1
|
Mulero-Russe A, Mora-Boza A, Marquez EN, Ziegelski M, Helmrath M, García AJ. Synthetic hydrogel substrate for human induced pluripotent stem cell definitive endoderm differentiation. Biomaterials 2025; 315:122920. [PMID: 39504708 PMCID: PMC11625597 DOI: 10.1016/j.biomaterials.2024.122920] [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: 06/27/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024]
Abstract
Human induced pluripotent stem cells (hiPSCs) can give rise to multiple lineages derived from three germ layers, endoderm, mesoderm and ectoderm. Definitive endoderm (DE) cell types and tissues have great potential for regenerative medicine applications. Current hiPSC differentiation protocols focus on the addition of soluble factors; however, extracellular matrix properties are known to also play a role in dictating cell fate. Matrigel™ is the gold standard for DE differentiation, but this xenogeneic, poorly defined basement membrane extract limits the clinical translatability of DE-derived tissues. Here we present a fully defined PEG-based hydrogel substrate to support hiPSC-derived DE differentiation. We screened hydrogel formulations presenting different adhesive peptides and matrix stiffness. Our results demonstrate that presenting a short peptide, cyclic RGD, on the engineered PEG hydrogel supports the transition from undifferentiated hiPSCs to DE using a serum-free, commercially available kit. We show that increasing substrate stiffness (G' = 1.0-4.0 kPa) results in an increased linear response in DE differentiation efficiency. We also include a temporal analysis of the expression of integrin and syndecan receptors as the hiPSCs undergo specification towards DE lineage. Finally, we show that focal adhesion kinase activity regulates hiPSC growth and DE differentiation efficiency. Overall, we present a fully defined matrix as a synthetic alternative for Matrigel™ supporting DE differentiation.
Collapse
Affiliation(s)
- Adriana Mulero-Russe
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elijah N Marquez
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Morgan Ziegelski
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael Helmrath
- Division of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
2
|
Spetz MR, Kim H, Chavarria D, Conger DJ, Shattuck-Brandt R, Shekharan SR, Shostak A, Ligocki AP, Brien HJ, Embalabala RJ, Mobley BC, Schrag MS, Lippmann ES, Brunger JM. Amyloid-β-regulated gene circuits for programmable Alzheimer's disease therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642808. [PMID: 40161792 PMCID: PMC11952467 DOI: 10.1101/2025.03.12.642808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized in part by the accumulation of the protein amyloid-β (Aβ). Monoclonal antibodies (mAbs) that target Aβ for clearance from the brain have received FDA approval; however, these therapies are accompanied by serious side effects, and their cognitive benefit for patients remains of tremendous debate. Here, we present a potential engineered cell therapy for AD in which we enlist cells of the central nervous system as programmable agents for sculpting the neurodegenerative niche toward one that mitigates glial reactivity and neuronal loss. We constructed a suite of Aβ-sensitive synthetic Notch (synNotch) receptors from clinically tested anti-Aβ mAbs and show that cells expressing these receptors can recognize synthetic Aβ42 and Aβ40 with differential sensitivity. We express these receptors in astrocytes, cells native to the brain that are known to become dysfunctional in AD. These synNotch astrocytes, which upregulate selected transgenes upon exposure to synthetic and human brain-derived amyloid, were engineered to express potential therapeutic transgenes in response to Aβ, including brain-derived neurotrophic factor and antagonists of the cytokines tumor necrosis factor and interleukin-1. SynNotch astrocytes that express such antagonists in response to Aβ partially attenuate a cytokine-induced reactive astrocyte phenotype and promote barrier properties in brain microvascular endothelial cells. Additionally, engineered Aβ-synNotch cells potently upregulate transgene expression in response to Aβ deposited in the 5xFAD mouse brain, demonstrating the capacity to recognize Aβ in situ. Overall, our work supports Aβ-synNotch receptors as promising tools to generate a cell-based therapy for AD with targeted functionalities to positively influence the AD niche.
Collapse
Affiliation(s)
- Madeline R Spetz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Daniel Chavarria
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Dylan J Conger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | | | - Swathi R Shekharan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Alena Shostak
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
| | - Alexander P Ligocki
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
| | - Hannah J Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Rebecca J Embalabala
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Bret C Mobley
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew S Schrag
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN
| | - Ethan S Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|
3
|
Critcher M, Pang JM, Huang ML. Mapping the FGF2 Interactome Identifies a Functional Proteoglycan Coreceptor. ACS Chem Biol 2025; 20:105-116. [PMID: 39704408 PMCID: PMC11858877 DOI: 10.1021/acschembio.4c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Fibroblast growth factor 2 (FGF2) is a multipotent growth factor and signaling protein that exhibits broad functions across multiple cell types. These functions are often initiated by binding to growth factor receptors and fine-tuned by glycosaminoglycan (GAG)-modified proteins called proteoglycans. The various outputs of FGF2 signaling and functions arise from a dynamic and cell type-specific set of binding partners. However, the interactome of FGF2 has yet to be comprehensively determined. Moreover, the identity of the proteoglycan proteins carrying GAG chains is often overlooked and remains unknown in most cell contexts. Here, we perform peroxidase-catalyzed live cell proximity labeling using an engineered APEX2-FGF2 fusion protein to map the interactome of FGF2. Across two cell lines with established and distinct FGF2-driven functions, we greatly expand upon the known FGF2 interactome, identifying >600 new putative FGF2 interactors. Notably, our results demonstrate a key role for the GAG binding capacity of FGF2 in modulating its interactome.
Collapse
Affiliation(s)
- Meg Critcher
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| | - Jia Meng Pang
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| | - Mia L Huang
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, California 92037, United States
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla California 92037, United States
| |
Collapse
|
4
|
Syangtan D, Al Mahbuba D, Masuko S, Li Q, Elton AC, Zaltsman Y, Wrighton PJ, Xia K, Han X, Ouyang Y, Zhang F, Linhardt RJ, Kiessling LL. Heparan sulfate regulates the fate decisions of human pluripotent stem cells. Stem Cell Reports 2025; 20:102384. [PMID: 39729990 PMCID: PMC11784485 DOI: 10.1016/j.stemcr.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Heparan sulfate (HS) is an anionic polysaccharide generated by all animal cells, but our understanding of its roles in human pluripotent stem cell (hPSC) self-renewal and differentiation is limited. We derived HS-deficient hPSCs by disrupting the EXT1 glycosyltransferase. These EXT1-/- hPSCs maintain self-renewal and pluripotency under standard culture conditions that contain high levels of basic fibroblast growth factor(bFGF), a requirement for sufficient bFGF signaling in the engineered cells. Intriguingly, Activin/Nodal signaling is also compromised in EXT1-/- hPSCs, highlighting HS's previously unexplored involvement in this pathway. As a result, EXT1-/- hPSCs fail to differentiate into mesoderm or endoderm lineages. Unexpectedly, HS is dispensable for early ectodermal differentiation of hPSCs but still critical in generating motor neurons. Those derived from HS-deficient hPSCs lack proper neuronal projections and show alterations in axonogenesis gene expression. Thus, our study uncovers expected and unexpected mechanistic roles of HS in hPSC fate decisions.
Collapse
Affiliation(s)
- Deepsing Syangtan
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Deena Al Mahbuba
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Sayaka Masuko
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Qiao Li
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Andrew C Elton
- School of Pharmacy, University of Wisconsin-Madison, 777 Highland Avenue, Madison, WI 53705, USA
| | - Yefim Zaltsman
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Paul J Wrighton
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 110 8th St., Troy, NY 12180, USA
| | - Xiaorui Han
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 110 8th St., Troy, NY 12180, USA
| | - Yilan Ouyang
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 110 8th St., Troy, NY 12180, USA
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 110 8th St., Troy, NY 12180, USA
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 110 8th St., Troy, NY 12180, USA
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI 53706, USA; Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142, USA; Koch Institute for Integrative Cancer Research at MIT, 500 Main St, Cambridge, MA 02142, USA.
| |
Collapse
|
5
|
Caetano-Silva SP, Marr N, Rawlinson SCF, Pitsillides AA. Using Cell and Organ Culture Models to Analyze Responses of Bone Cells to Mechanical Stimulation. Methods Mol Biol 2025; 2885:159-191. [PMID: 40448761 DOI: 10.1007/978-1-0716-4306-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
The techniques that are useful for applying mechanical strain to bone and bone cells are now more diverse than described in the second edition. Their output has also increased substantially and, perhaps most importantly, their significance is now broadly accepted. This growth in the use of methods for applying mechanical strain to bone and its constituent cells and increased awareness of the importance of the mechanical environment in controlling normal bone cell behavior has indeed heralded new therapeutic approaches. We have expanded the text to include additions and modifications made to the straining apparatus and updated the research cited to support this growing role of cell cultures, including co-culture systems and primary cells, tissue engineering, and organ culture models to analyze responses of bone cells to mechanical stimulation. We understand that there are approaches not covered here and appreciate that alternative strategies have their own value and utility.
Collapse
Affiliation(s)
- Soraia P Caetano-Silva
- Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College of London, London, UK
| | - Neil Marr
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Simon C F Rawlinson
- Queen Mary University of London, Barts & The London School of Medicine and Dentistry, Institute of Dentistry, London, UK
| | - Andrew A Pitsillides
- Skeletal Biology Group, Comparative Biomedical Sciences, The Royal Veterinary College, London, UK.
| |
Collapse
|
6
|
Holman AR, Tran S, Destici E, Farah EN, Li T, Nelson AC, Engler AJ, Chi NC. Single-cell multi-modal integrative analyses highlight functional dynamic gene regulatory networks directing human cardiac development. CELL GENOMICS 2024; 4:100680. [PMID: 39437788 PMCID: PMC11605693 DOI: 10.1016/j.xgen.2024.100680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Illuminating the precise stepwise genetic programs directing cardiac development provides insights into the mechanisms of congenital heart disease and strategies for cardiac regenerative therapies. Here, we integrate in vitro and in vivo human single-cell multi-omic studies with high-throughput functional genomic screening to reveal dynamic, cardiac-specific gene regulatory networks (GRNs) and transcriptional regulators during human cardiomyocyte development. Interrogating developmental trajectories reconstructed from single-cell data unexpectedly reveal divergent cardiomyocyte lineages with distinct gene programs based on developmental signaling pathways. High-throughput functional genomic screens identify key transcription factors from inferred GRNs that are functionally relevant for cardiomyocyte lineages derived from each pathway. Notably, we discover a critical heat shock transcription factor 1 (HSF1)-mediated cardiometabolic GRN controlling cardiac mitochondrial/metabolic function and cell survival, also observed in fetal human cardiomyocytes. Overall, these multi-modal genomic studies enable the systematic discovery and validation of coordinated GRNs and transcriptional regulators controlling the development of distinct human cardiomyocyte populations.
Collapse
Affiliation(s)
- Alyssa R Holman
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shaina Tran
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eugin Destici
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elie N Farah
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ting Li
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Aileena C Nelson
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92093, USA
| | - Neil C Chi
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
7
|
Brien H, Lee JC, Sharma J, Hamann CA, Spetz MR, Lippmann ES, Brunger JM. Templated Pluripotent Stem Cell Differentiation via Substratum-Guided Artificial Signaling. ACS Biomater Sci Eng 2024; 10:6465-6482. [PMID: 39352143 PMCID: PMC11480943 DOI: 10.1021/acsbiomaterials.4c00885] [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: 05/13/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024]
Abstract
The emerging field of synthetic morphogenesis implements synthetic biology tools to investigate the minimal cellular processes sufficient for orchestrating key developmental events. As the field continues to grow, there is a need for new tools that enable scientists to uncover nuances in the molecular mechanisms driving cell fate patterning that emerge during morphogenesis. Here, we present a platform that combines cell engineering with biomaterial design to potentiate artificial signaling in pluripotent stem cells (PSCs). This platform, referred to as PSC-MATRIX, extends the use of programmable biomaterials to PSCs competent to activate morphogen production through orthogonal signaling, giving rise to the opportunity to probe developmental events by initiating morphogenetic programs in a spatially constrained manner through non-native signaling channels. We show that the PSC-MATRIX platform enables temporal and spatial control of transgene expression in response to bulk, soluble inputs in synthetic Notch (synNotch)-engineered human PSCs for an extended culture of up to 11 days. Furthermore, we used PSC-MATRIX to regulate multiple differentiation events via material-mediated artificial signaling in engineered PSCs using the orthogonal ligand green fluorescent protein, highlighting the potential of this platform for probing and guiding fate acquisition. Overall, this platform offers a synthetic approach to interrogate the molecular mechanisms driving PSC differentiation that could be applied to a variety of differentiation protocols.
Collapse
Affiliation(s)
- Hannah
J. Brien
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Joanne C. Lee
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jhanvi Sharma
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Catherine A. Hamann
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Madeline R. Spetz
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Ethan S. Lippmann
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jonathan M. Brunger
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
8
|
Ma R, Bi H, Wang Y, Wang J, Zhang J, Yu X, Chen Z, Wang J, Lu C, Zheng J, Li Y, Ding X. Low concentrations of saracatinib promote definitive endoderm differentiation through inhibition of FAK-YAP signaling axis. Cell Commun Signal 2024; 22:300. [PMID: 38816763 PMCID: PMC11140888 DOI: 10.1186/s12964-024-01679-7] [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: 01/16/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024] Open
Abstract
Optimizing the efficiency of definitive endoderm (DE) differentiation is necessary for the generation of diverse organ-like structures. In this study, we used the small molecule inhibitor saracatinib (SAR) to enhance DE differentiation of human embryonic stem cells and induced pluripotent stem cells. SAR significantly improved DE differentiation efficiency at low concentrations. The interaction between SAR and Focal Adhesion Kinase (FAK) was explored through RNA-seq and molecular docking simulations, which further supported the inhibition of DE differentiation by p-FAK overexpression in SAR-treated cells. In addition, we found that SAR inhibited the nuclear translocation of Yes-associated protein (YAP), a downstream effector of FAK, which promoted DE differentiation. Moreover, the addition of SAR enabled a significant reduction in activin A (AA) from 50 to 10 ng/mL without compromising DE differentiation efficiency. For induction of the pancreatic lineage, 10 ng/ml AA combined with SAR at the DE differentiation stage yielded a comparative number of PDX1+/NKX6.1+ pancreatic progenitor cells to those obtained by 50 ng/ml AA treatment. Our study highlights SAR as a potential modulator that facilitates the cost-effective generation of DE cells and provides insight into the orchestration of cell fate determination.
Collapse
Affiliation(s)
- Ruiyang Ma
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Huanjing Bi
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Ying Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Jingwen Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Jiangwei Zhang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Xiaoyang Yu
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Zuhan Chen
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Jiale Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Cuinan Lu
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Yang Li
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi Province, 710061, China.
| |
Collapse
|
9
|
Lee JC, Brien HJ, Walton BL, Eidman ZM, Toda S, Lim WA, Brunger JM. Instructional materials that control cellular activity through synthetic Notch receptors. Biomaterials 2023; 297:122099. [PMID: 37023529 PMCID: PMC10320837 DOI: 10.1016/j.biomaterials.2023.122099] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
The field of regenerative engineering relies primarily on the dual technical platforms of cell selection/conditioning and biomaterial fabrication to support directed cell differentiation. As the field has matured, an appreciation for the influence of biomaterials on cell behaviors has resulted in engineered matrices that meet biomechanical and biochemical demands of target pathologies. Yet, despite advances in methods to produce designer matrices, regenerative engineers remain unable to reliably orchestrate behaviors of therapeutic cells in situ. Here, we present a platform named MATRIX whereby cellular responses to biomaterials can be custom defined by combining engineered materials with cells expressing cognate synthetic biology control modules. Such privileged channels of material-to-cell communication can activate synthetic Notch receptors and govern activities as diverse as transcriptome engineering, inflammation attenuation, and pluripotent stem cell differentiation, all in response to materials decorated with otherwise bioinert ligands. Further, we show that engineered cellular behaviors are confined to programmed biomaterial surfaces, highlighting the potential to use this platform to spatially organize cellular responses to bulk, soluble factors. This integrated approach of co-engineering cells and biomaterials for orthogonal interactions opens new avenues for reproducible control of cell-based therapies and tissue replacements.
Collapse
Affiliation(s)
- Joanne C Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Hannah J Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Zachary M Eidman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Satoshi Toda
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Wendell A Lim
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA.
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA.
| |
Collapse
|
10
|
Ma S, Li X, Cao R, Zhan G, Fu X, Xiao R, Yang Z. Developmentally regulated expression of integrin alpha-6 distinguishes neural crest derivatives in the skin. Front Cell Dev Biol 2023; 11:1140554. [PMID: 37255601 PMCID: PMC10225710 DOI: 10.3389/fcell.2023.1140554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023] Open
Abstract
Neural crest-derived cells play essential roles in skin function and homeostasis. However, how they interact with environmental cues and differentiate into functional skin cells remains unclear. Using a combination of single-cell data analysis, neural crest lineage tracing, and flow cytometry, we found that the expression of integrin α6 (ITGA6) in neural crest and its derivatives was developmentally regulated and that ITGA6 could serve as a functional surface marker for distinguishing neural crest derivatives in the skin. Based on the expression of ITGA6, Wnt1-Cre lineage neural crest derivatives in the skin could be categorized into three subpopulations, namely, ITGA6bright, ITGA6dim, and ITGA6neg, which were found to be Schwann cells, melanocytes, and fibroblasts, respectively. We further analyzed the signature genes and transcription factors that specifically enriched in each cell subpopulation, as well as the ligand or receptor molecules, mediating the potential interaction with other cells of the skin. Additionally, we found that Hmx1 and Lhx8 are specifically expressed in neural crest-derived fibroblasts, while Zic1 and homeobox family genes are expressed in mesoderm-derived fibroblasts, indicating the distinct development pathways of fibroblasts of different origins. Our study provides insights into the regulatory landscape of neural crest cell development and identifies potential markers that facilitate the isolation of different neural crest derivatives in the skin.
Collapse
Affiliation(s)
- Shize Ma
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiu Li
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Cao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Guoqin Zhan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Fu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Ran Xiao
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of External Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Sung TC, Wang T, Liu Q, Ling QD, Subbiah SK, Renuka RR, Hsu ST, Umezawa A, Higuchi A. Cell-binding peptides on the material surface guide stem cell fate of adhesion, proliferation and differentiation. J Mater Chem B 2023; 11:1389-1415. [PMID: 36727243 DOI: 10.1039/d2tb02601e] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Human cells, especially stem cells, need to communicate and interact with extracellular matrix (ECM) proteins, which not only serve as structural components but also guide and support cell fate and properties such as cell adhesion, proliferation, survival and differentiation. The binding of the cells with ECM proteins or ECM-derived peptides via cell adhesion receptors such as integrins activates several signaling pathways that determine the cell fate, morphological change, proliferation and differentiation. The development of synthetic ECM protein-derived peptides that mimic the biological and biochemical functions of natural ECM proteins will benefit academic and clinical application. Peptides derived from or inspired by specific ECM proteins can act as agonists of each ECM protein receptor. Given that most ECM proteins function in cell adhesion via integrin receptors, many peptides have been developed that bind to specific integrin receptors. In this review, we discuss the peptide sequence, immobilization design, reaction method, and functions of several ECM protein-derived peptides. Various peptide sequences derived from mainly ECM proteins, which are used for coating or grafting on dishes, scaffolds, hydrogels, implants or nanofibers, have been developed to improve the adhesion, proliferation or differentiation of stem cells and to culture differentiated cells. This review article will help to inform the optimal choice of ECM protein-derived peptides for the development of scaffolds, implants, hydrogels, nanofibers and 2D cell culture dishes to regulate the proliferation and direct the differentiation of stem cells into specific lineages.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qian Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Hsi-Chi City, Taipei 221, Taiwan
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Tambaram East, Chennai-73, 600078, India
| | - Remya Rajan Renuka
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, 173, Agaram Road, Tambaram East, Chennai-73, 600078, India
| | - Shih-Tien Hsu
- Department of Internal Medicine, Taiwan Landseed Hospital, 77 Kuangtai Road, Pingjen City, Tao-Yuan County 32405, Taiwan
| | - Akihiro Umezawa
- Department of Reproduction, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China. .,Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan. .,R & D Center for Membrane Technology, Chung Yuan Christian University, 200 Chung-Bei Rd., Jhongli, Taoyuan 320, Taiwan
| |
Collapse
|
12
|
Eguchi A, Gonzalez AFGS, Torres-Bigio SI, Koleckar K, Birnbaum F, Zhang JZ, Wang VY, Wu JC, Artandi SE, Blau HM. TRF2 rescues telomere attrition and prolongs cell survival in Duchenne muscular dystrophy cardiomyocytes derived from human iPSCs. Proc Natl Acad Sci U S A 2023; 120:e2209967120. [PMID: 36719921 PMCID: PMC9963063 DOI: 10.1073/pnas.2209967120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 12/29/2022] [Indexed: 02/01/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle wasting disease caused by the lack of dystrophin. Heart failure, driven by cardiomyocyte death, fibrosis, and the development of dilated cardiomyopathy, is the leading cause of death in DMD patients. Current treatments decrease the mechanical load on the heart but do not address the root cause of dilated cardiomyopathy: cardiomyocyte death. Previously, we showed that telomere shortening is a hallmark of DMD cardiomyocytes. Here, we test whether prevention of telomere attrition is possible in cardiomyocytes differentiated from patient-derived induced pluripotent stem cells (iPSC-CMs) and if preventing telomere shortening impacts cardiomyocyte function. We observe reduced cell size, nuclear size, and sarcomere density in DMD iPSC-CMs compared with healthy isogenic controls. We find that expression of just one telomere-binding protein, telomeric repeat-binding factor 2 (TRF2), a core component of the shelterin complex, prevents telomere attrition and rescues deficiencies in cell size as well as sarcomere density. We employ a bioengineered platform to micropattern cardiomyocytes for calcium imaging and perform Southern blots of telomere restriction fragments, the gold standard for telomere length assessments. Importantly, preservation of telomere lengths in DMD cardiomyocytes improves their viability. These data provide evidence that preventing telomere attrition ameliorates deficits in cell morphology, activation of the DNA damage response, and premature cell death, suggesting that TRF2 is a key player in DMD-associated cardiac failure.
Collapse
Affiliation(s)
- Asuka Eguchi
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, CA94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Adriana Fernanda G. S. Gonzalez
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, CA94305
| | - Sofía I. Torres-Bigio
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Kassie Koleckar
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, CA94305
| | - Foster Birnbaum
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, CA94305
| | - Joe Z. Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Vicky Y. Wang
- Stanford Department of Radiology, Stanford University School of Medicine, Stanford, CA94305
| | - Joseph C. Wu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
- Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Steven E. Artandi
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94035
| | - Helen M. Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford School of Medicine, Stanford, CA94305
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
13
|
Alarfaj AA, Hirad AH, Munusamy MA, Kumar SS, Higuchi A. Human embryonic stem cells cultured on hydrogels grafted with extracellular matrix protein-derived peptides with polyethylene glycol joint nanosegments. IET Nanobiotechnol 2022; 16:295-304. [PMID: 36200801 DOI: 10.1049/nbt2.12091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 11/19/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) can be proliferated on completely synthetic materials under xeno-free cultivation conditions using biomaterials grafted with extracellular matrix protein (ECM)-derived peptides. However, cell culture biomaterials grafted with ECM-derived peptides must be prepared using a high concentration of peptide reaction solution (e.g. 1000 μg/ml), whereas the ECM concentration of the ECM-coated surface for hPSC culture is typically 5 μg/ml. We designed a polyethylene glycol (PEG) joint nanosegment (linker) to be used between base cell culture biomaterials and bioactive ECM-derived peptides to enhance the probability of contact between ECM-derived peptides and cell binding receptors of hPSCs. Vitronectin-derived peptides with glycine joint nanosegments (GCGG) were conjugated onto poly (vinyl alcohol-co-itaconic acid) hydrogels via PEG joint nanosegments, and human embryonic stem cells (hESCs) were cultivated on these hydrogels. hESCs could successfully be cultivated on hydrogels while maintaining their pluripotency and differentiation potential to differentiate into cells that are induced from three germ layers in vitro and in vivo, where only a 50 μg/ml ECM-derived peptide concentration was used when the PEG joint nanosegments were introduced into peptides that were grafted onto hydrogel surfaces. The joint nanosegments between bioactive peptides and base cell culture biomaterials were found to contribute to efficient hESC attachment and proliferation.
Collapse
Affiliation(s)
- Abdullah A Alarfaj
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Abdurahman H Hirad
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Murugan A Munusamy
- Department of Botany and Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - S Suresh Kumar
- Department of Biotechnology, Bharath Institute of Higher Education and Research, Chennai-73, India
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan.,Department of Reproduction, National Center for Child Health and Development, Tokyo, Japan.,School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China.,R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| |
Collapse
|
14
|
Afzal J, Liu Y, Du W, Suhail Y, Zong P, Feng J, Ajeti V, Sayyad WA, Nikolaus J, Yankova M, Deymier AC, Yue L, Kshitiz. Cardiac ultrastructure inspired matrix induces advanced metabolic and functional maturation of differentiated human cardiomyocytes. Cell Rep 2022; 40:111146. [PMID: 35905711 DOI: 10.1016/j.celrep.2022.111146] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/26/2022] [Accepted: 07/07/2022] [Indexed: 12/21/2022] Open
Abstract
The vast potential of human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) in preclinical models of cardiac pathologies, precision medicine, and drug screening remains to be fully realized because hiPSC-CMs are immature without adult-like characteristics. Here, we present a method to accelerate hiPSC-CM maturation on a substrate, cardiac mimetic matrix (CMM), mimicking adult human heart matrix ligand chemistry, rigidity, and submicron ultrastructure, which synergistically mature hiPSC-CMs rapidly within 30 days. hiPSC-CMs matured on CMM exhibit systemic transcriptomic maturation toward an adult heart state, are aligned with high strain energy, metabolically rely on oxidative phosphorylation and fatty acid oxidation, and display enhanced redox handling capability, efficient calcium handling, and electrophysiological features of ventricular myocytes. Endothelin-1-induced pathological hypertrophy is mitigated on CMM, highlighting the role of a native cardiac microenvironment in withstanding hypertrophy progression. CMM is a convenient model for accelerated development of ventricular myocytes manifesting highly specialized cardiac-specific functions.
Collapse
Affiliation(s)
- Junaid Afzal
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Yamin Liu
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA
| | - Wenqiang Du
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA; Center for Cellular Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Pengyu Zong
- Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA; Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT 06032, USA
| | - Jianlin Feng
- Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA; Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT 06032, USA
| | - Visar Ajeti
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA; Center for Cellular Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Wasim A Sayyad
- Department of Cell Biology, Yale University, New Haven, CT 06510, USA
| | - Joerg Nikolaus
- West Campus Imaging Core, Yale University, New Haven, CT 06477, USA
| | - Maya Yankova
- Electron Microscopy Core, University of Connecticut Health, Farmington, CT 06032, USA
| | - Alix C Deymier
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA
| | - Lixia Yue
- Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA; Calhoun Cardiology Center, University of Connecticut Health, Farmington, CT 06032, USA
| | - Kshitiz
- Department of Biomedical Engineering, University of Connecticut Health, Farmington, CT 06032, USA; Center for Cellular Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA; Department of Cell Biology, University of Connecticut Health, Farmington, CT 06032, USA.
| |
Collapse
|
15
|
Melzer MK, Breunig M, Arnold F, Wezel F, Azoitei A, Roger E, Krüger J, Merkle J, Schütte L, Resheq Y, Hänle M, Zehe V, Zengerling F, Azoitei N, Klein L, Penz F, Singh SK, Seufferlein T, Hohwieler M, Bolenz C, Günes C, Gout J, Kleger A. Organoids at the PUB: The Porcine Urinary Bladder Serves as a Pancreatic Niche for Advanced Cancer Modeling. Adv Healthc Mater 2022; 11:e2102345. [PMID: 35114730 PMCID: PMC11468201 DOI: 10.1002/adhm.202102345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/17/2021] [Indexed: 12/17/2022]
Abstract
Despite intensive research and progress in personalized medicine, pancreatic ductal adenocarcinoma remains one of the deadliest cancer entities. Pancreatic duct-like organoids (PDLOs) derived from human pluripotent stem cells (PSCs) or pancreatic cancer patient-derived organoids (PDOs) provide unique tools to study early and late stage dysplasia and to foster personalized medicine. However, such advanced systems are neither rapidly nor easily accessible and require an in vivo niche to study tumor formation and interaction with the stroma. Here, the establishment of the porcine urinary bladder (PUB) is revealed as an advanced organ culture model for shaping an ex vivo pancreatic niche. This model allows pancreatic progenitor cells to enter the ductal and endocrine lineages, while PDLOs further mature into duct-like tissue. Accordingly, the PUB offers an ex vivo platform for earliest pancreatic dysplasia and cancer if PDLOs feature KRASG12D mutations. Finally, it is demonstrated that PDOs-on-PUB i) resemble primary pancreatic cancer, ii) preserve cancer subtypes, iii) enable the study of niche epithelial crosstalk by spiking in pancreatic stellate and immune cells into the grafts, and finally iv) allow drug testing. In summary, the PUB advances the existing pancreatic cancer models by adding feasibility, complexity, and customization at low cost and high flexibility.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Department of UrologyUlm UniversityUlm89081Germany
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Markus Breunig
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Frank Arnold
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Felix Wezel
- Department of UrologyUlm UniversityUlm89081Germany
| | - Anca Azoitei
- Department of UrologyUlm UniversityUlm89081Germany
| | - Elodie Roger
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Jana Krüger
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Jessica Merkle
- Department of Internal Medicine IUlm UniversityUlm89081Germany
- Core Facility OrganoidsUlm UniversityUlm89081Germany
| | - Lena Schütte
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Yazid Resheq
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Mark Hänle
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Viktor Zehe
- Department of UrologyUlm UniversityUlm89081Germany
| | | | - Ninel Azoitei
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Lukas Klein
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medicine GoettingenGoettingen37075Germany
| | - Frederike Penz
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medicine GoettingenGoettingen37075Germany
| | - Shiv K. Singh
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medicine GoettingenGoettingen37075Germany
| | | | - Meike Hohwieler
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | | | | | - Johann Gout
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Alexander Kleger
- Department of Internal Medicine IUlm UniversityUlm89081Germany
- Core Facility OrganoidsUlm UniversityUlm89081Germany
| |
Collapse
|
16
|
Mennen RH, Oldenburger MM, Piersma AH. Endoderm and mesoderm derivatives in embryonic stem cell differentiation and their use in developmental toxicity testing. Reprod Toxicol 2021; 107:44-59. [PMID: 34861400 DOI: 10.1016/j.reprotox.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Embryonic stem cell differentiation models have increasingly been applied in non-animal test systems for developmental toxicity. After the initial focus on cardiac differentiation, attention has also included an array of neuro-ectodermal differentiation routes. Alternative differentiation routes in the mesodermal and endodermal germ lines have received less attention. This review provides an inventory of achievements in the latter areas of embryonic stem cell differentiation, with a view to possibilities for their use in non-animal test systems in developmental toxicology. This includes murine and human stem cell differentiation models, and also gains information from the field of stem cell use in regenerative medicine. Endodermal stem cell derivatives produced in vitro include hepatocytes, pancreatic cells, lung epithelium, and intestinal epithelium, and mesodermal derivatives include cardiac muscle, osteogenic, vascular and hemopoietic cells. This inventory provides an overview of studies on the different cell types together with biomarkers and culture conditions that stimulate these differentiation routes from embryonic stem cells. These models may be used to expand the spectrum of embryonic stem cell based new approach methodologies in non-animal developmental toxicity testing.
Collapse
Affiliation(s)
- R H Mennen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | | | - A H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
17
|
Wetzel R, Hauser S, Lin W, Berg P, Werner C, Pietzsch J, Kempermann G, Zhang Y. Screening Arrays of Laminin Peptides on Modified Cellulose for Promotion of Adhesion of Primary Endothelial and Neural Precursor Cells. Adv Biol (Weinh) 2021. [DOI: 10.1002/adbi.201900303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Richard Wetzel
- B CUBE Center for Molecular Bioengineering Technische Universität Dresden Tatzberg 41 Dresden 01307 Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology Helmholtz‐Zentrum Dresden‐Rossendorf Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400 Dresden 01328 Germany
| | - Weilin Lin
- B CUBE Center for Molecular Bioengineering Technische Universität Dresden Tatzberg 41 Dresden 01307 Germany
| | - Peggy Berg
- B CUBE Center for Molecular Bioengineering Technische Universität Dresden Tatzberg 41 Dresden 01307 Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Hohe Str. 6 Dresden 01069 Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology Helmholtz‐Zentrum Dresden‐Rossendorf Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400 Dresden 01328 Germany
- Faculty of Chemistry and Food Chemistry, School of Science Technische Universität Dresden Dresden 01062 Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Tatzberg 41 Dresden 01307 Germany
- CRTD—Center for Regenerative Therapies Dresden Technische Universität Dresden Fetscherstraße 105 Dresden 01307 Germany
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering Technische Universität Dresden Tatzberg 41 Dresden 01307 Germany
| |
Collapse
|
18
|
McKee C, Brown C, Bakshi S, Walker K, Govind CK, Chaudhry GR. Transcriptomic Analysis of Naïve Human Embryonic Stem Cells Cultured in Three-Dimensional PEG Scaffolds. Biomolecules 2020; 11:E21. [PMID: 33379237 PMCID: PMC7824559 DOI: 10.3390/biom11010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
Naïve human embryonic stem cells (ESCs) are characterized by improved viability, proliferation, and differentiation capacity in comparison to traditionally derived primed human ESCs. However, currently used two-dimensional (2-D) cell culture techniques fail to mimic the three-dimensional (3-D) in vivo microenvironment, altering morphological and molecular characteristics of ESCs. Here, we describe the use of 3-D self-assembling scaffolds that support growth and maintenance of the naïve state characteristics of ESC line, Elf1. Scaffolds were formed via a Michael addition reaction upon the combination of two 8-arm polyethylene glycol (PEG) polymers functionalized with thiol (PEG-8-SH) and acrylate (PEG-8-Acr) end groups. 3-D scaffold environment maintained the naïve state and supported the long-term growth of ESCs. RNA-sequencing demonstrated significant changes in gene expression profiles between 2-D and 3-D grown cells. Gene ontology analysis revealed upregulation of biological processes involved in the regulation of transcription and translation, extracellular matrix organization, and chromatin remodeling in 3-D grown cells. 3-D culture conditions also induced upregulation of genes associated with Wnt and focal adhesion signaling, while p53 signaling pathway associated genes were downregulated. Our findings, for the first time, provide insight into the possible mechanisms of self-renewal of naïve ESCs stimulated by the transduction of mechanical signals from the 3-D microenvironment.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Christina Brown
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Shreeya Bakshi
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Keegan Walker
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Chhabi K. Govind
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - G. Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| |
Collapse
|
19
|
Embryonic Stem Cell-Derived Extracellular Vesicles Maintain ESC Stemness by Activating FAK. Dev Cell 2020; 56:277-291.e6. [PMID: 33321103 DOI: 10.1016/j.devcel.2020.11.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/02/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
It is critical that epiblast cells within blastocyst-stage embryos receive the necessary regulatory cues to remain pluripotent until the appropriate time when they are stimulated to undergo differentiation, ultimately to give rise to an entire organism. Here, we show that exposure of embryonic stem cells (ESCs), which are the in vitro equivalents of epiblasts, to ESC-derived extracellular vesicles (EVs) helps to maintain their stem cell properties even under culture conditions that would otherwise induce differentiation. EV-treated ESCs continued to express stemness genes, preserving their pluripotency and ability to generate chimeric mice. These effects were triggered by fibronectin bound to the surfaces of EVs, enabling them to interact with ESC-associated integrins and activate FAK more effectively than fibronectin alone. Overall, these findings highlight a potential regulatory mechanism whereby epiblast cells, via their shed EVs, create an environment within the blastocyst that prevents their premature differentiation and maintains their pluripotent state.
Collapse
|
20
|
Yadav A, Seth B, Chaturvedi RK. Brain Organoids: Tiny Mirrors of Human Neurodevelopment and Neurological Disorders. Neuroscientist 2020; 27:388-426. [PMID: 32723210 DOI: 10.1177/1073858420943192] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Unravelling the complexity of the human brain is a challenging task. Nowadays, modern neurobiologists have developed 3D model systems called "brain organoids" to overcome the technical challenges in understanding human brain development and the limitations of animal models to study neurological diseases. Certainly like most model systems in neuroscience, brain organoids too have limitations, as these minuscule brains lack the complex neuronal circuitry required to begin the operational tasks of human brain. However, researchers are hopeful that future endeavors with these 3D brain tissues could provide mechanistic insights into the generation of circuit complexity as well as reproducible creation of different regions of the human brain. Herein, we have presented the contemporary state of brain organoids with special emphasis on their mode of generation and their utility in modelling neurological disorders, drug discovery, and clinical trials.
Collapse
Affiliation(s)
- Anuradha Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
21
|
Notbohm J, Napiwocki B, deLange W, Stempien A, Saraswathibhatla A, Craven R, Salick M, Ralphe J, Crone W. Two-Dimensional Culture Systems to Enable Mechanics-Based Assays for Stem Cell-Derived Cardiomyocytes. EXPERIMENTAL MECHANICS 2019; 59:1235-1248. [PMID: 31680699 PMCID: PMC6824432 DOI: 10.1007/s11340-019-00473-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 01/09/2019] [Indexed: 06/10/2023]
Abstract
Well-controlled 2D cell culture systems advance basic investigations in cell biology and provide innovative platforms for drug development, toxicity testing, and diagnostic assays. These cell culture systems have become more advanced in order to provide and to quantify the appropriate biomechanical and biochemical cues that mimic the milieu of conditions present in vivo. Here we present an innovative 2D cell culture system to investigate human stem cell-derived cardiomyocytes, the muscle cells of the heart responsible for pumping blood throughout the body. We designed our 2D cell culture platform to control intracellular features to produce adult-like cardiomyocyte organization with connectivity and anisotropic conduction comparable to the native heart, and combined it with optical microscopy to quantify cell-cell and cell-substrate mechanical interactions. We show the measurement of forces and displacements that occur within individual cells, between neighboring cells, and between cells and their surrounding matrix. This system has broad potential to expand our understanding of tissue physiology, with particular advantages for the study of the mechanically active heart. Furthermore, this technique should prove valuable in screening potential drugs for efficacy and testing for toxicity.
Collapse
Affiliation(s)
- J. Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
| | - B.N. Napiwocki
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
| | - W.J. deLange
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - A. Stempien
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
| | - A. Saraswathibhatla
- Department of Engineering Physics, University of Wisconsin-Madison, Madison WI, USA
| | - R.J. Craven
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
| | - M.R. Salick
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison WI, USA
| | - J.C. Ralphe
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - W.C. Crone
- Department of Engineering Physics, University of Wisconsin-Madison, Madison WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison WI, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison WI, USA
| |
Collapse
|
22
|
Park Y, Chen S, Ahmad N, Hayami T, Kapila S. Estrogen Selectively Enhances TMJ Disc but Not Knee Meniscus Matrix Loss. J Dent Res 2019; 98:1532-1538. [PMID: 31526329 DOI: 10.1177/0022034519875956] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The preponderance of temporomandibular joint (TMJ) degenerative disorders in women and their early onset during reproductive years have implicated female sex hormones, particularly 17-β estradiol (E2), in the pathogenesis of these disorders. Nevertheless, the mechanisms by which E2 contributes to TMJ degenerative disorders and the reasons for its targeted effects on the TMJ but not other joints remain poorly understood. Here, we developed an ovariectomized mouse model in which systemic E2 concentrations mimicked those in cycling women, and we determined the effect of E2 on the targeted turnover of TMJ fibrocartilage matrix via E2-induced matrix metalloproteinases MMP9 and MMP13. Infusion of E2 and progesterone (P4; hormone control) over 7 d resulted in 5- and 8-fold greater serum E2 and P4 levels relative to controls, respectively, achieving systemic hormone levels similar to high baseline levels in cycling women. Administration of E2 but not P4 caused a significant loss of TMJ collagen and glycosaminoglycans, which was accompanied by amplification of ERα and specific increases in MMP9 and MMP13 expression. This dose of E2 had no effect on knee meniscus fibrocartilage, demonstrating the specificity of the degradative effect of E2. Dose-response experiments showed a greater sensitivity and a higher peak induction of MMP9 and MMP13 in TMJ fibrocartilaginous cells than knee meniscus cells to E2, providing an explanation for the differential responses of these tissues to E2. Using MMP9- and MMP13-null mice, we observed no discernible effects of each proteinase individually to E2-mediated TMJ matrix loss but noted a significant compensatory reciprocal induction of each MMP by E2 in the absence of the other. The redundancy in E2's induction of MMP9 and MMP13 suggests that the proteinases may together contribute to E2-mediated TMJ fibrocartilage loss. These results advance our understanding of E2-mediated upregulation of MMP9 and MMP13 on fibrocartilage matrix turnover targeted to the TMJ.
Collapse
Affiliation(s)
- Y Park
- Department of Human Genetics, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - S Chen
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - N Ahmad
- Biology Department, Henry Ford College, Dearborn, MI, USA.,Wayne County Community College, Detroit, MI, USA
| | - T Hayami
- Department of Internal Medicine, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - S Kapila
- Division of Orthodontics, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
23
|
Mawaribuchi S, Aiki Y, Ikeda N, Ito Y. mRNA and miRNA expression profiles in an ectoderm-biased substate of human pluripotent stem cells. Sci Rep 2019; 9:11910. [PMID: 31417139 PMCID: PMC6695399 DOI: 10.1038/s41598-019-48447-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 08/05/2019] [Indexed: 12/27/2022] Open
Abstract
The potential applications of human pluripotent stem cells, embryonic stem (ES) cells, and induced pluripotent stem (iPS) cells in cell therapy and regenerative medicine have been widely studied. The precise definition of pluripotent stem cell status during culture using biomarkers is essential for basic research and regenerative medicine. Culture conditions, including extracellular matrices, influence the balance between self-renewal and differentiation. Accordingly, to explore biomarkers for defining and monitoring the pluripotent substates during culture, we established different substates in H9 human ES cells by changing the extracellular matrix from vitronectin to Matrigel. The substate was characterised by low and high expression of the pluripotency marker R-10G epitope and the mesenchymal marker vimentin, respectively. Immunohistochemistry, induction of the three germ layers, and exhaustive expression analysis showed that the substate was ectoderm-biased, tended to differentiate into nerves, but retained the potential to differentiate into the three germ layers. Further integrated analyses of mRNA and miRNA microarrays and qPCR analysis showed that nine genes (COL9A2, DGKI, GBX2, KIF26B, MARCH1, PLXNA4, SLC24A4, TLR4, and ZHX3) were upregulated in the ectoderm-biased cells as ectoderm-biased biomarker candidates in pluripotent stem cells. Our findings provide important insights into ectoderm-biased substates of human pluripotent stem cells in the fields of basic research and regenerative medicine.
Collapse
Affiliation(s)
- Shuuji Mawaribuchi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Yasuhiko Aiki
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Nozomi Ikeda
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Yuzuru Ito
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan.
| |
Collapse
|
24
|
Dunn KK, Reichardt IM, Simmons AD, Jin G, Floy ME, Hoon KM, Palecek SP. Coculture of Endothelial Cells with Human Pluripotent Stem Cell-Derived Cardiac Progenitors Reveals a Differentiation Stage-Specific Enhancement of Cardiomyocyte Maturation. Biotechnol J 2019; 14:e1800725. [PMID: 30927511 PMCID: PMC6849481 DOI: 10.1002/biot.201800725] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/20/2019] [Indexed: 01/14/2023]
Abstract
Cardiomyocytes (CMs) generated from human pluripotent stem cells (hPSCs) are immature in their structure and function, limiting their potential in disease modeling, drug screening, and cardiac cellular therapies. Prior studies have demonstrated that coculture of hPSC-derived CMs with other cardiac cell types, including endothelial cells (ECs), can accelerate CM maturation. To address whether the CM differentiation stage at which ECs are introduced affects CM maturation, the authors coculture hPSC-derived ECs with hPSC-derived cardiac progenitor cells (CPCs) and CMs and analyze the molecular and functional attributes of maturation. ECs have a more significant effect on acceleration of maturation when cocultured with CPCs than with CMs. EC coculture with CPCs increases CM size, expression of sarcomere, and ion channel genes and proteins, the presence of intracellular membranous extensions, and chronotropic response compared to monoculture. Maturation is accelerated with an increasing EC:CPC ratio. This study demonstrates that EC incorporation at the CPC stage of CM differentiation expedites CM maturation, leading to cells that may be better suited for in vitro and in vivo applications of hPSC-derived CMs.
Collapse
Affiliation(s)
- Kaitlin K Dunn
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| | - Isabella M Reichardt
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| | - Aaron D Simmons
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| | - Gyuhyung Jin
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| | - Martha E Floy
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| | - Kelsey M Hoon
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Engineering Drive, 53706, Madison, WI, USA
| |
Collapse
|
25
|
Engineered biomaterials to mitigate growth factor cost in cell biomanufacturing. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2018.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
26
|
Wang D, Wang Y, Liu H, Tong C, Ying Q, Sachinidis A, Li L, Peng L. Laminin promotes differentiation of rat embryonic stem cells into cardiomyocytes by activating the integrin/FAK/PI3K p85 pathway. J Cell Mol Med 2019; 23:3629-3640. [PMID: 30907509 PMCID: PMC6484303 DOI: 10.1111/jcmm.14264] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 12/27/2022] Open
Abstract
The generation of germline competent rat embryonic stem cells (rESCs) allows the study of their lineage commitment. Here, we developed a highly efficient system for rESC-derived cardiomyocytes, and even the formation of three-dimensional (3D)-like cell clusters with cTNT and α-Actinin. We have validated that laminin can interact with membrane integrin to promote the phosphorylation of both phosphatidylinositol 3-kinase (PI3K) p85 and the focal adhesion kinase (FAK). In parallel, GATA4 was up-regulated. Upon inhibiting the integrin, laminin loses the effect on cardiomyocyte differentiation, accompanied with a down-regulation of phosphorylation level of PI3K p85 and FAK. Meanwhile, the expression of Gata4 was inhibited as well. Taken together, laminin is a crucial component in the differentiation of rESCs into cardiomyocytes through increasing their proliferation via interacting with integrin pathway. These results provide new insights into the pathways mediated by extracellular laminin involved in the fate of rESC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Duo Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Research Center for Translational Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Department of Pathology and PathophysiologyTongji University School of MedicineShanghaiChina
| | - Yumei Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Research Center for Translational Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Department of Pathology and PathophysiologyTongji University School of MedicineShanghaiChina
| | - Huan Liu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Research Center for Translational Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Department of Pathology and PathophysiologyTongji University School of MedicineShanghaiChina
| | - Chang Tong
- Research Center for Translational Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Qilong Ying
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Department of Stem Cell Biology and Regenerative Medicine, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCalifornia
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular MedicineUniversity of CologneCologneGermany
| | - Li Li
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Research Center for Translational Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Department of Pathology and PathophysiologyTongji University School of MedicineShanghaiChina
| | - Luying Peng
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Research Center for Translational Medicine, Shanghai East HospitalTongji University School of MedicineShanghaiChina
- Department of Pathology and PathophysiologyTongji University School of MedicineShanghaiChina
| |
Collapse
|
27
|
Zaltsman Y, Masuko S, Bensen JJ, Kiessling LL. Angiomotin Regulates YAP Localization during Neural Differentiation of Human Pluripotent Stem Cells. Stem Cell Reports 2019; 12:869-877. [PMID: 31006631 PMCID: PMC6523060 DOI: 10.1016/j.stemcr.2019.03.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/24/2019] [Accepted: 03/25/2019] [Indexed: 12/02/2022] Open
Abstract
Leveraging the extraordinary potential of human pluripotent stem cells (hPSCs) requires an understanding of the mechanisms underlying cell-fate decisions. Substrate elasticity can induce differentiation by signaling through the transcriptional coactivator Yes-associated protein (YAP). Cells cultured on surfaces mimicking brain elasticity exclude YAP from their nuclei and differentiate to neurons. How YAP localization is controlled during neural differentiation has been unclear. We employed CRISPR/Cas9 to tag endogenous YAP in hPSCs and used this fusion protein to identify YAP's interaction partners. This engineered cell line revealed that neural differentiation promotes a change in YAP interactors, including a dramatic increase in angiomotin (AMOT) interaction with YAP. AMOT regulates YAP localization during differentiation. AMOT expression increases during neural differentiation and leads to YAP nuclear exclusion. Our findings that AMOT-dependent regulation of YAP helps direct hPSC fate provide insight into the molecular mechanisms by which the microenvironment can induce neural differentiation. Endogenous tagging reveals YAP interactors in hPSCs AMOT-YAP complex concentration increases during neural differentiation AMOT regulates YAP localization in hPSCs hPSC cytoskeleton influences YAP localization via AMOT
Collapse
Affiliation(s)
- Yefim Zaltsman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sayaka Masuko
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Joshua J Bensen
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Laura L Kiessling
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
28
|
Klontzas ME, Reakasame S, Silva R, Morais JC, Vernardis S, MacFarlane RJ, Heliotis M, Tsiridis E, Panoskaltsis N, Boccaccini AR, Mantalaris A. Oxidized alginate hydrogels with the GHK peptide enhance cord blood mesenchymal stem cell osteogenesis: A paradigm for metabolomics-based evaluation of biomaterial design. Acta Biomater 2019; 88:224-240. [PMID: 30772514 DOI: 10.1016/j.actbio.2019.02.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023]
Abstract
Oxidized alginate hydrogels are appealing alternatives to natural alginate due to their favourable biodegradability profiles and capacity to self-crosslink with amine containing molecules facilitating functionalization with extracellular matrix cues, which enable modulation of stem cell fate, achieve highly viable 3-D cultures, and promote cell growth. Stem cell metabolism is at the core of cellular fate (proliferation, differentiation, death) and metabolomics provides global metabolic signatures representative of cellular status, being able to accurately identify the quality of stem cell differentiation. Herein, umbilical cord blood mesenchymal stem cells (UCB MSCs) were encapsulated in novel oxidized alginate hydrogels functionalized with the glycine-histidine-lysine (GHK) peptide and differentiated towards the osteoblastic lineage. The ADA-GHK hydrogels significantly improved osteogenic differentiation compared to gelatin-containing control hydrogels, as demonstrated by gene expression, alkaline phosphatase activity and bone extracellular matrix deposition. Metabolomics revealed the high degree of metabolic heterogeneity in the gelatin-containing control hydrogels, captured the enhanced osteogenic differentiation in the ADA-GHK hydrogels, confirmed the similar metabolism between differentiated cells and primary osteoblasts, and elucidated the metabolic mechanism responsible for the function of GHK. Our results suggest a novel paradigm for metabolomics-guided biomaterial design and robust stem cell bioprocessing. STATEMENT OF SIGNIFICANCE: Producing high quality engineered bone grafts is important for the treatment of critical sized bone defects. Robust and sensitive techniques are required for quality assessment of tissue-engineered constructs, which result to the selection of optimal biomaterials for bone graft development. Herein, we present a new use of metabolomics signatures in guiding the development of novel oxidised alginate-based hydrogels with umbilical cord blood mesenchymal stem cells and the glycine-histidine-lysine peptide, demonstrating that GHK induces stem cell osteogenic differentiation. Metabolomics signatures captured the enhanced osteogenesis in GHK hydrogels, confirmed the metabolic similarity between differentiated cells and primary osteoblasts, and elucidated the metabolic mechanism responsible for the function of GHK. In conclusion, our results suggest a new paradigm of metabolomics-driven design of biomaterials.
Collapse
|
29
|
Bicho D, Ajami S, Liu C, Reis RL, Oliveira JM. Peptide-biofunctionalization of biomaterials for osteochondral tissue regeneration in early stage osteoarthritis: challenges and opportunities. J Mater Chem B 2019; 7:1027-1044. [DOI: 10.1039/c8tb03173h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis is a degenerative joint disease characterized by the progressive deterioration of articular cartilage, synovial inflammation and changes in periarticular and subchondral bone, being a leading cause of disability.
Collapse
Affiliation(s)
- D. Bicho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| | - S. Ajami
- Institute of Orthopaedics and Musculo-Skeletal Sci, University College London, Royal National Orthopaedic Hospital
- Stanmore
- UK
| | - C. Liu
- Institute of Orthopaedics and Musculo-Skeletal Sci, University College London, Royal National Orthopaedic Hospital
- Stanmore
- UK
| | - R. L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| | - J. M. Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| |
Collapse
|
30
|
Shekari F, Han CL, Lee J, Mirzaei M, Gupta V, Haynes PA, Lee B, Baharvand H, Chen YJ, Hosseini Salekdeh G. Surface markers of human embryonic stem cells: a meta analysis of membrane proteomics reports. Expert Rev Proteomics 2018; 15:911-922. [PMID: 30358457 DOI: 10.1080/14789450.2018.1539669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
Human embryonic stem cells (hESCs) have unique biological features and attributes that make them attractive in various areas of biomedical research. With heightened applications, there is an ever increasing need for advancement of proteome analysis. Membrane proteins are one of the most important subset of hESC proteins as they can be used as surface markers. Areas covered: This review discusses commonly used surface markers of hESCs, and provides in-depth analysis of available hESC membrane proteome reports and the existence of these markers in many other cell types, especially cancer cells. Appreciating, existing ambiguity in the definition of a membrane protein, we have attempted a meta analysis of the published membrane protein reports of hESCs by using a combination of protein databases and prediction tools to find the most confident plasma membrane proteins in hESCs. Furthermore, responsiveness of plasma membrane proteins to differentiation has been discussed based on available transcriptome profiling data bank. Expert commentary: Combined transcriptome and membrane proteome analysis highlighted additional proteins that may eventually find utility as new cell surface markers.
Collapse
Affiliation(s)
- Faezeh Shekari
- a Department of Molecular Systems Biology at Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- b Department of Developmental Biology , University of Science and Culture, ACECR , Tehran , Iran
| | - Chia-Li Han
- c Chemical Biology and Molecular Biophysics Program , Institute of Chemistry , Taipei , Taiwan , Republic of China
| | - Jaesuk Lee
- d Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute , Gachon University , Incheon , Republic of Korea
| | - Mehdi Mirzaei
- e Department of Molecular Sciences , Macquarie University , Sydney , NSW , Australia
- f Australian Proteome Analysis Facility , Macquarie University , Sydney , NSW , Australia
- g Department of Clinical Medicine , Macquarie University , Sydney , NSW , Australia
| | - Vivek Gupta
- g Department of Clinical Medicine , Macquarie University , Sydney , NSW , Australia
| | - Paul A Haynes
- e Department of Molecular Sciences , Macquarie University , Sydney , NSW , Australia
| | - Bonghee Lee
- d Center for Genomics and Proteomics, Lee Gil Ya Cancer and Diabetes Institute , Gachon University , Incheon , Republic of Korea
| | - Hossein Baharvand
- b Department of Developmental Biology , University of Science and Culture, ACECR , Tehran , Iran
- h Department of Stem Cells and Developmental Biology at Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Yu-Ju Chen
- c Chemical Biology and Molecular Biophysics Program , Institute of Chemistry , Taipei , Taiwan , Republic of China
| | - Ghasem Hosseini Salekdeh
- a Department of Molecular Systems Biology at Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- e Department of Molecular Sciences , Macquarie University , Sydney , NSW , Australia
- i Department of Systems and Synthetic biology , Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research, Education, and Extension Organization , Karaj , Iran
| |
Collapse
|
31
|
Naticchia MR, Laubach LK, Tota EM, Lucas TM, Huang ML, Godula K. Embryonic Stem Cell Engineering with a Glycomimetic FGF2/BMP4 Co-Receptor Drives Mesodermal Differentiation in a Three-Dimensional Culture. ACS Chem Biol 2018; 13:2880-2887. [PMID: 30157624 DOI: 10.1021/acschembio.8b00436] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell surface glycans, such as heparan sulfate (HS), are increasingly identified as co-regulators of growth factor signaling in early embryonic development; therefore, chemical tailoring of HS activity within the cellular glycocalyx of stem cells offers an opportunity to control their differentiation. The growth factors FGF2 and BMP4 are involved in mediating the exit of murine embryonic stem cells (mESCs) from their pluripotent state and their differentiation toward mesodermal cell types, respectively. Here, we report a method for remodeling the glycocalyx of mutant Ext1-/- mESCs with defective biosynthesis of HS to drive their mesodermal differentiation in an embryoid body culture. Lipid-functionalized synthetic HS-mimetic glycopolymers with affinity for both FGF2 and BMP4 were introduced into the plasma membrane of Ext1-/- mESCs, where they acted as functional co-receptors of these growth factors and facilitated signal transduction through associated MAPK and Smad signaling pathways. We demonstrate that these materials can be employed to remodel Ext1-/- mESCs within three-dimensional embryoid body structures, providing enhanced association of BMP4 at the cell surface and driving mesodermal differentiation. As a more complete understanding of the function of HS in regulating development continues to emerge, this simple glycocalyx engineering method is poised to enable precise control over growth factor signaling activity and outcomes of differentiation in stem cells.
Collapse
Affiliation(s)
- Matthew R. Naticchia
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive #0358, La Jolla, California 92093-0358, United States
| | - Logan K. Laubach
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive #0358, La Jolla, California 92093-0358, United States
| | - Ember M. Tota
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive #0358, La Jolla, California 92093-0358, United States
| | - Taryn M. Lucas
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive #0358, La Jolla, California 92093-0358, United States
| | - Mia L. Huang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive #0358, La Jolla, California 92093-0358, United States
| | - Kamil Godula
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive #0358, La Jolla, California 92093-0358, United States
| |
Collapse
|
32
|
Yan Z, Qin H, Ren J, Qu X. Photocontrolled Multidirectional Differentiation of Mesenchymal Stem Cells on an Upconversion Substrate. Angew Chem Int Ed Engl 2018; 57:11182-11187. [DOI: 10.1002/anie.201803939] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Zhengqing Yan
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
- University of Chinese Academy of Sciences; Beijing 100039 China
| | - Hongshuang Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| |
Collapse
|
33
|
Yan Z, Qin H, Ren J, Qu X. Photocontrolled Multidirectional Differentiation of Mesenchymal Stem Cells on an Upconversion Substrate. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201803939] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Zhengqing Yan
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
- University of Chinese Academy of Sciences; Beijing 100039 China
| | - Hongshuang Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| |
Collapse
|
34
|
Chen X, Li J, Huang Y, Liu P, Fan Y. Insoluble Microenvironment Facilitating the Generation and Maintenance of Pluripotency. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:267-278. [PMID: 29327674 DOI: 10.1089/ten.teb.2017.0415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Induced pluripotent stem cells (iPSCs) hold enormous potential as a tool to generate cells for tissue engineering and regenerative medicine. Since the initial report of iPSCs in 2006, many different methods have been developed to enhance the safety and efficiency of this technology. Recent studies indicate that the extracellular signals can promote the production of iPSCs, and even replace the Yamanaka factors. Noticeably, abundant evidences suggest that the insoluble microenvironment, including the culture substrate and neighboring cells, directly regulates the expression of core pluripotency genes and the epigenetic modification of the chromatins, hence, impacts the reprogramming dynamics. These studies provide new strategies for developing safer and more efficient method for iPSC generation. In this review, we examine the publications addressing the insoluble extracellular microenvironment that boosts iPSC generation and self-renewal. We also discuss cell adhesion-mediated molecular mechanisms, through which the insoluble extracellular cues interplay with reprogramming.
Collapse
Affiliation(s)
- Xiaofang Chen
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Jiaqi Li
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
| | - Yan Huang
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
| | - Peng Liu
- 3 Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University , Beijing, China
| | - Yubo Fan
- 1 Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University , Beijing, China
- 2 Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University , Beijing, China
- 4 National Research Center for Rehabilitation Technical Aids , Beijing, China
| |
Collapse
|
35
|
Elastic polyurethane bearing pendant TGF-β1 affinity peptide for potential tissue engineering applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 83:67-77. [DOI: 10.1016/j.msec.2017.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 09/05/2017] [Accepted: 10/11/2017] [Indexed: 12/18/2022]
|
36
|
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
|
37
|
Vitillo L, Baxter M, Iskender B, Whiting P, Kimber SJ. Integrin-Associated Focal Adhesion Kinase Protects Human Embryonic Stem Cells from Apoptosis, Detachment, and Differentiation. Stem Cell Reports 2017; 7:167-76. [PMID: 27509133 PMCID: PMC4983079 DOI: 10.1016/j.stemcr.2016.07.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/08/2016] [Accepted: 07/08/2016] [Indexed: 11/25/2022] Open
Abstract
Human embryonic stem cells (hESCs) can be maintained in a fully defined niche on extracellular matrix substrates, to which they attach through integrin receptors. However, the underlying integrin signaling mechanisms, and their contribution to hESC behavior, are largely unknown. Here, we show that focal adhesion kinase (FAK) transduces integrin activation and supports hESC survival, substrate adhesion, and maintenance of the undifferentiated state. After inhibiting FAK kinase activity we show that hESCs undergo cell detachment-dependent apoptosis or differentiation. We also report deactivation of FAK downstream targets, AKT and MDM2, and upregulation of p53, all key players in hESC regulatory networks. Loss of integrin activity or FAK also induces cell aggregation, revealing a role in the cell-cell interactions of hESCs. This study provides insight into the integrin signaling cascade activated in hESCs and reveals in FAK a key player in the maintenance of hESC survival and undifferentiated state.
Collapse
Affiliation(s)
- Loriana Vitillo
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Melissa Baxter
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Banu Iskender
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Paul Whiting
- Pfizer Neusentis, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
| | - Susan J Kimber
- North West Embryonic Stem Cell Centre, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK.
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW Human pluripotent stem cells (hPSCs) are anchorage-dependent cells that can be cultured on a variety of matrices and express integrins and the machinery for integrin signaling. Until recently, there has been limited understanding of exactly how integrin signaling regulates pluripotent stem cell (PSC) behavior. This review summarizes our knowledge of how integrins and focal adhesion kinase (FAK) regulate different aspects of hPSC biology. RECENT FINDINGS The latest research suggests that mouse and human embryonic stem cells utilize similar integrin signaling players but with different biological outcomes, reflecting the known developmental difference in their pluripotent status. Notably, attachment cues via FAK signaling are crucial for hPSCs survival and pluripotency maintenance. FAK may be found cortically but also in the nucleus of hPSCs intersecting core pluripotency networks. SUMMARY Integrins and FAK have been consigned to the conventional role of cell adhesion receptor systems in PSCs. This review highlights data indicating that they are firmly integrated in pluripotency circuits, with implications for both research PSC culture and scale up and use in clinical applications.
Collapse
Affiliation(s)
- Loriana Vitillo
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, University of Manchester, Michael Smith Building, Oxford Rd, Manchester, M13 9PT UK
| | - Susan J. Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, University of Manchester, Michael Smith Building, Oxford Rd, Manchester, M13 9PT UK
| |
Collapse
|
39
|
Valdez J, Cook CD, Ahrens CC, Wang AJ, Brown A, Kumar M, Stockdale L, Rothenberg D, Renggli K, Gordon E, Lauffenburger D, White F, Griffith L. On-demand dissolution of modular, synthetic extracellular matrix reveals local epithelial-stromal communication networks. Biomaterials 2017; 130:90-103. [PMID: 28371736 DOI: 10.1016/j.biomaterials.2017.03.030] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/19/2017] [Accepted: 03/21/2017] [Indexed: 02/06/2023]
Abstract
Methods to parse paracrine epithelial-stromal communication networks are a vital need in drug development, as disruption of these networks underlies diseases ranging from cancer to endometriosis. Here, we describe a modular, synthetic, and dissolvable extracellular matrix (MSD-ECM) hydrogel that fosters functional 3D epithelial-stromal co-culture, and that can be dissolved on-demand to recover cells and paracrine signaling proteins intact for subsequent analysis. Specifically, synthetic polymer hydrogels, modified with cell-interacting adhesion motifs and crosslinked with peptides that include a substrate for cell-mediated proteolytic remodeling, can be rapidly dissolved by an engineered version of the microbial transpeptidase Sortase A (SrtA) if the crosslinking peptide includes a SrtA substrate motif and a soluble second substrate. SrtA-mediated dissolution affected only 1 of 31 cytokines and growth factors assayed, whereas standard protease degradation methods destroyed about half of these same molecules. Using co-encapsulated endometrial epithelial and stromal cells as one model system, we show that the dynamic cytokine and growth factor response of co-cultures to an inflammatory cue is richer and more nuanced when measured from SrtA-dissolved gel microenvironments than from the culture supernate. This system employs accessible, reproducible reagents and facile protocols; hence, has potential as a tool in identifying and validating therapeutic targets in complex diseases.
Collapse
Affiliation(s)
- Jorge Valdez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christi D Cook
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Caroline Chopko Ahrens
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alex J Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander Brown
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Manu Kumar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Linda Stockdale
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Rothenberg
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kasper Renggli
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Elizabeth Gordon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Forest White
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Linda Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
40
|
Varun D, Srinivasan GR, Tsai YH, Kim HJ, Cutts J, Petty F, Merkley R, Stephanopoulos N, Dolezalova D, Marsala M, Brafman DA. A robust vitronectin-derived peptide for the scalable long-term expansion and neuronal differentiation of human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs). Acta Biomater 2017; 48:120-130. [PMID: 27989923 DOI: 10.1016/j.actbio.2016.10.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 10/03/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022]
Abstract
Despite therapeutic advances, neurodegenerative diseases and disorders remain some of the leading causes of mortality and morbidity in the United States. Therefore, cell-based therapies to replace lost or damaged neurons and supporting cells of the central nervous system (CNS) are of great therapeutic interest. To that end, human pluripotent stem cell (hPSC) derived neural progenitor cells (hNPCs) and their neuronal derivatives could provide the cellular 'raw material' needed for regenerative medicine therapies for a variety of CNS disorders. In addition, hNPCs derived from patient-specific hPSCs could be used to elucidate the underlying mechanisms of neurodegenerative diseases and identify potential drug candidates. However, the scientific and clinical application of hNPCs requires the development of robust, defined, and scalable substrates for their long-term expansion and neuronal differentiation. In this study, we rationally designed a vitronectin-derived peptide (VDP) that served as an adhesive growth substrate for the long-term expansion of several hNPC lines. Moreover, VDP-coated surfaces allowed for the directed neuronal differentiation of hNPC at levels similar to cells differentiated on traditional extracellular matrix protein-based substrates. Overall, the ability of VDP to support the long-term expansion and directed neuronal differentiation of hNPCs will significantly advance the future translational application of these cells in treating injuries, disorders, and diseases of the CNS.
Collapse
|
41
|
Vas WJ, Shah M, Al Hosni R, Owen HC, Roberts SJ. Biomimetic strategies for fracture repair: Engineering the cell microenvironment for directed tissue formation. J Tissue Eng 2017; 8:2041731417704791. [PMID: 28491274 PMCID: PMC5406151 DOI: 10.1177/2041731417704791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/21/2017] [Indexed: 12/20/2022] Open
Abstract
Complications resulting from impaired fracture healing have major clinical implications on fracture management strategies. Novel concepts taken from developmental biology have driven research strategies towards the elaboration of regenerative approaches that can truly harness the complex cellular events involved in tissue formation and repair. Advances in polymer technology and a better understanding of naturally derived scaffolds have given rise to novel biomaterials with an increasing ability to recapitulate native tissue environments. This coupled with advances in the understanding of stem cell biology and technology has opened new avenues for regenerative strategies with true clinical translatability. These advances have provided the impetus to develop alternative approaches to enhance the fracture repair process. We provide an update on these advances, with a focus on the development of novel biomimetic approaches for bone regeneration and their translational potential.
Collapse
Affiliation(s)
- Wollis J Vas
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| | - Mittal Shah
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| | - Rawiya Al Hosni
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| | - Helen C Owen
- Department of Natural Sciences, School of Science & Technology, Middlesex University, London, UK
| | - Scott J Roberts
- Department of Materials & Tissue, Institute of Orthopaedics & Musculoskeletal Science, University College London, Stanmore, UK
| |
Collapse
|
42
|
Tan Y, Wood AR, Jia Q, Zhou W, Luo J, Yang F, Chen J, Chen J, Sun J, Seong J, Tajik A, Singh R, Wang N. Soft matrices downregulate FAK activity to promote growth of tumor-repopulating cells. Biochem Biophys Res Commun 2016; 483:456-462. [PMID: 28007596 DOI: 10.1016/j.bbrc.2016.12.122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 12/19/2016] [Indexed: 12/21/2022]
Abstract
Tumor-repopulating cells (TRCs) are a tumorigenic sub-population of cancer cells that drives tumorigenesis. We have recently reported that soft fibrin matrices maintain TRC growth by promoting histone 3 lysine 9 (H3K9) demethylation and Sox2 expression and that Cdc42 expression influences H3K9 methylation. However, the underlying mechanisms of how soft matrices induce H3K9 demethylation remain elusive. Here we find that TRCs exhibit lower focal adhesion kinase (FAK) and H3K9 methylation levels in soft fibrin matrices than control melanoma cells on 2D rigid substrates. Silencing FAK in control melanoma cells decreases H3K9 methylation, whereas overexpressing FAK in tumor-repopulating cells enhances H3K9 methylation. Overexpressing Cdc42 or RhoA in the presence of FAK knockdown restores H3K9 methylation levels. Importantly, silencing FAK, Cdc42, or RhoA promotes Sox2 expression and proliferation of control melanoma cells in stiff fibrin matrices, whereas overexpressing each gene suppresses Sox2 expression and reduces growth of TRCs in soft but not in stiff fibrin matrices. Our findings suggest that low FAK mediated by soft fibrin matrices downregulates H3K9 methylation through reduction of Cdc42 and RhoA and promotes TRC growth.
Collapse
Affiliation(s)
- Youhua Tan
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Interdisciplinary Division of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Kowloon, Hong Kong SAR, China
| | - Adam Richard Wood
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Qiong Jia
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Wenwen Zhou
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Junyu Luo
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Fang Yang
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Junwei Chen
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Junjian Chen
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Jian Sun
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jihye Seong
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 136-791, South Korea
| | - Arash Tajik
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Rishi Singh
- Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Ning Wang
- Laboratory for Cell Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China; Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
43
|
Mutoji K, Singh A, Nguyen T, Gildersleeve H, Kaucher AV, Oatley MJ, Oatley JM, Velte EK, Geyer CB, Cheng K, McCarrey JR, Hermann BP. TSPAN8 Expression Distinguishes Spermatogonial Stem Cells in the Prepubertal Mouse Testis. Biol Reprod 2016; 95:117. [PMID: 27733379 PMCID: PMC5315423 DOI: 10.1095/biolreprod.116.144220] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/13/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022] Open
Abstract
Precise separation of spermatogonial stem cells (SSCs) from progenitor spermatogonia that lack stem cell activity and are committed to differentiation remains a challenge. To distinguish between these spermatogonial subtypes, we identified genes that exhibited bimodal mRNA levels at the single-cell level among undifferentiated spermatogonia from Postnatal Day 6 mouse testes, including Tspan8, Epha2, and Pvr, each of which encode cell surface proteins useful for cell selection. Transplantation studies provided definitive evidence that a TSPAN8-high subpopulation is enriched for SSCs. RNA-seq analyses identified genes differentially expressed between TSPAN8-high and -low subpopulations that clustered into multiple biological pathways potentially involved in SSC renewal or differentiation, respectively. Methyl-seq analysis identified hypomethylated domains in the promoters of these genes in both subpopulations that colocalized with peaks of histone modifications defined by ChIP-seq analysis. Taken together, these results demonstrate functional heterogeneity among mouse undifferentiated spermatogonia and point to key biological characteristics that distinguish SSCs from progenitor spermatogonia.
Collapse
Affiliation(s)
- Kazadi Mutoji
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Anukriti Singh
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Thu Nguyen
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Heidi Gildersleeve
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
- Genomics Core Facility, University of Texas at San Antonio, San Antonio, Texas
| | - Amy V Kaucher
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Melissa J Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Ellen K Velte
- Department of Anatomy and Cell Biology and East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology and East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Keren Cheng
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - John R McCarrey
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Brian P Hermann
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas
- Genomics Core Facility, University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
44
|
Jia J, Coyle RC, Richards DJ, Berry CL, Barrs RW, Biggs J, James Chou C, Trusk TC, Mei Y. Development of peptide-functionalized synthetic hydrogel microarrays for stem cell and tissue engineering applications. Acta Biomater 2016; 45:110-120. [PMID: 27612960 PMCID: PMC5146757 DOI: 10.1016/j.actbio.2016.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 10/21/2022]
Abstract
Synthetic polymer microarray technology holds remarkable promise to rapidly identify suitable biomaterials for stem cell and tissue engineering applications. However, most of previous microarrayed synthetic polymers do not possess biological ligands (e.g., peptides) to directly engage cell surface receptors. Here, we report the development of peptide-functionalized hydrogel microarrays based on light-assisted copolymerization of poly(ethylene glycol) diacrylates (PEGDA) and methacrylated-peptides. Using solid-phase peptide/organic synthesis, we developed an efficient route to synthesize methacrylated-peptides. In parallel, we identified PEG hydrogels that effectively inhibit non-specific cell adhesion by using PEGDA-700 (M. W.=700) as a monomer. The combined use of these chemistries enables the development of a powerful platform to prepare peptide-functionalized PEG hydrogel microarrays. Additionally, we identified a linker composed of 4 glycines to ensure sufficient exposure of the peptide moieties from hydrogel surfaces. Further, we used this system to directly compare cell adhesion abilities of several related RGD peptides: RGD, RGDS, RGDSG and RGDSP. Finally, we combined the peptide-functionalized hydrogel technology with bioinformatics to construct a library composed of 12 different RGD peptides, including 6 unexplored RGD peptides, to develop culture substrates for hiPSC-derived cardiomyocytes (hiPSC-CMs), a cell type known for poor adhesion to synthetic substrates. 2 out of 6 unexplored RGD peptides showed substantial activities to support hiPSC-CMs. Among them, PMQKMRGDVFSP from laminin β4 subunit was found to support the highest adhesion and sarcomere formation of hiPSC-CMs. With bioinformatics, the peptide-functionalized hydrogel microarrays accelerate the discovery of novel biological ligands to develop biomaterials for stem cell and tissue engineering applications. STATEMENT OF SIGNIFICANCE In this manuscript, we described the development of a robust approach to prepare peptide-functionalized synthetic hydrogel microarrays. Combined with bioinformatics, this technology enables us to rapidly identify novel biological ligands for the development of the next generation of functional biomaterials for stem cell and tissue engineering applications.
Collapse
Affiliation(s)
- Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert C Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | | | - Ryan Walker Barrs
- College of Engineering and Computing, University of South Carolina, Columbia, SC 29208, USA
| | - Joshua Biggs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - C James Chou
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Thomas C Trusk
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
45
|
Biological Effects of Culture Substrates on Human Pluripotent Stem Cells. Stem Cells Int 2016; 2016:5380560. [PMID: 27656216 PMCID: PMC5021488 DOI: 10.1155/2016/5380560] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/04/2016] [Indexed: 01/03/2023] Open
Abstract
In recent years, as human pluripotent stem cells (hPSCs) have been commonly cultured in feeder-free conditions, a number of cell culture substrates have been applied or developed. However, the functional roles of these substrates in maintaining hPSC self-renewal remain unclear. Here in this review, we summarize the types of these substrates and their effect on maintaining hPSC self-renewal. Endogenous extracellular matrix (ECM) protein expression has been shown to be crucial in maintaining hPSC self-renewal. These ECM molecules interact with integrin cell-surface receptors and transmit their cellular signaling. We discuss the possible effect of integrin-mediated signaling pathways on maintaining hPSC self-renewal. Activation of integrin-linked kinase (ILK), which transmits ECM-integrin signaling to AKT (also known as protein kinase B), has been shown to be critical in maintaining hPSC self-renewal. Also, since naïve pluripotency has been widely recognized as an alternative pluripotent state of hPSCs, we discuss the possible effects of culture substrates and integrin signaling on naïve hPSCs based on the studies of mouse embryonic stem cells. Understanding the role of culture substrates in hPSC self-renewal and differentiation enables us to control hPSC behavior precisely and to establish scalable or microfabricated culture technologies for regenerative medicine and drug development.
Collapse
|
46
|
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
|
47
|
Coyle R, Jia J, Mei Y. Polymer microarray technology for stem cell engineering. Acta Biomater 2016; 34:60-72. [PMID: 26497624 PMCID: PMC4811723 DOI: 10.1016/j.actbio.2015.10.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/10/2015] [Accepted: 10/19/2015] [Indexed: 12/12/2022]
Abstract
Stem cells hold remarkable promise for applications in tissue engineering and disease modeling. During the past decade, significant progress has been made in developing soluble factors (e.g., small molecules and growth factors) to direct stem cells into a desired phenotype. However, the current lack of suitable synthetic materials to regulate stem cell activity has limited the realization of the enormous potential of stem cells. This can be attributed to a large number of materials properties (e.g., chemical structures and physical properties of materials) that can affect stem cell fate. This makes it challenging to design biomaterials to direct stem cell behavior. To address this, polymer microarray technology has been developed to rapidly identify materials for a variety of stem cell applications. In this article, we summarize recent developments in polymer array technology and their applications in stem cell engineering. STATEMENT OF SIGNIFICANCE Stem cells hold remarkable promise for applications in tissue engineering and disease modeling. In the last decade, significant progress has been made in developing chemically defined media to direct stem cells into a desired phenotype. However, the current lack of the suitable synthetic materials to regulate stem cell activities has been limiting the realization of the potential of stem cells. This can be attributed to the number of variables in material properties (e.g., chemical structures and physical properties) that can affect stem cells. Polymer microarray technology has shown to be a powerful tool to rapidly identify materials for a variety of stem cell applications. Here we summarize recent developments in polymer array technology and their applications in stem cell engineering.
Collapse
Affiliation(s)
- Robert Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
48
|
Wrighton PJ, Kiessling LL. Forces of Change: Mechanics Underlying Formation of Functional 3D Organ Buds. Cell Stem Cell 2016; 16:453-4. [PMID: 25957897 DOI: 10.1016/j.stem.2015.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
3D organ buds that can recapitulate organ function have myriad applications for regenerative and personalized medicine. Here, Takebe et al. (2015) describe a generalized method for organ bud formation, demonstrating that mechanosensitive mesenchymal stem cells drive condensation of heterotypic cell mixtures to create buds from diverse organs.
Collapse
Affiliation(s)
- Paul J Wrighton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Laura L Kiessling
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53792, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53792, USA.
| |
Collapse
|
49
|
Crowder SW, Leonardo V, Whittaker T, Papathanasiou P, Stevens MM. Material Cues as Potent Regulators of Epigenetics and Stem Cell Function. Cell Stem Cell 2016; 18:39-52. [PMID: 26748755 PMCID: PMC5409508 DOI: 10.1016/j.stem.2015.12.012] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Biophysical signals act as potent regulators of stem cell function, lineage commitment, and epigenetic status. In recent years, synthetic biomaterials have been used to study a wide range of outside-in signaling events, and it is now well appreciated that material cues modulate the epigenome. Here, we review the role of extracellular signals in guiding stem cell behavior via epigenetic regulation, and we stress the role of physicochemical material properties as an often-overlooked modulator of intracellular signaling. We also highlight promising new research tools for ongoing interrogation of the stem cell-material interface.
Collapse
Affiliation(s)
- Spencer W Crowder
- Department of Materials, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Institute for Biomedical Engineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
| | - Vincent Leonardo
- Department of Materials, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Institute for Biomedical Engineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
| | - Thomas Whittaker
- Department of Materials, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Institute for Biomedical Engineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
| | - Peter Papathanasiou
- Department of Materials, Imperial College London, Prince Consort Road, London SW7 2AZ, UK
| | - Molly M Stevens
- Department of Materials, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Department of Bioengineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK; Institute for Biomedical Engineering, Imperial College London, Prince Consort Road, London SW7 2AZ, UK.
| |
Collapse
|
50
|
Grim JC, Marozas IA, Anseth KS. Thiol-ene and photo-cleavage chemistry for controlled presentation of biomolecules in hydrogels. J Control Release 2015; 219:95-106. [PMID: 26315818 DOI: 10.1016/j.jconrel.2015.08.040] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
Hydrogels have emerged as promising scaffolds in regenerative medicine for the delivery of biomolecules to promote healing. However, increasing evidence suggests that the context that biomolecules are presented to cells (e.g., as soluble verses tethered signals) can influence their bioactivity. A common approach to deliver biomolecules in hydrogels involves physically entrapping them within the network, such that they diffuse out over time to the surrounding tissues. While simple and versatile, the release profiles in such system are highly dependent on the molecular weight of the entrapped molecule relative to the network structure, and it can be difficult to control the release of two different signals at independent rates. In some cases, supraphysiologically high loadings are used to achieve therapeutic local concentrations, but uncontrolled release can then cause deleterious off-target side effects. In vivo, many growth factors and cytokines are stored in the extracellular matrix (ECM) and released on demand as needed during development, growth, and wound healing. Thus, emerging strategies in biomaterial chemistry have focused on ways to tether or sequester biological signals and engineer these bioactive scaffolds to signal to delivered cells or endogenous cells. While many strategies exist to achieve tethering of peptides, protein, and small molecules, this review focuses on photochemical methods, and their usefulness as a mild reaction that proceeds with fast kinetics in aqueous solutions and at physiological conditions. Photo-click and photo-caging methods are particularly useful because one can direct light to specific regions of the hydrogel to achieve spatial patterning. Recent methods have even demonstrated reversible introduction of biomolecules to mimic the dynamic changes of native ECM, enabling researchers to explore how the spatial and dynamic context of biomolecular signals influences important cell functions. This review will highlight how two photochemical methods have led to important advances in the tissue regeneration community, namely the thiol-ene photo-click reaction for bioconjugation and photocleavage reactions that allow for the removal of protecting groups. Specific examples will be highlighted where these methodologies have been used to engineer hydrogels that control and direct cell function with the aim of inspiring their use in regenerative medicine.
Collapse
Affiliation(s)
- Joseph C Grim
- Howard Hughes Medical Institute, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Ian A Marozas
- Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Kristi S Anseth
- Howard Hughes Medical Institute, University of Colorado at Boulder, Boulder, CO 80309, USA; Department of Chemical and Biological Engineering, University of Colorado at Boulder, Boulder, CO 80309, USA; BioFrontiers Institute, University of Colorado at Boulder, Boulder, CO 80309, USA.
| |
Collapse
|