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
|
Baude JA, Li MD, Jackson SM, Sharma A, Walter DI, Stowers RS. Engineered basement membrane mimetic hydrogels to study mammary epithelial morphogenesis and invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640825. [PMID: 40093166 PMCID: PMC11908212 DOI: 10.1101/2025.02.28.640825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Reconstituted basement membrane (rBM) products like Matrigel are widely used in 3D culture models of epithelial tissues and cancer. However, their utility is hindered by key limitations, including batch variability, xenogenic contaminants, and a lack of tunability. To address these challenges, we engineered a 3D basement membrane (eBM) matrix by conjugating defined extracellular matrix (ECM) adhesion peptides (IKVAV, YIGSR, RGD) to an alginate hydrogel network with precisely tunable stiffness and viscoelasticity. We optimized the mechanical and biochemical properties of the engineered basement membranes (eBMs) to support mammary acinar morphogenesis in MCF10A cells, similar to rBM. We found that IKVAV-modified, fast-relaxing (τ1/2 = 30-150 s), and soft (E = 200 Pa) eBMs best promoted polarized acinar structures. Clusters became invasive and lost polarity only when the IKVAV-modified eBM exhibited both similar stiffness to a malignant breast tumor (E = 4000 Pa) and slow stress relaxation (τ1/2 = 600-1100 s). Notably, tumor-like stiffness alone was not sufficient to drive invasion in fast stress relaxing matrices modified with IKVAV. In contrast, RGD-modified matrices promoted a malignant phenotype regardless of mechanical properties. We also utilized this system to interrogate the mechanism driving acinar and tumorigenic phenotypes in response to microenvironmental parameters. A balance in activity between β1- and β4-integrins was observed in the context of IKVAV-modified eBMs, prompting further investigation into the downstream mechanisms. We found differences in hemidesmosome formation and production of endogenous laminin in response to peptide type, stress relaxation, and stiffness. We also saw that inhibiting either focal adhesion kinase or hemidesmosome signaling in IKVAV eBMs prevented acinus formation. This eBM matrix is a powerful, reductionist, xenogenic-free system, offering a robust platform for both fundamental research and translational applications in tissue engineering and disease modeling.
Collapse
Affiliation(s)
- Jane A Baude
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
| | - Megan D Li
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
| | - Sabrina M Jackson
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
| | - Abhishek Sharma
- University of California, Santa Barbara, Department of Mechanical Engineering
| | - Daniella I Walter
- University of California, Santa Barbara, Department of Mechanical Engineering
| | - Ryan S Stowers
- University of California, Santa Barbara, Department of Molecular, Cellular, and Developmental Biology
- University of California, Santa Barbara, Department of Mechanical Engineering
- University of California, Santa Barbara, Department of Bioengineering
| |
Collapse
|
3
|
Truong AT, Lee SJ, Hamada K, Kiyomi A, Guo H, Yamada Y, Kikkawa Y, Okamoto CT, Nomizu M, MacKay JA. Synergy between Laminin-Derived Elastin-like Polypeptides (LELPs) Optimizes Cell Spreading. Biomacromolecules 2024; 25:4001-4013. [PMID: 38814168 DOI: 10.1021/acs.biomac.4c00144] [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: 05/31/2024]
Abstract
A major component of the extracellular matrix (ECM), laminins, modulates cells via diverse receptors. Their fragments have emerging utility as components of "ECM-mimetics" optimized to promote cell-based therapies. Recently, we reported that a bioactive laminin peptide known as A99 enhanced cell binding and spreading via fusion to an elastin-like polypeptide (ELP). The ELP "handle" serves as a rapid, noncovalent strategy to concentrate bioactive peptide mixtures onto a surface. We now report that this strategy can be further generalized across an expanded panel of additional laminin-derived elastin-like polypeptides (LELPs). A99 (AGTFALRGDNPQG), A2G80 (VQLRNGFPYFSY), AG73 (RKRLQVQLSIRT), and EF1m (LQLQEGRLHFMFD) all promote cell spreading while showing morphologically distinct F-actin formation. Equimolar mixtures of A99:A2G80-LELPs have synergistic effects on adhesion and spreading. Finally, three of these ECM-mimetics promote the neurite outgrowth of PC-12 cells. The evidence presented here demonstrates the potential of ELPs to deposit ECM-mimetics with applications in regenerative medicine, cell therapy, and tissue engineering.
Collapse
Affiliation(s)
- Anh T Truong
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Shin-Jae Lee
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Anna Kiyomi
- Department of Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Curtis T Okamoto
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, United States
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, United States
| |
Collapse
|
4
|
Yamada Y. Characterization of Novel Cell-Adhesive Peptides for Biomaterial Development. Biol Pharm Bull 2024; 47:1072-1078. [PMID: 38825460 DOI: 10.1248/bpb.b24-00030] [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: 06/04/2024]
Abstract
In previous studies, my group developed cell-adhesive peptide-polysaccharide complexes as biomaterials for tissue engineering. Having a wide variety of cell-adhesive peptides is important as the biological functions of peptide-polysaccharide complexes are highly dependent on the biological activity of peptides. This paper reviews the biological activities of two types of recently characterized cell-adhesive peptides. The first is peptides rich in basic amino acids originating from octaarginine. We analyzed the relationships between the amino acid composition of basic peptides and cell adhesion, elongation, and proliferation and identified the most suitable peptide for cell culture. The second was arginine-glycine-aspartic acid (RGD)-containing peptides that promote the adhesion of induced pluripotent stem cells (iPSCs). We identified the RGD-surrounding sequences necessary for iPSC adhesion, clarified the underlying mechanism, and improved cell adhesion by modifying the structure-activity relationships. The novel cell-adhesive peptides identified in our previous studies may aid in the development of novel peptide-based biomaterials.
Collapse
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
5
|
Wang D, Hedayati M, Stuart JD, Madruga LYC, Popat KC, Snow CD, Kipper MJ. Ligand Presentation Inside Protein Crystal Nanopores: Tunable Interfacial Adhesion Noncovalently Modulates Cell Attachment. MATERIALS TODAY. NANO 2023; 24:100432. [PMID: 38370345 PMCID: PMC10871713 DOI: 10.1016/j.mtnano.2023.100432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Protein crystals with sufficiently large solvent pores can non-covalently adsorb polymers in the pores. In principle, if these polymers contain cell adhesion ligands, the polymer-laden crystals could present ligands to cells with tunable adhesion strength. Moreover, porous protein crystals can store an internal ligand reservoir, so that the surface can be replenished. In this study, we demonstrate that poly(ethylene glycol) terminated with a cyclic cell adhesion ligand peptide (PEG-RGD) can be loaded into porous protein crystals by diffusion. Through atomic force microscopy (AFM), force-distance correlations of the mechanical interactions between activated AFM tips and protein crystals were precisely measured. The activation of AFM tips allows the tips to interact with PEG-RGD that was pre-loaded in the protein crystal nanopores, mimicking how a cell might attach to and pull on the ligand through integrin receptors. The AFM experiments also simultaneously reveal the detailed morphology of the buffer-immersed nanoporous protein crystal surface. We also show that porous protein crystals (without and with loaded PEG-RGD) serve as suitable substrates for attachment and spreading of adipose-derived stem cells. This strategy can be used to design surfaces that non-covalently present multiple different ligands to cells with tunable adhesive strength for each ligand, and with an internal reservoir to replenish the precisely defined crystalline surface.
Collapse
Affiliation(s)
- Dafu Wang
- Department of Chemical and Biological Engineering, Colorado State University, 1370Campus Delivery, Fort Collins, CO 80523, U.S.A
- School of Advanced Materials Discovery, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, U.S.A
| | - Mohammadhasan Hedayati
- Department of Chemical and Biological Engineering, Colorado State University, 1370Campus Delivery, Fort Collins, CO 80523, U.S.A
| | - Julius D Stuart
- Department of Chemistry, Colorado State University, 1872 Campus Delivery, FortCollins, CO 80523, U.S.A
| | - Liszt Y C Madruga
- Department of Chemical and Biological Engineering, Colorado State University, 1370Campus Delivery, Fort Collins, CO 80523, U.S.A
| | - Ketul C Popat
- Department of Chemical and Biological Engineering, Colorado State University, 1370Campus Delivery, Fort Collins, CO 80523, U.S.A
| | - Christopher D Snow
- Department of Chemical and Biological Engineering, Colorado State University, 1370Campus Delivery, Fort Collins, CO 80523, U.S.A
- School of Advanced Materials Discovery, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, U.S.A
- Department of Chemistry, Colorado State University, 1872 Campus Delivery, FortCollins, CO 80523, U.S.A
- School of Biomedical Engineering, Colorado State University, 1301 Campus Delivery, Fort Collins, CO 80523, U.S.A
| | - Matt J Kipper
- Department of Chemical and Biological Engineering, Colorado State University, 1370Campus Delivery, Fort Collins, CO 80523, U.S.A
- School of Advanced Materials Discovery, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, U.S.A
- School of Biomedical Engineering, Colorado State University, 1301 Campus Delivery, Fort Collins, CO 80523, U.S.A
| |
Collapse
|
6
|
Hiroshima K, Sakata N, Konogami T, Shimamoto S, Hidaka Y. The Cell Adhesion Activity of the Joining Peptide of Proopiomelanocortin. Molecules 2023; 28:7754. [PMID: 38067484 PMCID: PMC10707936 DOI: 10.3390/molecules28237754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Proopiomelanocortin (POMC) is a precursor protein of several peptide hormones, such as ACTH and β-endorphin. Almost all of the peptide hormones in POMC have been drastically investigated in terms of their biological activities. However, the biological activity of the joining peptide region (JP) in POMC is unknown. Therefore, to explore the biological activity of JP, sequence analyses of mammalian POMC were performed. We found an -Arg-Gly-Asp- (RGD) motif in several mammalian species, such as porcine, suggesting that JP has cell adhesion activity. To validate this hypothesis, the cell adhesion activities of the synthetic porcine JP peptides were examined using 293T cells. Cell adhesions were observed in a concentration-dependent manner of the JP peptides. In addition, the JP peptide competitively inhibited cell adhesion to the POMC-coated plates. Moreover, the cell adhesion activity of the joining peptide was inhibited by the addition of EDTA, indicating that the JP peptide mediates the cell adhesion activity via a receptor protein, integrin. Interestingly, a human JP peptide, which possesses an -Arg-Ser-Asp- (RSD) sequence in place of the RGD sequence, exhibited a higher ability in the cell adhesion activity than that of the porcine JP peptide, suggesting that the cell adhesion activity of the joining peptide is developed during the molecular evolution of POMC. In conclusion, our results reveal that the joining peptide in POMC plays an important role during cell adhesion and provide useful information related to signal transduction of nerve peptide hormones derived from POMC.
Collapse
Affiliation(s)
| | | | | | - Shigeru Shimamoto
- Faculty of Science and Engineering, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan
| | - Yuji Hidaka
- Faculty of Science and Engineering, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|
7
|
Wang T, Yu T, Tsai CY, Hong ZY, Chao WH, Su YS, Subbiah SK, Renuka RR, Hsu ST, Wu GJ, Higuchi A. Xeno-free culture and proliferation of hPSCs on 2D biomaterials. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:63-107. [PMID: 37678982 DOI: 10.1016/bs.pmbts.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Human pluripotent stem cells (human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs)) have unlimited proliferative potential, whereas adult stem cells such as bone marrow-derived stem cells and adipose-derived stem cells have problems with aging. When hPSCs are intended to be cultured on feeder-free or xeno-free conditions without utilizing mouse embryonic fibroblasts or human fibroblasts, they cannot be cultured on conventional tissue culture polystyrene dishes, as adult stem cells can be cultured but should be cultivated on material surfaces grafted or coated with (a) natural or recombinant extracellular matrix (ECM) proteins, (b) ECM protein-derived peptides and specific synthetic polymer surfaces in xeno-free and/or chemically defined conditions. This review describes current developing cell culture biomaterials for the proliferation of hPSCs while maintaining the pluripotency and differentiation potential of the cells into 3 germ layers. Biomaterials for the cultivation of hPSCs without utilizing a feeder layer are essential to decrease the risk of xenogenic molecules, which contributes to the potential clinical usage of hPSCs. ECM proteins such as human recombinant vitronectin, laminin-511 and laminin-521 have been utilized instead of Matrigel for the feeder-free cultivation of hPSCs. The following biomaterials are also discussed for hPSC cultivation: (a) decellularized ECM, (b) peptide-grafted biomaterials derived from ECM proteins, (c) recombinant E-cadherin-coated surface, (d) polysaccharide-immobilized surface, (e) synthetic polymer surfaces with and without bioactive sites, (f) thermoresponsive polymer surfaces with and without bioactive sites, and (g) synthetic microfibrous scaffolds.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Tao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Chang-Yen Tsai
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Zhao-Yu Hong
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Wen-Hui Chao
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Yi-Shuo Su
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Remya Rajan Renuka
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| | - Shih-Tien Hsu
- Department of Internal Medicine, Landseed International Hospital, Pingjen City, Taoyuan, Taiwan
| | - Gwo-Jang Wu
- Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
8
|
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: 10] [Impact Index Per Article: 5.0] [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
|
9
|
Guo Y, Cheng N, Sun H, Hou J, Zhang Y, Wang D, Zhang W, Chen Z. Advances in the development and optimization strategies of the hemostatic biomaterials. Front Bioeng Biotechnol 2023; 10:1062676. [PMID: 36714615 PMCID: PMC9873964 DOI: 10.3389/fbioe.2022.1062676] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/29/2022] [Indexed: 01/12/2023] Open
Abstract
Most injuries are accompanied by acute bleeding. Hemostasis is necessary to relieve pain and reduce mortality in these accidents. In recent years, the traditional hemostatic materials, including inorganic, protein-based, polysaccharide-based and synthetic materials have been widely used in the clinic. The most prominent of these are biodegradable collagen sponges (Helistat®, United States), gelatin sponges (Ethicon®, SURGIFOAM®, United States), chitosan (AllaQuixTM, ChitoSAMTM, United States), cellulose (Tabotamp®, SURGICEL®, United States), and the newly investigated extracellular matrix gels, etc. Although these materials have excellent hemostatic properties, they also have their advantages and disadvantages. In this review, the performance characteristics, hemostatic effects, applications and hemostatic mechanisms of various biomaterials mentioned above are presented, followed by several strategies to improve hemostasis, including modification of single materials, blending of multiple materials, design of self-assembled peptides and their hybrid materials. Finally, the exploration of more novel hemostatic biomaterials and relative coagulation mechanisms will be essential for future research on hemostatic methods.
Collapse
Affiliation(s)
- Yayuan Guo
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China
| | - Nanqiong Cheng
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China
| | - Hongxiao Sun
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China
| | - Jianing Hou
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China
| | - Yuchen Zhang
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China
| | - Du Wang
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China
| | - Wei Zhang
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China,School of Medicine, Northwest University, Xi’an, Shaanxi Province, China
| | - Zhuoyue Chen
- Faculty of Life Science, Northwest University, Xi’an, Shaanxi Province, China,School of Medicine, Northwest University, Xi’an, Shaanxi Province, China,*Correspondence: Zhuoyue Chen,
| |
Collapse
|
10
|
Huang WY, Otaka A, Fujita S, Yamaoka T. Bioactive peptide-bearing polylactic acid fibers as a model of the brain tumor-stimulating microenvironment. Polym J 2023. [DOI: 10.1038/s41428-022-00743-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
11
|
Structural Requirement of hA5G18 Peptide (DDFVFYVGGYPS) from Laminin α5 Chain for Amyloid-like Fibril Formation and Cell Adhesion. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196610. [PMID: 36235147 PMCID: PMC9573118 DOI: 10.3390/molecules27196610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022]
Abstract
The hA5G18 peptide (DDFVFYVGGYPS) identified from the human laminin α5 chain G domain promotes cell attachment and spreading when directly coated on a plastic plate, but does not show activity when it is conjugated on a chitosan matrix. Here, we focused on the structural requirement of hA5G18 for activity. hA5G18 was stained with Congo red and formed amyloid-like fibrils. A deletion analysis of hA5G18 revealed that FVFYV was a minimum active sequence for the formation of amyloid-like fibrils, but FVFYV did not promote cell attachment. Next, we designed functional fibrils using FVFYV as a template for amyloid-like fibrils. When we conjugated an integrin binding sequence Arg-Gly-Asp (RGD) to the FVFYV peptide with Gly-Gly (GG) as a spacer, FVFYVGGRGD promoted cell attachment in a plate coat assay, but a negative control sequence RGE conjugated peptide, FVFYVGGRGE, also showed activity. However, when the peptides were conjugated to Sepharose beads, the FVFYVGGRGD beads showed cell attachment activity, but the FVFYVGGRGE beads did not. These results suggest that RGD and RGE similarly contribute to cell attachment activity in amyloid-like fibrils, but only RGD contributes the activity on the Sepharose beads. Further, we conjugated a basic amino acid (Arg, Lys, and His) to the FVFYV peptide. Arg or Lys-conjugated FVFYV peptides, FVFYVGGR and FVFYVGGK, showed cell attachment activity when they were coated on a plate, but a His-conjugated FVFYV peptide FVFYVGGH did not show activity. None of the basic amino acid-conjugated peptides showed cell attachment in a Sepharose bead assay. The cell attachment and spreading on FVFYVGGR and FVFYVGGK were inhibited by an anti-integrin β1 antibody. These results suggest that the Arg and Lys residues play critical roles in the interaction with integrins in amyloid-like fibrils. FVFYV is useful to use as a template for amyloid-like fibrils and to develop multi-functional biomaterials.
Collapse
|
12
|
Wilson KL, Pérez SCL, Naffaa MM, Kelly SH, Segura T. Stoichiometric Post-Modification of Hydrogel Microparticles Dictates Neural Stem Cell Fate in Microporous Annealed Particle Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201921. [PMID: 35731241 PMCID: PMC9645378 DOI: 10.1002/adma.202201921] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/07/2022] [Indexed: 05/16/2023]
Abstract
Microporous annealed particle (MAP) scaffolds are generated from assembled hydrogel microparticles (microgels). It has been previously demonstrated that MAP scaffold are porous, biocompatible, and recruit neural progenitor cells (NPCs) to the stroke cavity after injection into the stroke core. Here, the goal is to study NPC fate inside MAP scaffolds in vitro. To create plain microgels that can later be converted to contain different types of bioactivities, the inverse electron-demand Diels-Alder reaction between tetrazine and norbornene is utilized, which allows the post-modification of plain microgels stoichiometrically. As a result of adhesive peptide attachment, NPC spreading leads to contractile force generation which can be recorded by tracking microgel displacement. Alternatively, non-adhesive peptide integration results in neurosphere formation that grows within the void space of MAP scaffolds. Although the formed neurospheres do not impose a contractile force on the scaffolds, they are seen to continuously transverse the scaffolds. It is concluded that MAP scaffolds can be engineered to either promote neurogenesis or enhance stemness depending on the chosen post-modifications of the microgels, which can be key in modulating their phenotypes in various applications in vivo.
Collapse
Affiliation(s)
- Katrina L Wilson
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708-0281, USA
| | - Sasha Cai Lesher Pérez
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, Building 28, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA
| | - Moawiah M Naffaa
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Psychology and Neuroscience, Duke University, Durham, NC, 27708, USA
| | - Sean H Kelly
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708-0281, USA
| | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708-0281, USA
- Department of Neurology, Duke University, Durham, NC, 27708-0281, USA
- Department of Dermatology, Duke University, Durham, NC, 27708-0281, USA
| |
Collapse
|
13
|
Ravikrishnan A, Fowler EW, Stuffer AJ, Jia X. Hydrogel-Supported, Engineered Model of Vocal Fold Epithelium. ACS Biomater Sci Eng 2021; 7:4305-4317. [PMID: 33635635 DOI: 10.1021/acsbiomaterials.0c01741] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is a critical need for the establishment of an engineered model of the vocal fold epithelium that can be used to gain understanding of its role in vocal fold health, disease, and facilitate the development of new treatment options. Toward this goal, we isolated primary vocal fold epithelial cells (VFECs) from healthy porcine larynxes and used them within passage 3. Culture-expanded VFECs expressed the suprabasal epithelial marker cytokeratin 13 and intercellular junctional proteins occludin, E-cadherin, and zonula occludens-1. To establish the engineered model, we cultured VFECs on a hyaluronic acid-derived synthetic basement membrane displaying fibronectin-derived integrin-binding peptide (RGDSP) and/or laminin 111-derived syndecan-binding peptide AG73 (RKRLQVQLSIRT). Our results show that matrix stiffness and composition cooperatively regulate the adhesion, proliferation, and stratification of VFECs. Cells cultured on hydrogels with physiological stiffness (elastic shear modulus, G' = 1828 Pa) adopted a cobblestone morphology with close cell-cell contacts, whereas those on softer matrices (G' = 41 Pa) were spindle shaped with extensive intracellular stress fibers. The development of stratified epithelium with proliferating basal cells and additional (1-2) suprabasal layers requires the presence of both RGDSP and AG73 peptide signals. Supplementation of cytokines produced by vimentin positive primary porcine vocal fold fibroblasts in the VFEC culture led to the establishment of 4-5 distinct cell layers. The engineered vocal fold epithelium resembled native tissue morphologically; expressed cytokeratin 13, mucin 1, and tight/adherens junction markers; and secreted basement membrane proteins collagen IV and laminin 5. Collectively, our results demonstrate that stiffness matching, cell-matrix engagement, and paracrine signaling cooperatively contribute to the stratification of VFECs. The engineered epithelium can be used as a versatile tool for investigations of genetic and molecular mechanisms in vocal fold health and disease.
Collapse
Affiliation(s)
- Anitha Ravikrishnan
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Eric W Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Alexander J Stuffer
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States.,Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States.,Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States.,Delaware Biotechnology Institute, University of Delaware, Newark, Delaware 19711, United States
| |
Collapse
|
14
|
Mrówczyńska E, Mazur AJ. Integrin-Linked Kinase (ILK) Plays an Important Role in the Laminin-Dependent Development of Dorsal Root Ganglia during Chicken Embryogenesis. Cells 2021; 10:cells10071666. [PMID: 34359835 PMCID: PMC8304069 DOI: 10.3390/cells10071666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022] Open
Abstract
Integrin-linked kinase (ILK) is mainly localized in focal adhesions where it interacts and modulates the downstream signaling of integrins affecting cell migration, adhesion, and survival. The interaction of dorsal root ganglia (DRG) cells, being part of the peripheral nervous system (PNS), with the extracellular matrix (ECM) via integrins is crucial for proper PNS development. A few studies have focused on ILK’s role in PNS development, but none of these have focused on chicken. Therefore, we decided to investigate ILK’s role in the development of Gallus gallus domesticus’s DRG. First, using RT-PCR, Western blotting, and in situ hybridization, we show that ILK is expressed in DRG. Next, by immunocytochemistry, we show ILK’s localization both intracellularly and on the cell membrane of DRG neurons and Schwann cell precursors (SCPs). Finally, we describe ILK’s involvement in multiple aspects of DRG development by performing functional experiments in vitro. IgG-mediated interruption of ILK’s action improved DRG neurite outgrowth, modulated their directionality, stimulated SCPs migration, and impacted growth cone morphology in the presence of laminin-1 or laminin-1 mimicking peptide IKVAV. Taken together, our results show that ILK is important for chicken PNS development, probably via its exposure to the ECM.
Collapse
Affiliation(s)
- Ewa Mrówczyńska
- Correspondence: (E.M.); (A.J.M.); Tel.: +48-71-375-7972 (E.M.); +48-71-375-6206 (A.J.M.)
| | - Antonina Joanna Mazur
- Correspondence: (E.M.); (A.J.M.); Tel.: +48-71-375-7972 (E.M.); +48-71-375-6206 (A.J.M.)
| |
Collapse
|
15
|
Speer J, Barcellona M, Jing L, Liu B, Lu M, Kelly M, Buchowski J, Zebala L, Luhmann S, Gupta M, Setton L. Integrin-mediated interactions with a laminin-presenting substrate modulate biosynthesis and phenotypic expression for cells of the human nucleus pulposus. Eur Cell Mater 2021; 41:793-810. [PMID: 34160056 PMCID: PMC8378851 DOI: 10.22203/ecm.v041a50] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
With aging and pathology, cells of the nucleus pulposus (NP) de-differentiate towards a fibroblast-like phenotype, a change that contributes to degeneration of the intervertebral disc (IVD). Laminin isoforms are a component of the NP extracellular matrix during development but largely disappear in the adult NP tissue. Exposing human adult NP cells to hydrogels made from PEGylated-laminin-111 (PEGLM) has been shown to regulate NP cell behaviors and promote cells to assume a biosynthetically active state with gene/protein expression and morphology consistent with those observed in juvenile NP cells. However, the mechanism regulating this effect has remained unknown. In the present study, the integrin subunits that promote adult degenerative NP cell interactions with laminin-111 are identified by performing integrin blocking studies along with assays of intracellular signaling and cell phenotype. The findings indicate that integrin α3 is a primary regulator of cell attachment to laminin and is associated with phosphorylation of signaling molecules downstream of integrin engagement (ERK 1/2 and GSK3β). Sustained effects of blocking integrin α3 were also demonstrated including decreased expression of phenotypic markers, reduced biosynthesis, and altered cytoskeletal organization. Furthermore, blocking both integrin α3 and additional integrin subunits elicited changes in cell clustering, but did not alter the phenotype of single cells. These findings reveal that integrin- mediated interactions through integrin α3 are critical in the process by which NP cells sense and alter phenotype in response to culture upon laminin and further suggest that targeting integrin α3 has potential for reversing or slowing degenerative changes to the NP cell.
Collapse
Affiliation(s)
- J. Speer
- Department of Biomedical Engineering, Washington University in St. Louis; St. Louis, MO, USA
| | - M. Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis; St. Louis, MO, USA
| | - L. Jing
- Department of Biomedical Engineering, Washington University in St. Louis; St. Louis, MO, USA
| | - B. Liu
- Department of Biomedical Engineering, Washington University in St. Louis; St. Louis, MO, USA
| | - M. Lu
- Department of Biomedical Engineering, Washington University in St. Louis; St. Louis, MO, USA
| | - M. Kelly
- Department of Orthopedic Surgery, Washington University School of Medicine; St. Louis, MO, USA
| | - J. Buchowski
- Department of Orthopedic Surgery, Washington University School of Medicine; St. Louis, MO, USA
| | - L. Zebala
- Department of Orthopedic Surgery, Washington University School of Medicine; St. Louis, MO, USA
| | - S. Luhmann
- Department of Orthopedic Surgery, Washington University School of Medicine; St. Louis, MO, USA
| | - M. Gupta
- Department of Orthopedic Surgery, Washington University School of Medicine; St. Louis, MO, USA
| | - L. Setton
- Department of Biomedical Engineering, Washington University in St. Louis; St. Louis, MO, USA,Department of Orthopedic Surgery, Washington University School of Medicine; St. Louis, MO, USA,Address for correspondence: Dr. Lori A. Setton, Department of Biomedical Engineering, Washington University in St. Louis, 1 Brookings Drive, Campus Box 1097, St. Louis, MO 63130, USA. Telephone number: +1 3149356164,
| |
Collapse
|
16
|
Speer JE, Barcellona MN, Lu MY, Zha Z, Jing L, Gupta MC, Buchowski JM, Kelly MP, Setton LA. Development of a library of laminin-mimetic peptide hydrogels for control of nucleus pulposus cell behaviors. J Tissue Eng 2021; 12:20417314211021220. [PMID: 34188794 PMCID: PMC8211742 DOI: 10.1177/20417314211021220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022] Open
Abstract
The nucleus pulposus (NP) of the intervertebral disc plays a critical role in
distributing mechanical loads to the axial skeleton. Alterations in NP cells and,
consequently, NP matrix are some of the earliest changes in the development of disc
degeneration. Previous studies demonstrated a role for laminin-presenting biomaterials in
promoting a healthy phenotype for human NP cells from degenerated tissue. Here we
investigate the use of laminin-mimetic peptides presented individually or in combination
on a poly(ethylene) glycol hydrogel as a platform to modulate the behaviors of
degenerative human NP cells. Data confirm that NP cells attach to select laminin-mimetic
peptides that results in cell signaling downstream of integrin and syndecan binding.
Furthermore, the peptide-functionalized hydrogels demonstrate an ability to promote cell
behaviors that mimic that of full-length laminins. These results identify a set of
peptides that can be used to regulate NP cell behaviors toward a regenerative engineering
strategy.
Collapse
Affiliation(s)
- Julie E Speer
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Marcos N Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Y Lu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Zizhen Zha
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Munish C Gupta
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob M Buchowski
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael P Kelly
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
17
|
Amyloid Aggregates of Smooth-Muscle Titin Impair Cell Adhesion. Int J Mol Sci 2021; 22:ijms22094579. [PMID: 33925514 PMCID: PMC8123791 DOI: 10.3390/ijms22094579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022] Open
Abstract
Various amyloid aggregates, in particular, aggregates of amyloid β-proteins, demonstrate in vitro and in vivo cytotoxic effects associated with impairment of cell adhesion. We investigated the effect of amyloid aggregates of smooth-muscle titin on smooth-muscle-cell cultures. The aggregates were shown to impair cell adhesion, which was accompanied by disorganization of the actin cytoskeleton, formation of filopodia, lamellipodia, and stress fibers. Cells died after a 72-h contact with the amyloid aggregates. To understand the causes of impairment, we studied the effect of the microtopology of a titin-amyloid-aggregate-coated surface on fibroblast adhesion by atomic force microscopy. The calculated surface roughness values varied from 2.7 to 4.9 nm, which can be a cause of highly antiadhesive properties of this surface. As all amyloids have the similar structure and properties, it is quite likely that the antiadhesive effect is also intrinsic to amyloid aggregates of other proteins. These results are important for understanding the mechanisms of the negative effect of amyloids on cell adhesion.
Collapse
|
18
|
Ramasubramanian A, Muckom R, Sugnaux C, Fuentes C, Ekerdt BL, Clark DS, Healy KE, Schaffer DV. High-Throughput Discovery of Targeted, Minimally Complex Peptide Surfaces for Human Pluripotent Stem Cell Culture. ACS Biomater Sci Eng 2021; 7:1344-1360. [PMID: 33750112 DOI: 10.1021/acsbiomaterials.0c01462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human pluripotent stem cells harbor an unlimited capacity to generate therapeutically relevant cells for applications in regenerative medicine. However, to utilize these cells in the clinic, scalable culture systems that activate defined receptors and signaling pathways to sustain stem cell self-renewal are required; and synthetic materials offer considerable promise to meet these needs. De novo development of materials that target novel pathways has been stymied by a limited understanding of critical receptor interactions maintaining pluripotency. Here, we identify peptide agonists for the human pluripotent stem cell (hPSC) laminin receptor and pluripotency regulator, α6-integrin, through unbiased, library-based panning strategies. Biophysical characterization of adhesion suggests that identified peptides bind hPSCs through α6-integrin with sub-μM dissociation constants similar to laminin. By harnessing a high-throughput microculture platform, we developed predictive guidelines for presenting these integrin-targeting peptides alongside canonical binding motifs at optimal stoichiometries to generate nascent culture surfaces. Finally, when presented as self-assembled monolayers, predicted peptide combinations supported hPSC expansion, highlighting how unbiased screens can accelerate the discovery of targeted biomaterials.
Collapse
Affiliation(s)
- Anusuya Ramasubramanian
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Riya Muckom
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Caroline Sugnaux
- Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Christina Fuentes
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Barbara L Ekerdt
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Douglas S Clark
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, California 94720, United States
| | - David V Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
19
|
Bobyleva LG, Shumeyko SA, Yakupova EI, Surin AK, Galzitskaya OV, Kihara H, Timchenko AA, Timchenko MA, Penkov NV, Nikulin AD, Suvorina MY, Molochkov NV, Lobanov MY, Fadeev RS, Vikhlyantsev IM, Bobylev AG. Myosin Binding Protein-C Forms Amyloid-Like Aggregates In Vitro. Int J Mol Sci 2021; 22:ijms22020731. [PMID: 33450960 PMCID: PMC7828380 DOI: 10.3390/ijms22020731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 11/17/2022] Open
Abstract
This work investigated in vitro aggregation and amyloid properties of skeletal myosin binding protein-C (sMyBP-C) interacting in vivo with proteins of thick and thin filaments in the sarcomeric A-disc. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) found a rapid (5–10 min) formation of large (>2 μm) aggregates. sMyBP-C oligomers formed both at the initial 5–10 min and after 16 h of aggregation. Small angle X-ray scattering (SAXS) and DLS revealed sMyBP-C oligomers to consist of 7–10 monomers. TEM and atomic force microscopy (AFM) showed sMyBP-C to form amorphous aggregates (and, to a lesser degree, fibrillar structures) exhibiting no toxicity on cell culture. X-ray diffraction of sMyBP-C aggregates registered reflections attributed to a cross-β quaternary structure. Circular dichroism (CD) showed the formation of the amyloid-like structure to occur without changes in the sMyBP-C secondary structure. The obtained results indicating a high in vitro aggregability of sMyBP-C are, apparently, a consequence of structural features of the domain organization of proteins of this family. Formation of pathological amyloid or amyloid-like sMyBP-C aggregates in vivo is little probable due to amino-acid sequence low identity (<26%), alternating ordered/disordered regions in the protein molecule, and S–S bonds providing for general stability.
Collapse
Affiliation(s)
- Liya G. Bobyleva
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Sergey A. Shumeyko
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Elmira I. Yakupova
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
| | - Alexey K. Surin
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
- Biological Testing Laboratory, Branch of the Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
- Laboratory of the Biochemistry of Pathogenic Microorganisms, State Research Centre for Applied Microbiology and Biotechnology, Obolensk, 142279 Serpukhov District, Russia
| | - Oxana V. Galzitskaya
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Hiroshi Kihara
- Department of Early Childhood Education, Himeji-Hinomoto College, 890 Koro, Kodera-cho, Himeji 679-2151, Japan;
| | - Alexander A. Timchenko
- Group of Experimental Research and Engineering of Oligomeric Structures, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Maria A. Timchenko
- Laboratory of Applied Enzymology, FRC PSCBR, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Nikita V. Penkov
- Laboratory of the Methods of Optical Spectral Analysis, Institute of Cell Biophysics, Russian Academy of Sciences, FRC PSCBR RAS, 142290 Pushchino, Russia;
| | - Alexey D. Nikulin
- Laboratory for Structural Studies of the Translational Apparatus, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Mariya Yu. Suvorina
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Nikolay V. Molochkov
- Laboratory of NMR Investigations of Biosystems, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Mikhail Yu. Lobanov
- Laboratory of Bioinformatics and Proteomics, Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (A.K.S.); (M.Y.S.); (M.Y.L.)
| | - Roman S. Fadeev
- Laboratory of Pharmacological Regulation of Cell Resistance, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Ivan M. Vikhlyantsev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Correspondence: (I.M.V.); (A.G.B.)
| | - Alexander G. Bobylev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.B.); (S.A.S.); (E.I.Y.); (O.V.G.)
- Correspondence: (I.M.V.); (A.G.B.)
| |
Collapse
|
20
|
Sasaki E, Hayashi Y, Kimura Y, Sashida S, Hamano N, Nirasawa K, Hamada K, Katagiri F, Kikkawa Y, Sakai T, Yoshida A, Kawada M, Hirashima SI, Miura T, Endo-Takahashi Y, Nomizu M, Negishi Y. Alpha-dystroglycan binding peptide A2G80-modified stealth liposomes as a muscle-targeting carrier for Duchenne muscular dystrophy. J Control Release 2021; 329:1037-1045. [PMID: 33080271 DOI: 10.1016/j.jconrel.2020.10.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 01/16/2023]
Abstract
Safe and efficient gene therapy for the treatment of Duchenne muscular dystrophy (DMD), a genetic disorder, is required. For this, the muscle-targeting delivery system of genes and nucleic acids is ideal. In this study, we focused on the A2G80 peptide, which has an affinity for α-dystroglycan expressed on muscle cell membranes, as a muscle targeted nanocarrier for DMD and developed A2G80-modified liposomes. We also prepared A2G80-modified liposomes coated with long- and short-chain PEG, called A2G80-LSP-Lip, to improve the blood circulation of liposomes using microfluidics. The liposomes had a particle size of approximately 80 nm. A2G80-LSP-Lip showed an affinity for the muscle tissue section of mice by overlay assay. When the liposomes were administered to DMD model mice (mdx mice) via the tail vein, A2G80-LSP-Lip accumulated efficiently in muscle tissue compared to control liposomes. These results suggest that A2G80-LSP-Lip can function as a muscle-targeting liposome for DMD via systemic administration, and may be a useful tool for DMD treatment.
Collapse
Affiliation(s)
- Eri Sasaki
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoshihiro Hayashi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yuka Kimura
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Sanae Sashida
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Nobuhito Hamano
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Kei Nirasawa
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takaaki Sakai
- Department of Pharmaceutical Chemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Akihiro Yoshida
- Department of Pharmaceutical Chemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Masahiro Kawada
- Department of Pharmaceutical Chemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Shin-Ichi Hirashima
- Department of Pharmaceutical Chemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Tsuyoshi Miura
- Department of Pharmaceutical Chemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoko Endo-Takahashi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
21
|
Hayashi H, Horinokita I, Yamada Y, Hamada K, Takagi N, Nomizu M. Effects of laminin-111 peptide coatings on rat neural stem/progenitor cell culture. Exp Cell Res 2020; 400:112440. [PMID: 33359470 DOI: 10.1016/j.yexcr.2020.112440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/26/2020] [Accepted: 12/12/2020] [Indexed: 10/22/2022]
Abstract
Neurons require adhesive scaffolds for their growth and differentiation. Laminins are a major cell adhesive component of basement membranes and have various biological activities in the peripheral and central nervous systems. Here, we evaluated the biological activities of 5 peptides derived from laminin-111 as a scaffold for mouse neuroblastoma Neuro2a cells and rat neural stem/progenitor cells (NPCs). The 5 peptides showed Neuro2a cell attachment activity similar to that of poly-d-lysine. However, when NPCs were cultured on the peptides, 2 syndecan-binding peptides, AG73 (RKRLQVQLSIRT, mouse laminin α1 chain 2719-2730) and C16 (KAFDITYVRLKF, laminin γ1 chain 139-150), demonstrated significantly higher cell attachment and neurite extension activities than other peptides including integrin-binding ones. Long-term cell culture experiments showed that both AG73 and C16 supported the growth of neurons and astrocytes that had differentiated from NPCs. Furthermore, C16 markedly promoted the expression of neuronal markers such as synaptosomal-associated protein-25 and syntaxin 1A. These results indicate that AG73 and C16 are useful for NPC cultures and that C16 can be applied to specialized research on synapses in differentiated neurons. These peptides have the potential for use as valuable biomaterials for NPC research.
Collapse
Affiliation(s)
- Hideki Hayashi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Hachioji, 192-0392, Japan.
| | - Ichiro Horinokita
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Hachioji, 192-0392, Japan
| | - Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Hachioji, 192-0392, Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Hachioji, 192-0392, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Hachioji, 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Hachioji, 192-0392, Japan
| |
Collapse
|
22
|
Ishikawa M, Hamada K, Yamada Y, Kumai J, Katagiri F, Kikkawa Y, Nomizu M. Conformational dependence of integrin‐binding peptides derived from homologous loop regions in the laminin α chains. J Pept Sci 2020; 26:e3284. [DOI: 10.1002/psc.3284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/28/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Masaya Ishikawa
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Yuji Yamada
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Jun Kumai
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry School of Pharmacy, Tokyo University of Pharmacy and Life Sciences Tokyo Japan
| |
Collapse
|
23
|
Yamada Y, Yoshida C, Hamada K, Kikkawa Y, Nomizu M. Development of Three-Dimensional Cell Culture Scaffolds Using Laminin Peptide-Conjugated Agarose Microgels. Biomacromolecules 2020; 21:3765-3771. [DOI: 10.1021/acs.biomac.0c00871] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chihiro Yoshida
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
24
|
Jia J, Jeon EJ, Li M, Richards DJ, Lee S, Jung Y, Barrs RW, Coyle R, Li X, Chou JC, Yost MJ, Gerecht S, Cho SW, Mei Y. Evolutionarily conserved sequence motif analysis guides development of chemically defined hydrogels for therapeutic vascularization. SCIENCE ADVANCES 2020; 6:eaaz5894. [PMID: 32923589 PMCID: PMC7455498 DOI: 10.1126/sciadv.aaz5894] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 04/10/2020] [Indexed: 05/04/2023]
Abstract
Biologically active ligands (e.g., RGDS from fibronectin) play critical roles in the development of chemically defined biomaterials. However, recent decades have shown only limited progress in discovering novel extracellular matrix-protein-derived ligands for translational applications. Through motif analysis of evolutionarily conserved RGD-containing regions in laminin (LM) and peptide-functionalized hydrogel microarray screening, we identified a peptide (a1) that showed superior supports for endothelial cell (EC) functions. Mechanistic studies attributed the results to the capacity of a1 engaging both LM- and Fn-binding integrins. RNA sequencing of ECs in a1-functionalized hydrogels showed ~60% similarities with Matrigel in "vasculature development" gene ontology terms. Vasculogenesis assays revealed the capacity of a1-formulated hydrogels to improve EC network formation. Injectable alginates functionalized with a1 and MMPQK (a vascular endothelial growth factor-mimetic peptide with a matrix metalloproteinase-degradable linker) increased blood perfusion and functional recovery over decellularized extracellular matrix and (RGDS + MMPQK)-functionalized hydrogels in an ischemic hindlimb model, illustrating the power of this approach.
Collapse
Affiliation(s)
- Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Eun Je Jeon
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
- Department of Biomaterials Science and Engineering, Yonsei University, Seoul, Republic of Korea
| | - Mei Li
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Cardiology, Medical University of South Carolina, Charleston, SC, USA
| | - Dylan J. Richards
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Soojin Lee
- Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Robert Coyle
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaoyang Li
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
- Ocean University of China, School of Medicine and Pharmacy, Qingdao, Shandong, China
| | - James C. Chou
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC, USA
| | - Michael J. Yost
- Department of Surgery, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, and Johns Hopkins Physical Sciences–Oncology Center, The Johns Hopkins University, Baltimore, MD, USA
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
25
|
Karimi F, Thombare VJ, Hutton CA, O'Connor AJ, Qiao GG, Heath DE. Biomaterials functionalized with nanoclusters of integrin- and syndecan-binding ligands improve cell adhesion and mechanosensing under shear flow conditions. J Biomed Mater Res A 2020; 109:313-325. [PMID: 32490581 DOI: 10.1002/jbm.a.37024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022]
Abstract
We have engineered biomaterials that display nanoclusters of ligands that bind both integrin and syndecan-4 cell receptors. These surfaces regulate cell behaviors under static conditions including adhesion, spreading, actin stress fiber formation, and migration. The syndecan-4 receptors are also critical mediators of cellular mechanotransduction. In this contribution we assess whether this novel class of materials can regulate the response of cells to applied mechanical stimulation, using the shear stress imparted by laminar fluid flow as a model stimulus. Specifically, we assess endothelial cell detachment due to flow, cell alignment due to flow, and cell adhesion from the flowing fluid. A high degree of cell retention was observed on surfaces containing integrin-binding ligands or a mixed population of integrin- and syndecan-binding ligands. However, the presence of both ligand types was necessary for the cells to align in the direction of flow. These results imply that integrin engagement is necessary for adhesion strength, but engagement of both receptor types aids in appropriate mechanotransduction. Additionally, it was found that surfaces functionalized with both ligand types were able to scavenge a larger number of cells from flow, and to do so at a faster rate, compared to surfaces functionalized with only integrin- or syndecan-binding ligands. These results show that interfaces functionalized with both integrin- and syndecan-binding ligands regulate a significant range of biophysical cell behaviors in response to shear stress.
Collapse
Affiliation(s)
- Fatemeh Karimi
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, University of Melbourne, Parkville, Victoria, Australia.,Polymer Science Group, Department of Chemical Engineering, Particulate Fluid Processing Centre, University of Melbourne, Parkville, Victoria, Australia.,Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Varsha Jagannath Thombare
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia
| | - Craig A Hutton
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Greg G Qiao
- Polymer Science Group, Department of Chemical Engineering, Particulate Fluid Processing Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel E Heath
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
26
|
Cherian DS, Bhuvan T, Meagher L, Heng TSP. Biological Considerations in Scaling Up Therapeutic Cell Manufacturing. Front Pharmacol 2020; 11:654. [PMID: 32528277 PMCID: PMC7247829 DOI: 10.3389/fphar.2020.00654] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Cell therapeutics - using cells as living drugs - have made advances in many areas of medicine. One of the most clinically studied cell-based therapy products is mesenchymal stromal cells (MSCs), which have shown promising results in promoting tissue regeneration and modulating inflammation. However, MSC therapy requires large numbers of cells, the generation of which is not feasible via conventional planar tissue culture methods. Scale-up manufacturing methods (e.g., propagation on microcarriers in stirred-tank bioreactors), however, are not specifically tailored for MSC expansion. These processes may, in principle, alter the cell secretome, a vital component underlying the immunosuppressive properties and clinical effectiveness of MSCs. This review outlines our current understanding of MSC properties and immunomodulatory function, expansion in commercial manufacturing systems, and gaps in our knowledge that need to be addressed for effective up-scaling commercialization of MSC therapy.
Collapse
Affiliation(s)
- Darshana S Cherian
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tejasvini Bhuvan
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, Australia
| | - Tracy S P Heng
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
27
|
Barcellona MN, Speer JE, Fearing BV, Jing L, Pathak A, Gupta MC, Buchowski JM, Kelly M, Setton LA. Control of adhesive ligand density for modulation of nucleus pulposus cell phenotype. Biomaterials 2020; 250:120057. [PMID: 32361392 DOI: 10.1016/j.biomaterials.2020.120057] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/29/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
Cells of the nucleus pulposus have been observed to undergo a shift from their notochordal-like juvenile phenotype to a more fibroblast-like state with age and maturation. It has been demonstrated that culture of degenerative adult human nucleus pulposus cells upon soft (<1 kPa) full length laminin-containing hydrogel substrates promotes increased levels of a panel of markers associated with the juvenile nucleus pulposus cell phenotype. In the current work, we observed an ability to use soft polymeric substrates functionalized with short laminin-mimetic peptide sequences to recapitulate the behaviors elicited by soft, full-length laminin containing materials. Furthermore, our work suggests an ability to mimic features of soft systems through control of peptide density upon stiffer substrates. Specifically, results suggest that stiffer polymer-peptide hydrogel substrates can be used to promote the expression of a more juvenile-like phenotype for cells of the nucleus pulposus by reducing adhesive ligand presentation. Here we show how polymer stiffness combined with adhesive ligand presentation can be controlled to be supportive of nucleus pulposus cell phenotype and biosynthesis.
Collapse
Affiliation(s)
- Marcos N Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, USA
| | - Julie E Speer
- Department of Biomedical Engineering, Washington University in St. Louis, USA
| | - Bailey V Fearing
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Department of Orthopedic Surgery, Atrium Health Musculoskeletal Institute, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, USA
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, USA
| | - Munish C Gupta
- Department of Orthopedic Surgery, Washington University School of Medicine, USA
| | - Jacob M Buchowski
- Department of Orthopedic Surgery, Washington University School of Medicine, USA
| | - Michael Kelly
- Department of Orthopedic Surgery, Washington University School of Medicine, USA
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Department of Orthopedic Surgery, Washington University School of Medicine, USA.
| |
Collapse
|
28
|
Barros D, Amaral IF, Pêgo AP. Laminin-Inspired Cell-Instructive Microenvironments for Neural Stem Cells. Biomacromolecules 2019; 21:276-293. [PMID: 31789020 DOI: 10.1021/acs.biomac.9b01319] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Laminin is a heterotrimeric glycoprotein with a key role in the formation and maintenance of the basement membrane architecture and properties, as well as on the modulation of several biological functions, including cell adhesion, migration, differentiation and matrix-mediated signaling. In the central nervous system (CNS), laminin is differentially expressed during development and homeostasis, with an impact on the modulation of cell function and fate. Within neurogenic niches, laminin is one of the most important and well described extracellular matrix (ECM) proteins. Specifically, efforts have been made to understand laminin assembly, domain architecture, and interaction of its different bioactive domains with cell surface receptors, soluble signaling molecules, and ECM proteins, to gain insight into the role of this ECM protein and its receptors on the modulation of neurogenesis, both in homeostasis and during repair. This is also expected to provide a rational basis for the design of biomaterial-based matrices mirroring the biological properties of the basement membrane of neural stem cell niches, for application in neural tissue repair and cell transplantation. This review provides a general overview of laminin structure and domain architecture, as well as the main biological functions mediated by this heterotrimeric glycoprotein. The expression and distribution of laminin in the CNS and, more specifically, its role within adult neural stem cell niches is summarized. Additionally, a detailed overview on the use of full-length laminin and laminin derived peptide/recombinant laminin fragments for the development of hydrogels for mimicking the neurogenic niche microenvironment is given. Finally, the main challenges associated with the development of laminin-inspired hydrogels and the hurdles to overcome for these to progress from bench to bedside are discussed.
Collapse
Affiliation(s)
- Daniela Barros
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar , UPorto , Porto 4200-153 , Portugal
| | - Isabel F Amaral
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,FEUP - Faculdade de Engenharia , UPorto , Porto 4200-153 , Portugal
| | - Ana P Pêgo
- i3S - Instituto de Investigação e Inovação em Saúde , Universidade do Porto (UPorto) , Porto 4200-153 , Portugal.,INEB - Instituto de Engenharia Biomédica , UPorto , Porto 4200-153 , Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar , UPorto , Porto 4200-153 , Portugal.,FEUP - Faculdade de Engenharia , UPorto , Porto 4200-153 , Portugal
| |
Collapse
|
29
|
Kumai J, Yamada Y, Hamada K, Katagiri F, Hozumi K, Kikkawa Y, Nomizu M. Identification of active sequences in human laminin α5 G domain. J Pept Sci 2019; 25:e3218. [PMID: 31755207 DOI: 10.1002/psc.3218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/31/2019] [Accepted: 08/30/2019] [Indexed: 11/06/2022]
Abstract
Human laminin-511 (α5β1γ1) and its truncated protein, laminin-511 E8 fragment, bind to integrin α6β1 and have been widely used for embryonic stem cell and induced pluripotent stem cell culture under feeder-free conditions. In this study, we focused on human laminin α5 chain G domain, which is thought to be critical for the biological functions of laminin-511, and screened its biologically active sequences using a synthetic peptide library. We synthesized 115 peptides (hA5G1-hA5G115) covering the entire laminin α5 chain G domain and evaluated cell attachment activity using both the peptide-coated plate and peptide-chitosan matrix (peptide-ChtM) assays. Seventeen peptides demonstrated cell attachment activity in the assays. Both hA5G18 and hA5G26-coated plates and hA5G74-ChtMs promoted integrin β1-mediated cell attachment. These findings are useful for the study of molecular mechanisms of laminin-511, and the active peptides have a potential for use as a molecular probe for cell adhesion receptors.
Collapse
Affiliation(s)
- Jun Kumai
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Keisuke Hamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Kentaro Hozumi
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| |
Collapse
|
30
|
Gois Beghini D, Iwao Horita S, Monteiro da Fonseca Cardoso L, Anastacio Alves L, Nagaraju K, Henriques-Pons A. A Promising Future for Stem-Cell-Based Therapies in Muscular Dystrophies-In Vitro and In Vivo Treatments to Boost Cellular Engraftment. Int J Mol Sci 2019; 20:ijms20215433. [PMID: 31683627 PMCID: PMC6861917 DOI: 10.3390/ijms20215433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/28/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophies (MD) are a group of genetic diseases that lead to skeletal muscle wasting and may affect many organs (multisystem). Unfortunately, no curative therapies are available at present for MD patients, and current treatments mainly address the symptoms. Thus, stem-cell-based therapies may present hope for improvement of life quality and expectancy. Different stem cell types lead to skeletal muscle regeneration and they have potential to be used for cellular therapies, although with several limitations. In this review, we propose a combination of genetic, biochemical, and cell culture treatments to correct pathogenic genetic alterations and to increase proliferation, dispersion, fusion, and differentiation into new or hybrid myotubes. These boosted stem cells can also be injected into pretreate recipient muscles to improve engraftment. We believe that this combination of treatments targeting the limitations of stem-cell-based therapies may result in safer and more efficient therapies for MD patients. Matricryptins have also discussed.
Collapse
Affiliation(s)
- Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Samuel Iwao Horita
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | | | - Luiz Anastacio Alves
- Laboratório de Comunicação Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, NY 13902, USA.
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| |
Collapse
|
31
|
Bobylev AG, Kraevaya OA, Bobyleva LG, Khakina EA, Fadeev RS, Zhilenkov AV, Mishchenko DV, Penkov NV, Teplov IY, Yakupova EI, Vikhlyantsev IM, Troshin PA. Anti-amyloid activities of three different types of water-soluble fullerene derivatives. Colloids Surf B Biointerfaces 2019; 183:110426. [PMID: 31421408 DOI: 10.1016/j.colsurfb.2019.110426] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/16/2019] [Accepted: 08/04/2019] [Indexed: 01/21/2023]
Abstract
Anti-amyloid activity, aggregation behaviour, cytotoxicity and acute toxicity were investigated for three water-soluble fullerene derivatives with different types of solubilizing addends. All investigated compounds showed a strong anti-amyloid effect in vitrocaused by interaction of the water-soluble fullerene derivatives with the Ab(1-42)-peptide and followed by destruction of the amyloid fibrils. Notably, all of the studied fullerene derivatives showed very low cytotoxicity and low acute toxicity in mice (most promising compound 3 was more than four times less toxic than aspirin). Strong anti-amyloid effect of the fullerene derivatives together with low toxicity reveals high potential of these compounds as drug candidates for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexander G Bobylev
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya St. 3, Pushchino, Moscow Region, 142290, Russia
| | - Olga A Kraevaya
- Skolkovo Institute of Science and Technology, Nobel St. 3, Moscow, 143026, Russia; Institute for Problems of Chemical Physics of the Russian Academy of Sciences, Semenov Prospect 1, Chernogolovka, 141432, Russia
| | - Liya G Bobyleva
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya St. 3, Pushchino, Moscow Region, 142290, Russia
| | - Ekaterina A Khakina
- Institute for Problems of Chemical Physics of the Russian Academy of Sciences, Semenov Prospect 1, Chernogolovka, 141432, Russia
| | - Roman S Fadeev
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya St. 3, Pushchino, Moscow Region, 142290, Russia
| | - Alexander V Zhilenkov
- Institute for Problems of Chemical Physics of the Russian Academy of Sciences, Semenov Prospect 1, Chernogolovka, 141432, Russia
| | - Denis V Mishchenko
- Institute for Problems of Chemical Physics of the Russian Academy of Sciences, Semenov Prospect 1, Chernogolovka, 141432, Russia
| | - Nikita V Penkov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institutskaya St., 3, Pushchino, Moscow Region, 142290, Russia
| | - Ilia Y Teplov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institutskaya St., 3, Pushchino, Moscow Region, 142290, Russia
| | - Elmira I Yakupova
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya St. 3, Pushchino, Moscow Region, 142290, Russia
| | - Ivan M Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, Institutskaya St. 3, Pushchino, Moscow Region, 142290, Russia
| | - Pavel A Troshin
- Skolkovo Institute of Science and Technology, Nobel St. 3, Moscow, 143026, Russia; Institute for Problems of Chemical Physics of the Russian Academy of Sciences, Semenov Prospect 1, Chernogolovka, 141432, Russia.
| |
Collapse
|
32
|
Caires-Dos-Santos L, da Silva SV, Smuczek B, de Siqueira AS, Cruz KSP, Barbuto JAM, Augusto TM, Freitas VM, Carvalho HF, Jaeger RG. Laminin-derived peptide C16 regulates Tks expression and reactive oxygen species generation in human prostate cancer cells. J Cell Physiol 2019; 235:587-598. [PMID: 31254281 DOI: 10.1002/jcp.28997] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 06/01/2019] [Accepted: 06/04/2019] [Indexed: 01/05/2023]
Abstract
Laminin peptides influence cancer biology. We investigated the role of a laminin-derived peptide C16 regulating invadopodia molecules in human prostate cancer cells (DU145). C16 augmented invadopodia activity of DU145 cells, and stimulated expression Tks4, Tks5, cortactin, and membrane-type matrix metalloproteinase 1. Reactive oxygen species generation is also related to invadopodia formation. This prompted us to address whether C16 would induce reactive oxygen species generation in DU145 cells. Quantitative fluorescence and flow cytometry showed that the peptide C16 increased reactive oxygen species in DU145 cells. Furthermore, significant colocalization between Tks5 and reactive oxygen species was observed in C16-treated cells. Results suggested that the peptide C16 increased Tks5 and reactive oxygen species in prostate cancer cells. The role of C16 increasing Tks and reactive oxygen species are novel findings on invadopodia activity.
Collapse
Affiliation(s)
- Livia Caires-Dos-Santos
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Suély V da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Basilio Smuczek
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Department of Biology, UNICENTRO State University, Guarapuava, PR, Brazil
| | - Adriane S de Siqueira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,School of Dentistry, Positivo University, Curitiba, PR, Brazil
| | - Karen S P Cruz
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil.,Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, AL, Brazil.,Faculty of Nutrition, Federal University of Alagoas, Maceio, AL, Brazil
| | - José Alexandre M Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Taize M Augusto
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil.,Department of Morphology and Basic Pathology, School of Medicine of Jundiai, Jundiai, SP, Brazil
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Ruy G Jaeger
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
33
|
Galliou PA, Verrou KM, Koliakos G. Phosphorylation mapping of laminin α1-chain: Kinases in association with active sites. Comput Biol Chem 2019; 80:480-497. [PMID: 31174160 DOI: 10.1016/j.compbiolchem.2019.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/25/2019] [Indexed: 11/25/2022]
Abstract
Laminin-111 is a trimeric glycoprotein of the extracellular matrix (ECM) that holds a significant role in cell adhesion, migration and differentiation. Laminin-111 is the most studied laminin isoform, composed of three chains; α1, β1 and γ1. Phosphorylation is the most common eukaryotic post - translational modification and has regulatory effect on protein function. Using bioinformatic tools we computationally predicted all the possible phosphorylation sites on human laminin α1-chain sequence (LAMA1) according to kinases binding motifs. Thus, we predicted, for the first time, the possibly responsible kinases for fifteen of the nineteen already published experimentally observed phosphorylated residues in LAMA1. Searching the literature extensively, we recorded all the known functional sites (active sites) in LAMA1. We combined the experimentally observed and predicted phosphorylated residues as well as the active sites in LAMA1, generating an analytic phosphorylation map of human laminin α1-chain, which is useful for further analysis. Our results indicated fourteen kinases that might be important for the phosphorylation of human laminin α1-chain, out of which three kinases with reported ecto-phosphorylation activity (PKA, PKC and CKII) were suggested to have a more significant role. Six cancer associated-active sites were correlated with kinases, three out which were correlated with only the above ecto - kinases.
Collapse
Affiliation(s)
- Panagiota Angeliki Galliou
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124, Greece..
| | - Kleio-Maria Verrou
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124, Greece.; School of Medicine, University of Crete, 71500, Greece..
| | - George Koliakos
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124, Greece..
| |
Collapse
|
34
|
Negishi Y, Nomizu M. Laminin-derived peptides: Applications in drug delivery systems for targeting. Pharmacol Ther 2019; 202:91-97. [PMID: 31158392 DOI: 10.1016/j.pharmthera.2019.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/29/2022]
Abstract
Recently, the development of drug delivery systems (DDSs) for clinical application of anticancer drugs and gene therapy has rapidly progressed. In particular, DDS carriers used for chemotherapy and gene therapy are required to selectively deliver drugs and genes to cancer cells. Both the carrier and the molecule must in combination be highly selective in most cases. Possible candidate targeting molecules are the laminins, major basement membrane proteins that interact with various cells through their multiple constituent active peptide sequences. Laminin-derived peptides bind to various cellular receptors and have been used for DDSs as a targeting moiety. Here, we review the progress in laminin-derived peptide-conjugated DDSs. Drug and gene carriers as well as ultrasound diagnostic contrast agents utilizing laminin-derived peptides for selective targeting are useful components of DDSs and play important roles in cancer and in the neovasculature.
Collapse
Affiliation(s)
- Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
35
|
The Laminin- α1 Chain-Derived Peptide, AG73, Binds to Syndecans on MDA-231 Breast Cancer Cells and Alters Filopodium Formation. Anal Cell Pathol (Amst) 2019; 2019:9192516. [PMID: 31183318 PMCID: PMC6515157 DOI: 10.1155/2019/9192516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 02/17/2019] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is one of the most common forms of cancer affecting women in the United States, second only to skin cancers. Although treatments have been developed to combat primary breast cancer, metastasis remains a leading cause of death. An early step of metastasis is cancer cell invasion through the basement membrane. However, this process is not yet well understood. AG73, a synthetic laminin-α1 chain peptide, plays an important role in cell adhesion and has previously been linked to migration, invasion, and metastasis. Thus, we aimed to identify the binding partner of AG73 on breast cancer cells that could mediate cancer progression. We performed adhesion assays using MCF10A, T47D, SUM1315, and MDA-231 breast cell lines and found that AG73 binds to syndecans (Sdcs) 1, 2, and 4. This interaction was inhibited when we silenced Sdcs 1 and/or 4 in MDA-231 cells, indicating the importance of these receptors in this relationship. Through actin staining, we found that silencing of Sdc 1, 2, and 4 expression in MDA-231 cells exhibits a decrease in the length and number of filopodia bound to AG73. Expression of mouse Sdcs 1, 2, and 4 in MDA-231 cells provides rescue in filopodia, and overexpression of Sdcs 1 and 2 leads to increased filopodium length and number. Our findings demonstrate an intrinsic interaction between AG73 in the tumor environment and the Sdcs on breast cancer cells in supporting tumor cell adhesion and invasion through filopodia, an important step in cancer metastasis.
Collapse
|
36
|
Farrukh A, Zhao S, Paez JI, Kavyanifar A, Salierno M, Cavalié A, Del Campo A. In Situ, Light-Guided Axon Growth on Biomaterials via Photoactivatable Laminin Peptidomimetic IK(HANBP)VAV. ACS APPLIED MATERIALS & INTERFACES 2018; 10:41129-41137. [PMID: 30387978 DOI: 10.1021/acsami.8b15517] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The ability to guide the growth of neurites is relevant for reconstructing neural networks and for nerve tissue regeneration. Here, a biofunctional hydrogel that allows light-based directional control of axon growth in situ is presented. The gel is covalently modified with a photoactivatable derivative of the short laminin peptidomimetic IKVAV. This adhesive peptide contains the photoremovable group 2-(4'-amino-4-nitro-[1,1'-biphenyl]-3-yl)propan-1-ol (HANBP) on the Lys rest that inhibits its activity. The modified peptide is highly soluble in water and can be simply conjugated to -COOH containing hydrogels via its terminal -NH2 group. Light exposure allows presentation of the IKVAV adhesive motif on a soft hydrogel at desired concentration and at defined position and time point. The photoactivated gel supports neurite outgrowth in embryonic neural progenitor cells culture and allows site-selective guidance of neurites extension. In situ exposure of cell cultures using a scanning laser allows outgrowth of neurites in desired pathways.
Collapse
Affiliation(s)
- Aleeza Farrukh
- INM-Leibniz Institute for New Materials , Campus D2 2 , 66123 Saarbrücken , Germany
- Max Planck Graduate Center , Forum Universitatis 2 , Building 1111, 55122 Mainz , Germany
| | - Shifang Zhao
- INM-Leibniz Institute for New Materials , Campus D2 2 , 66123 Saarbrücken , Germany
- Chemistry Department , Saarland University , 66123 Saarbrücken , Germany
| | - Julieta I Paez
- INM-Leibniz Institute for New Materials , Campus D2 2 , 66123 Saarbrücken , Germany
| | - Atria Kavyanifar
- Institute of Physiological Chemistry , University Medical Center Johannes Gutenberg University , Hanns-Dieter-Hüsch-Weg 19 , D-55128 Mainz , Germany
| | - Marcelo Salierno
- Institute of Physiological Chemistry , University Medical Center Johannes Gutenberg University , Hanns-Dieter-Hüsch-Weg 19 , D-55128 Mainz , Germany
| | - Adolfo Cavalié
- Experimental and Clinical Pharmacology and Toxicology , Saarland University , 66421 Homburg , Germany
| | - Aránzazu Del Campo
- INM-Leibniz Institute for New Materials , Campus D2 2 , 66123 Saarbrücken , Germany
- Chemistry Department , Saarland University , 66123 Saarbrücken , Germany
| |
Collapse
|
37
|
Beyond RGD; nanoclusters of syndecan- and integrin-binding ligands synergistically enhance cell/material interactions. Biomaterials 2018; 187:81-92. [DOI: 10.1016/j.biomaterials.2018.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/22/2022]
|
38
|
Sivaraman K, Shanthi C. Matrikines for therapeutic and biomedical applications. Life Sci 2018; 214:22-33. [PMID: 30449450 DOI: 10.1016/j.lfs.2018.10.056] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/25/2022]
Abstract
Matrikines, peptides originating from the fragmentation of extracellular matrix proteins are identified to play important role in both health and disease. They possess biological activities, much different from their parent protein. Identification of such bioactive cryptic regions in the extracellular matrix proteins has attracted the researchers all over the world in the recent decade. These bioactive peptides could find use in preparation of biomaterials and tissue engineering applications. Matrikines identified in major extracellular matrix (ECM) proteins like collagen, elastin, fibronectin, and laminin are being extensively studied for use in tissue engineering and regenerative medicine. They are identified to modulate cellular activity like cell growth, proliferation, migration and may induce apoptosis. RGD, a well-known peptide identified in fibronectin with cell adhesive property is being investigated in designing biomaterials. Collagen hexapeptide GFOGER was found to promote cell adhesion and differentiation. Laminin also possesses regions with strong cell adhesion property. Recently, cell-penetrating peptides from elastin are used as a targeted delivery system for therapeutic drugs. The continued search for cryptic sequences in the extracellular matrix proteins along with advanced peptide coupling chemistries would lead to biomaterials with improved surface properties. This review article outlines the peptides derived from extracellular matrix and some of the possible applications of these peptides in therapeutics and tissue engineering applications.
Collapse
Affiliation(s)
- K Sivaraman
- School of Biosciences and Technology, VIT, Vellore 632014, Tamilnadu, India
| | - C Shanthi
- School of Biosciences and Technology, VIT, Vellore 632014, Tamilnadu, India.
| |
Collapse
|
39
|
Biological activities of laminin-111-derived peptide-chitosan matrices in a primary culture of rat cortical neurons. Arch Biochem Biophys 2018; 648:53-59. [DOI: 10.1016/j.abb.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/09/2018] [Accepted: 04/12/2018] [Indexed: 01/10/2023]
|
40
|
Llacua LA, Faas MM, de Vos P. Extracellular matrix molecules and their potential contribution to the function of transplanted pancreatic islets. Diabetologia 2018; 61:1261-1272. [PMID: 29306997 PMCID: PMC6449002 DOI: 10.1007/s00125-017-4524-8] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/18/2017] [Indexed: 12/18/2022]
Abstract
Extracellular matrix (ECM) molecules are responsible for structural and biochemical support, as well as for regulation of molecular signalling and tissue repair in many organ structures, including the pancreas. In pancreatic islets, collagen type IV and VI, and laminins are the most abundant molecules, but other ECM molecules are also present. The ECM interacts with specific combinations of integrin α/β heterodimers on islet cells and guides many cellular processes. More specifically, some ECM molecules are involved in beta cell survival, function and insulin production, while others can fine tune the susceptibility of islet cells to cytokines. Further, some ECM induce release of growth factors to facilitate tissue repair. During enzymatic isolation of islets for transplantation, the ECM is damaged, impacting islet function. However, restoration of the ECM in human islets (for example by adding ECM to the interior of immunoprotective capsules) has been shown to enhance islet function. Here, we provide current insight into the role of ECM molecules in islet function and discuss the clinical potential of ECM manipulation to enhance pancreatic islet function and survival.
Collapse
Affiliation(s)
- L Alberto Llacua
- Section of Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1 EA11, 9700 RB, Groningen, the Netherlands.
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Marijke M Faas
- Section of Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1 EA11, 9700 RB, Groningen, the Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Paul de Vos
- Section of Immunoendocrinology, Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1 EA11, 9700 RB, Groningen, the Netherlands
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
41
|
Ovadia EM, Colby DW, Kloxin AM. Designing well-defined photopolymerized synthetic matrices for three-dimensional culture and differentiation of induced pluripotent stem cells. Biomater Sci 2018; 6:1358-1370. [PMID: 29675520 PMCID: PMC6126667 DOI: 10.1039/c8bm00099a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are of interest for the study of disease, where these cells can be derived from patients and have the potential to be differentiated into any cell type; however, three-dimensional (3D) culture and differentiation of iPSCs within well-defined synthetic matrices for these applications remains limited. Here, we aimed to establish synthetic cell-degradable hydrogels that allow precise presentation of specific biochemical cues for 3D culture of iPSCs with relevance for hypothesis testing and lineage-specific differentiation. We synthesized poly(ethylene glycol)-(PEG)-peptide-based hydrogels by photoinitiated step growth polymerization and used them to test the hypothesis that the viability of iPSCs within these matrices could be rescued with appropriate biochemical cues inspired by proteins and integrins important for iPSC culture on Matrigel. Specifically, we selected a range of motifs inspired by iPSC binding to Matrigel, including laminin-derived IKVAV and YIGSR, α5β1-binding PHSRNG10RGDS, αvβ5-binding KKQRFRHRNRKG, and RGDS that is known to bind a variety of integrins for generally promoting cell adhesion. YIGSR and PHSRNG10RGDS resulted in the highest iPSC viability, where binding of β1 integrin was key, and these permissive compositions also allowed iPSC differentiation into neural progenitor cells (NPCs) (decreased oct4 expression and increased pax6 expression) in response to soluble factors. The resulting NPCs formed clusters of different sizes in response to each peptide, suggesting that matrix biochemical cues affect iPSC proliferation and clustering in 3D culture. In summary, we have established photopolymerizable synthetic matrices for the encapsulation, culture, and differentiation of iPSCs for studies of cell-matrix interactions and deployment in disease models.
Collapse
Affiliation(s)
- Elisa M Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| | | | | |
Collapse
|
42
|
Siqueira AS, Pinto MP, Cruz MC, Smuczek B, Cruz KSP, Barbuto JAM, Hoshino D, Weaver AM, Freitas VM, Jaeger RG. Laminin-111 peptide C16 regulates invadopodia activity of malignant cells through β1 integrin, Src and ERK 1/2. Oncotarget 2018; 7:47904-47917. [PMID: 27323814 PMCID: PMC5216987 DOI: 10.18632/oncotarget.10062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 06/04/2016] [Indexed: 01/22/2023] Open
Abstract
Laminin peptides influence tumor behavior. In this study, we addressed whether laminin peptide C16 (KAFDITYVRLKF, γ1 chain) would increase invadopodia activity of cells from squamous cell carcinoma (CAL27) and fibrosarcoma (HT1080). We found that C16 stimulates invadopodia activity over time in both cell lines. Rhodamine-conjugated C16 decorates the edge of cells, suggesting a possible binding to membrane receptors. Flow cytometry showed that C16 increases activated β1 integrin, and β1 integrin miRNA-mediated depletion diminishes C16-induced invadopodia activity in both cell lines. C16 stimulates Src and ERK 1/2 phosphorylation, and ERK 1/2 inhibition decreases peptide-induced invadopodia activity. C16 also increases cortactin phosphorylation in both cells lines. Based on our findings, we propose that C16 regulates invadopodia activity over time of squamous carcinoma and fibrosarcoma cells, probably through β1 integrin, Src and ERK 1/2 signaling pathways.
Collapse
Affiliation(s)
- Adriane S Siqueira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil.,School of Dentistry, Positivo University, Curitiba, PR, 81280-330, Brazil
| | - Monique P Pinto
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Mário C Cruz
- ICB Core Facility, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Basilio Smuczek
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Karen S P Cruz
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - José Alexandre M Barbuto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Daisuke Hoshino
- Division of Cancer Cell Research, Kanagawa Cancer Center, Yokohama, Kanagawa, 241-8515, Japan
| | - Alissa M Weaver
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Ruy G Jaeger
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| |
Collapse
|
43
|
Farrukh A, Ortega F, Fan W, Marichal N, Paez JI, Berninger B, Campo AD, Salierno MJ. Bifunctional Hydrogels Containing the Laminin Motif IKVAV Promote Neurogenesis. Stem Cell Reports 2017; 9:1432-1440. [PMID: 28988991 PMCID: PMC5829305 DOI: 10.1016/j.stemcr.2017.09.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 10/29/2022] Open
Abstract
Engineering of biomaterials with specific biological properties has gained momentum as a means to control stem cell behavior. Here, we address the effect of bifunctionalized hydrogels comprising polylysine (PL) and a 19-mer peptide containing the laminin motif IKVAV (IKVAV) on embryonic and adult neuronal progenitor cells under different stiffness regimes. Neuronal differentiation of embryonic and adult neural progenitors was accelerated by adjusting the gel stiffness to 2 kPa and 20 kPa, respectively. While gels containing IKVAV or PL alone failed to support long-term cell adhesion, in bifunctional gels, IKVAV synergized with PL to promote differentiation and formation of focal adhesions containing β1-integrin in embryonic cortical neurons. Furthermore, in adult neural stem cell culture, bifunctionalized gels promoted neurogenesis via the expansion of neurogenic clones. These data highlight the potential of synthetic matrices to steer stem and progenitor cell behavior via defined mechano-adhesive properties.
Collapse
Affiliation(s)
- Aleeza Farrukh
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | - Felipe Ortega
- Biochemistry and Molecular Biology Department IV, Faculty of Veterinary Medicine, Complutense University, Madrid, Spain; Institute of Neurochemistry (IUIN), 28040 Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Wenqiang Fan
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 19, 55128 Mainz, Germany; Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Nicolás Marichal
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 19, 55128 Mainz, Germany; Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Julieta I Paez
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | - Benedikt Berninger
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 19, 55128 Mainz, Germany; Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany; Saarland University, Campus Saarbrücken D2 2, 66123 Saarbrücken, Germany
| | - Marcelo J Salierno
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 19, 55128 Mainz, Germany; Focus Program Translational Neuroscience, Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| |
Collapse
|
44
|
Silva J, Bento AR, Barros D, Laundos TL, Sousa SR, Quelhas P, Sousa MM, Pêgo AP, Amaral IF. Fibrin functionalization with synthetic adhesive ligands interacting with α6β1 integrin receptor enhance neurite outgrowth of embryonic stem cell-derived neural stem/progenitors. Acta Biomater 2017; 59:243-256. [PMID: 28694236 DOI: 10.1016/j.actbio.2017.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 12/25/2022]
Abstract
To enhance fibrin hydrogel affinity towards pluripotent stem cell-derived neural stem/progenitor cells (NSPCs) and its capacity to support NSPC migration and neurite extension, we explored the tethering of synthetic peptides engaging integrin α6β1, a cell receptor enriched in NSPCs. Six α6β1 integrin ligands were tested for their ability to support integrin α6β1-mediated adhesion of embryonic stem cell-derived NSPCs (ES-NSPs) and sustain ES-NSPC viability, migration, and neuronal differentiation. Due to their better performance, peptides T1, HYD1, and A5G81 were immobilized into fibrin and functionalized gels characterized in terms of peptide binding efficiency, structure and viscoelastic properties. Tethering of T1 or HYD1 successfully enhanced cell outgrowth from ES-NSPC neurospheres (up to 2.4-fold increase), which exhibited a biphasic response to peptide concentration. Inhibition assays evidenced the involvement of α6β1 and α3β1 integrins in mediating radial outgrowth on T1-/HYD1-functionalized gels. Fibrin functionalization also promoted neurite extension of single ES-NSPCs in fibrin, without affecting cell proliferation and neuronal differentiation. Finally, HYD1-functionalized gels were found to provide a permissive environment for axonal regeneration, leading up to a 2.0-fold increase in neurite extension from rat dorsal root ganglia explants as compared to unmodified fibrin, and to significant improved locomotor function after spinal cord injury (complete transection), along with a trend toward a higher area positive for growth associated protein 43 (marker for axonal growth cone formation). Our results suggest that conjugation of α6β1 integrin-binding motifs is of interest to increase the biofunctionality of hydrogels used in 3D platforms for ES-NSPC culture and potentially, in matrix-assisted ES-NSPC transplantation. STATEMENT OF SIGNIFICANCE Impact statement: The transplantation of NSPCs derived from pluripotent stem cells holds much promise for the treatment of central nervous system disorders. Moreover, the combinatorial use of biodegradable hydrogels with NSPCs was shown to contribute to the establishment of a more permissive environment for survival and integration of transplanted cells. In this study, fibrin hydrogels functionalized with a synthetic peptide engaging integrin α6β1 (HYD1) were shown to promote neurite extension of ES-NSPCs, which is fundamental for the formation of functional neuronal relay circuits after NSPC transplantation. Notably, HYD1-functionalized fibrin per se led to enhanced axonal growth ex vivo and to an improvement in locomotor function after implantation in a rat model of spinal cord injury. Conjugation of α6β1 integrin-binding motifs may therefore be of interest to confer bioactivity to NSPC hydrogel vehicles.
Collapse
Affiliation(s)
- Joana Silva
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Ana R Bento
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Portugal
| | - Daniela Barros
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Tiago L Laundos
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Susana R Sousa
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; ISEP - Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Portugal
| | - Pedro Quelhas
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Mónica M Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Nerve Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
| | - Ana P Pêgo
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Isabel F Amaral
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Portugal.
| |
Collapse
|
45
|
Regulation of human nucleus pulposus cells by peptide-coupled substrates. Acta Biomater 2017; 55:100-108. [PMID: 28433788 DOI: 10.1016/j.actbio.2017.04.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/20/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022]
Abstract
Nucleus pulposus (NP) cells are derived from the notochord and differ from neighboring cells of the intervertebral disc in phenotypic marker expression and morphology. Adult human NP cells lose this phenotype and morphology with age in a pattern that contributes to progressive disc degeneration and pathology. Select laminin-mimetic peptide ligands and substrate stiffnesses were examined for their ability to regulate human NP cell phenotype and biosynthesis through the expression of NP-specific markers aggrecan, N-cadherin, collagen types I and II, and GLUT1. Peptide-conjugated substrates demonstrated an ability to promote expression of healthy NP-specific markers, as well as increased biosynthetic activity. We show an ability to re-express markers of the juvenile NP cell and morphology through control of peptide presentation and stiffness on well-characterized polyacrylamide substrates. NP cells cultured on surfaces conjugated with α3 integrin receptor peptides P4 and P678, and on α2, α5, α6, β1 integrin-recognizing peptide AG10, show increased expression of aggrecan, N-cadherin, and types I and II collagen, suggesting a healthier, more juvenile-like phenotype. Multi-cell cluster formation was also observed to be more prominent on peptide-conjugated substrates. These findings indicate a critical role for cell-matrix interactions with specific ECM-mimetic peptides in supporting and maintaining a healthy NP cell phenotype and bioactivity. STATEMENT OF SIGNIFICANCE NP cells reside in a laminin-rich environment that deteriorates with age, including a loss of water content and changes in the extracellular matrix (ECM) structure that may lead to the development of a degenerated IVD. There is great interest in methods to re-express healthy, biosynthetically active NP cells using laminin-derived biomimetic peptides toward the goal of using autologous cell sources for tissue regeneration. Here, we describe a novel study utilizing several laminin mimetic peptides conjugated to polyacrylamide gels that are able to support an immature, healthy NP phenotype after culture on "soft" peptide gels. These findings can support future studies in tissue regeneration where cells may be directed to a desired regenerative phenotype using niche-specific ECM peptides.
Collapse
|
46
|
Fujimori C, Kumai J, Nakamura K, Gu Y, Katagiri F, Hozumi K, Kikkawa Y, Nomizu M. Biological activity of peptide-conjugated polyion complex matrices consisting of alginate and chitosan. Biopolymers 2017; 108. [PMID: 27603160 DOI: 10.1002/bip.22983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/20/2016] [Accepted: 08/29/2016] [Indexed: 11/09/2022]
Abstract
Peptide-conjugated polysaccharide matrices using bioactive laminin-derived peptides are useful biomaterials for tissue and cell engineering. Here, we demonstrate an easy handling preparation method for peptide-polysaccharide matrices using polyion complex with both alginate and chitosan. First, aldehyde-alginate was synthesized by oxidization of alginate using NaIO4 , and then, reacted with Cys-peptides. Next, the peptide-alginate solution was added to a chitosan-coated plate, and the peptide-polyion complex matrices (peptide-PCMs) were prepared. The peptide-PCMs using an integrin αvβ3-binding peptide (A99a: ALRGDN, mouse laminin α1 chain 1145-1150) and an integrin α2β1-binding peptide (EF1XmR: RLQLQEGRLHFXFD, X = Nle, mouse laminin α1 chain 2751-2763) showed strong cell attachment activity in a dose-dependent manner. When we examined the effect of various spacers on the biological activity of A99a-PCM, hydrophobic and long spacers enhanced the cell attachment activity. Further, the A99a-PCM with the spacers strongly promoted neurite outgrowth. The polyion complex method is an easy way to obtain insolubilized matrix and is widely applicable for various polysaccharides. The peptide-PCM is useful as a biomaterial for cell and tissue engineering.
Collapse
Affiliation(s)
- Chikara Fujimori
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Jun Kumai
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Kyotaro Nakamura
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Yingzi Gu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Kentaro Hozumi
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| |
Collapse
|
47
|
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
|
48
|
Gjorevski N, Sachs N, Manfrin A, Giger S, Bragina ME, Ordóñez-Morán P, Clevers H, Lutolf MP. Designer matrices for intestinal stem cell and organoid culture. Nature 2016; 539:560-564. [PMID: 27851739 DOI: 10.1038/nature20168] [Citation(s) in RCA: 940] [Impact Index Per Article: 104.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 10/18/2016] [Indexed: 12/30/2022]
Abstract
Epithelial organoids recapitulate multiple aspects of real organs, making them promising models of organ development, function and disease. However, the full potential of organoids in research and therapy has remained unrealized, owing to the poorly defined animal-derived matrices in which they are grown. Here we used modular synthetic hydrogel networks to define the key extracellular matrix (ECM) parameters that govern intestinal stem cell (ISC) expansion and organoid formation, and show that separate stages of the process require different mechanical environments and ECM components. In particular, fibronectin-based adhesion was sufficient for ISC survival and proliferation. High matrix stiffness significantly enhanced ISC expansion through a yes-associated protein 1 (YAP)-dependent mechanism. ISC differentiation and organoid formation, on the other hand, required a soft matrix and laminin-based adhesion. We used these insights to build a fully defined culture system for the expansion of mouse and human ISCs. We also produced mechanically dynamic matrices that were initially optimal for ISC expansion and subsequently permissive to differentiation and intestinal organoid formation, thus creating well-defined alternatives to animal-derived matrices for the culture of mouse and human stem-cell-derived organoids. Our approach overcomes multiple limitations of current organoid cultures and greatly expands their applicability in basic and clinical research. The principles presented here can be extended to identify designer matrices that are optimal for long-term culture of other types of stem cells and organoids.
Collapse
Affiliation(s)
- Nikolce Gjorevski
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Norman Sachs
- Hubrecht Institute and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - Andrea Manfrin
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sonja Giger
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maiia E Bragina
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Paloma Ordóñez-Morán
- Swiss Institute for Experimental Cancer Research (ISREC), EPFL, Lausanne, Switzerland
| | - Hans Clevers
- Hubrecht Institute and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Institute of Chemical Sciences and Engineering, School of Basic Science, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
49
|
Shim G, Yu YH, Lee S, Kim J, Oh YK. Surface-modified liposomes for syndecan 2–targeted delivery of edelfosine. Asian J Pharm Sci 2016. [DOI: 10.1016/j.ajps.2016.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
50
|
Rogers RS, Nishimune H. The role of laminins in the organization and function of neuromuscular junctions. Matrix Biol 2016; 57-58:86-105. [PMID: 27614294 DOI: 10.1016/j.matbio.2016.08.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/10/2016] [Accepted: 08/17/2016] [Indexed: 01/11/2023]
Abstract
The synapse between motor neurons and skeletal muscle is known as the neuromuscular junction (NMJ). Proper alignment of presynaptic and post-synaptic structures of motor neurons and muscle fibers, respectively, is essential for efficient motor control of skeletal muscles. The synaptic cleft between these two cells is filled with basal lamina. Laminins are heterotrimer extracellular matrix molecules that are key members of the basal lamina. Laminin α4, α5, and β2 chains specifically localize to NMJs, and these laminin isoforms play a critical role in maintenance of NMJs and organization of synaptic vesicle release sites known as active zones. These individual laminin chains exert their role in organizing NMJs by binding to their receptors including integrins, dystroglycan, and voltage-gated calcium channels (VGCCs). Disruption of these laminins or the laminin-receptor interaction occurs in neuromuscular diseases including Pierson syndrome and Lambert-Eaton myasthenic syndrome (LEMS). Interventions to maintain proper level of laminins and their receptor interactions may be insightful in treating neuromuscular diseases and aging related degeneration of NMJs.
Collapse
Affiliation(s)
- Robert S Rogers
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| |
Collapse
|