signaling pathways underlying these differences. Matrigel promotes integrin signaling, which in turn inhibits mesendoderm differentiation. The data indicate that integrin-activating surfaces stimulate Akt signaling via integrin-linked kinase (ILK), which is antagonistic to endoderm differentiation. The ability to attribute cellular responses to specific interactions between the cell and the substrate offers new opportunities for revealing and controlling the pathways governing cell fate.
Collapse
|
40
|
Substratum-induced differentiation of human pluripotent stem cells reveals the coactivator YAP is a potent regulator of neuronal specification. Proc Natl Acad Sci U S A 2014; 111:13805-10. [PMID: 25201954 DOI: 10.1073/pnas.1415330111] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Physical stimuli can act in either a synergistic or antagonistic manner to regulate cell fate decisions, but it is less clear whether insoluble signals alone can direct human pluripotent stem (hPS) cell differentiation into specialized cell types. We previously reported that stiff materials promote nuclear localization of the Yes-associated protein (YAP) transcriptional coactivator and support long-term self-renewal of hPS cells. Here, we show that even in the presence of soluble pluripotency factors, compliant substrata inhibit the nuclear localization of YAP and promote highly efficient differentiation of hPS cells into postmitotic neurons. In the absence of neurogenic factors, the effective substrata produce neurons rapidly (2 wk) and more efficiently (>75%) than conventional differentiation methods. The neurons derived from substrate induction express mature markers and possess action potentials. The hPS differentiation observed on compliant surfaces could be recapitulated on stiff surfaces by adding small-molecule inhibitors of F-actin polymerization or by depleting YAP. These studies reveal that the matrix alone can mediate differentiation of hPS cells into a mature cell type, independent of soluble inductive factors. That mechanical cues can override soluble signals suggests that their contributions to early tissue development and lineage commitment are profound.
Collapse
|
41
|
Barthes J, Özçelik H, Hindié M, Ndreu-Halili A, Hasan A, Vrana NE. Cell microenvironment engineering and monitoring for tissue engineering and regenerative medicine: the recent advances. BIOMED RESEARCH INTERNATIONAL 2014; 2014:921905. [PMID: 25143954 PMCID: PMC4124711 DOI: 10.1155/2014/921905] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/15/2014] [Indexed: 01/01/2023]
Abstract
In tissue engineering and regenerative medicine, the conditions in the immediate vicinity of the cells have a direct effect on cells' behaviour and subsequently on clinical outcomes. Physical, chemical, and biological control of cell microenvironment are of crucial importance for the ability to direct and control cell behaviour in 3-dimensional tissue engineering scaffolds spatially and temporally. In this review, we will focus on the different aspects of cell microenvironment such as surface micro-, nanotopography, extracellular matrix composition and distribution, controlled release of soluble factors, and mechanical stress/strain conditions and how these aspects and their interactions can be used to achieve a higher degree of control over cellular activities. The effect of these parameters on the cellular behaviour within tissue engineering context is discussed and how these parameters are used to develop engineered tissues is elaborated. Also, recent techniques developed for the monitoring of the cell microenvironment in vitro and in vivo are reviewed, together with recent tissue engineering applications where the control of cell microenvironment has been exploited. Cell microenvironment engineering and monitoring are crucial parts of tissue engineering efforts and systems which utilize different components of the cell microenvironment simultaneously can provide more functional engineered tissues in the near future.
Collapse
Affiliation(s)
- Julien Barthes
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR-S 1121, “Biomatériaux et Bioingénierie”, 11 rue Humann, 67085 Strasbourg Cedex, France
| | - Hayriye Özçelik
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR-S 1121, “Biomatériaux et Bioingénierie”, 11 rue Humann, 67085 Strasbourg Cedex, France
| | - Mathilde Hindié
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, Université de Cergy-Pontoise, 2 Avenue Adolphe Chauvin, 95302 Cergy Pontoise, France
| | | | - Anwarul Hasan
- Biomedical Engineering and Department of Mechanical Engineering, American University of Beirut, Beirut 1107 2020, Lebanon
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nihal Engin Vrana
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR-S 1121, “Biomatériaux et Bioingénierie”, 11 rue Humann, 67085 Strasbourg Cedex, France
- Protip SAS, 8 Place de l'Hôpital, 67000, Strasbourg, France
| |
Collapse
|
42
|
Yang K, Lee J, Cho SW. Engineering biomaterials for feeder-free maintenance of human pluripotent stem cells. Int J Stem Cells 2014; 5:1-5. [PMID: 24298348 DOI: 10.15283/ijsc.2012.5.1.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2012] [Indexed: 02/03/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are capable of differentiating into any type of somatic cell, a characteristic that imparts significant therapeutic potential. Human embryonic stem cells and induced pluripotent stem cells are types of hPSCs. Although hPSCs have high therapeutic potential, their clinical relevance is limited by the requirement for animal feeder layers, which maintain their pluripotency and self-renewal. hPSCs grown on animal feeder cells are at high risk for pathogen contamination and can be affected by the immunogenicity of the feeder layer. The presence of animal feeder cells also limits the scalability of hPSCs in culture because of the high cost of culturing and batch-to-batch variations. Therefore, development of feeder-free systems is imperative for robust, lower-cost, xeno-free, scalable culture of hPSCs. Biomaterials engineered with bioactive molecules such as adhesion proteins and extracellular matrix proteins, or synthetic materials such as peptides and polymers, may provide alternative substrates to animal feeder cells. This article reviews biomaterial-based, feeder-free systems for hPSC growth and maintenance, which provide clinically relevant alternatives to feeder cell systems.
Collapse
Affiliation(s)
- Kisuk Yang
- Department of Biotechnology, Yonsei University, Seoul, Korea
| | | | | |
Collapse
|
43
|
Dang LTH, Feric NT, Laschinger C, Chang WY, Zhang B, Wood GA, Stanford WL, Radisic M. Inhibition of apoptosis in human induced pluripotent stem cells during expansion in a defined culture using angiopoietin-1 derived peptide QHREDGS. Biomaterials 2014; 35:7786-99. [PMID: 24930852 DOI: 10.1016/j.biomaterials.2014.05.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 05/08/2014] [Indexed: 02/07/2023]
Abstract
Adhesion molecule signaling is critical to human pluripotent stem cell (hPSC) survival, self-renewal, and differentiation. Thus, hPSCs are grown as clumps of cells on feeder cell layers or poorly defined extracellular matrices such as Matrigel. We sought to define a small molecule that would initiate adhesion-based signaling to serve as a basis for a defined substrate for hPSC culture. Soluble angiopoeitin-1 (Ang-1)-derived peptide QHREDGS added to defined serum-free media increased hPSC colony cell number and size during long- and short-term culture when grown on feeder cell layers or Matrigel, i.e. on standard substrates, without affecting hPSC morphology, growth rate or the ability to differentiate into multiple lineages both in vitro and in vivo. Importantly, QHREDGS treatment decreased hPSC apoptosis during routine passaging and single-cell dissociation. Mechanistically, the interaction of QHREDGS with β1-integrins increased expression of integrin-linked kinase (ILK), increased expression and activation of extracellular signal-regulated kinases 1/2 (ERK1/2), and decreased caspase-3/7 activity. QHREDGS immobilization to polyethylene glycol hydrogels significantly increased cell adhesion in a dose-dependent manner. We propose QHREDGS as a small molecule inhibitor of hPSC apoptosis and the basis of an affordable defined substrate for hPSC maintenance.
Collapse
Affiliation(s)
- Lan T H Dang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; The Heart and Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Nicole T Feric
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Carol Laschinger
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Wing Y Chang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Sprott Centre for Stem Cell Research, The Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Boyang Zhang
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - William L Stanford
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; Sprott Centre for Stem Cell Research, The Ottawa Hospital Research Institute, Ottawa, Ontario K1H 8L6, Canada; Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada; The Heart and Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada.
| |
Collapse
|
44
|
Wang L, Zhao M, Li S, Erasquin UJ, Wang H, Ren L, Chen C, Wang Y, Cai C. "Click" immobilization of a VEGF-mimetic peptide on decellularized endothelial extracellular matrix to enhance angiogenesis. ACS APPLIED MATERIALS & INTERFACES 2014; 6:8401-8406. [PMID: 24749832 PMCID: PMC4059262 DOI: 10.1021/am501309d] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/21/2014] [Indexed: 05/29/2023]
Abstract
We show that coating of decellularized extracellular matrix (DC-ECM) on substrate surfaces is an efficient way to generate a platform mimicking the native ECM environment. Moreover, the DC-ECM can be modified with a peptide (QK) mimicking vascular endothelial growth factor without apparently compromising its integrity. The modification was achieved through metabolic incorporation of a "clickable" handle to DC-ECM followed by rapid attachment of the QK peptide with an azido tag using copper-catalyzed click reaction. The attachment of the QK peptide on to DC-ECM in this way further enhanced the angiogenic responses (formation of branched tubular networks) of endothelial cells.
Collapse
Affiliation(s)
- Lin Wang
- Department
of Chemistry, University of Houston, Houston, Texas 77204, United States
- Biomaterials
Research Center, South China University
of Technology, Guangzhou 510640, China
| | - Meirong Zhao
- Department
of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Siheng Li
- Department
of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Uriel J. Erasquin
- Department
of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Hao Wang
- Molecular
Surgeon Research Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Li Ren
- Biomaterials
Research Center, South China University
of Technology, Guangzhou 510640, China
| | - Changyi Chen
- Molecular
Surgeon Research Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Yingjun Wang
- Biomaterials
Research Center, South China University
of Technology, Guangzhou 510640, China
| | - Chengzhi Cai
- Department
of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
45
|
Zhang D, Kilian KA. Peptide microarrays for the discovery of bioactive surfaces that guide cellular processes: a single step azide-alkyne "click" chemistry approach. J Mater Chem B 2014; 2:4280-4288. [PMID: 32261566 DOI: 10.1039/c4tb00375f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cell behavior in vivo is guided by a complex microenvironment containing many different molecules including extracellular matrix (ECM) proteins, growth factors, and proteoglycans. Controlling the interaction between these various components at the cell-material interface will be invaluable in developing new materials for biomedical devices and tissue engineering applications. We report a single step approach to forming mixed peptide conjugated self-assembled monolayers on gold using copper-catalyzed azide-alkyne cycloaddition chemistry to study the combinatorial effects of different peptide ligands on cellular processes. We synthesized ECM adhesion peptides (YIGSR, GRGDS), a bone morphogenetic protein 7 (BMP-7) derived peptide (KPSSAPTQLN), and a heparin binding peptide (KRSR), and arrayed them, alone and in combination, onto gold coated coverslips. SAMs were characterized by X-ray photoelectron spectroscopy (XPS) and matrix-assisted laser desorption/ionization (MALDI) mass spectrometry, and arrayed peptide combinations were seen to differentially bind to adipose derived stem cells (ADSCs) and mouse embryonic fibroblasts (MEFs). We further investigated the osteogenesis of ADSCs on SAMs containing combinations of adhesion peptide and BMP-7 peptide in both standard culture and osteogenic differentiation media. We demonstrate enhanced expression of osteogenic markers Runx2 and osteopontin when ADSCs are adherent to BMP-7 derived peptide alone or in combination with ECM adhesion peptides. The platform presented here enables immobilization of multiple peptides in a single step using a commercially available microarray spotter which will prove useful in fabricating biomolecule interfaces for cell biology studies and biochemical assays.
Collapse
Affiliation(s)
- Douglas Zhang
- Department of Materials Science and Engineering, University of Illinois, Urbana-Champaign, Illinois 61801, USA.
| | | |
Collapse
|
46
|
Kandasamy K, Narayanan K, Ni M, Du C, Wan ACA, Zink D. Polysulfone Membranes Coated with Polymerized 3,4-Dihydroxy-l-phenylalanine are a Versatile and Cost-Effective Synthetic Substrate for Defined Long-Term Cultures of Human Pluripotent Stem Cells. Biomacromolecules 2014; 15:2067-78. [DOI: 10.1021/bm5001907] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Karthikeyan Kandasamy
- Institute of Bioengineering and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Karthikeyan Narayanan
- Institute of Bioengineering and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Ming Ni
- Institute of Bioengineering and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Chan Du
- Institute of Bioengineering and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Andrew C. A. Wan
- Institute of Bioengineering and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, 31
Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|
47
|
Deiss F, Matochko WL, Govindasamy N, Lin EY, Derda R. Flow‐Through Synthesis on Teflon‐Patterned Paper To Produce Peptide Arrays for Cell‐Based Assays. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201402037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Frédérique Deiss
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2 (Canada)
| | - Wadim L. Matochko
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2 (Canada)
| | - Natasha Govindasamy
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2 (Canada)
| | - Edith Y. Lin
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2 (Canada)
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2 (Canada)
| |
Collapse
|
48
|
Flow‐Through Synthesis on Teflon‐Patterned Paper To Produce Peptide Arrays for Cell‐Based Assays. Angew Chem Int Ed Engl 2014; 53:6374-7. [DOI: 10.1002/anie.201402037] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Indexed: 11/07/2022]
|
49
|
Loring JF, McDevitt TC, Palecek SP, Schaffer DV, Zandstra PW, Nerem RM. A global assessment of stem cell engineering. Tissue Eng Part A 2014; 20:2575-89. [PMID: 24428577 DOI: 10.1089/ten.tea.2013.0468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Over the last 2 years a global assessment of stem cell engineering (SCE) was conducted with the sponsorship of the National Science Foundation, the National Cancer Institute at the National Institutes of Health, and the National Institute of Standards and Technology. The purpose was to gather information on the worldwide status and trends in SCE, that is, the involvement of engineers and engineering approaches in the stem cell field, both in basic research and in the translation of research into clinical applications and commercial products. The study was facilitated and managed by the World Technology Evaluation Center. The process involved site visits in both Asia and Europe, and it also included several different workshops. From this assessment, the panel concluded that there needs to be an increased role for engineers and the engineering approach. This will provide a foundation for the generation of new markets and future economic growth. To do this will require an increased investment in engineering, applied research, and commercialization as it relates to stem cell research and technology. It also will require programs that support interdisciplinary teams, new innovative mechanisms for academic-industry partnerships, and unique translational models. In addition, the global community would benefit from forming strategic partnerships between countries that can leverage existing and emerging strengths in different institutions. To implement such partnerships will require multinational grant programs with appropriate review mechanisms.
Collapse
Affiliation(s)
- Jeanne F Loring
- 1 Director, Center for Regenerative Medicine, the Scripps Research Institute , LaJolla, California
| | | | | | | | | | | |
Collapse
|
50
|
Tan KY, Lin H, Ramstedt M, Watt FM, Huck WTS, Gautrot JE. Decoupling geometrical and chemical cues directing epidermal stem cell fate on polymer brush-based cell micro-patterns. Integr Biol (Camb) 2014; 5:899-910. [PMID: 23572192 DOI: 10.1039/c3ib40026c] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The intricacy of the different parameters involved in cell adhesion to biomaterials and fate decision (e.g. proliferation, differentiation, apoptosis) makes the decoupling of the respective effects of surface properties, extra-cellular matrix protein adsorption and ultimately cell behaviour difficult. This work presents a micro-patterned polymer brush platform to control the adsorption of extra-cellular matrix (ECM) proteins to well defined micron-size areas and consequently control cell adhesion, spreading and shape independently of other chemical and physical surface properties. Protein patterns can be readily generated with brushes presenting a range of hydrophilicity and surface charge density. The surface properties of the selected brushes are fully characterised using a combination of FTIR, XPS, ellipsometry, atomic force microscopy, water contact goniometry, dynamic light scattering and ζ-potential measurements. Interactions of proteins relevant to cell patterning and culture with these brushes are studied by surface plasmon resonance, dynamic light scattering, ellipsometry and immuno-fluorescence microscopy. Finally this platform is used in an assay investigating the relative contributions of matrix geometry and surface chemistry on epidermal stem cell differentiation. It is found that moderate hydrophobicity does not impact stem cell commitment, whereas strongly negative surface potential increases the incidence of differentiation. This correlates with a marked decrease in the formation of focal adhesions (but not cell spreading).
Collapse
Affiliation(s)
- Khooi Y Tan
- Melville Laboratory for Polymer Synthesis, Department of Chemistry, University of Cambridge, Cambridge, UK
| | | | | | | | | | | |
Collapse
|