• No results found

University of Groningen Topography-mediated Control of Cellular Response: Migration, Intracellular Crowding, and Gene-delivery Ge, Lu

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Topography-mediated Control of Cellular Response: Migration, Intracellular Crowding, and Gene-delivery Ge, Lu"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Topography-mediated Control of Cellular Response: Migration, Intracellular Crowding, and Gene-delivery

Ge, Lu

DOI:

10.33612/diss.146106454

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Ge, L. (2020). Topography-mediated Control of Cellular Response: Migration, Intracellular Crowding, and Gene-delivery. University of Groningen. https://doi.org/10.33612/diss.146106454

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

109

CHAPTER 6

General Discussion & Future

Perspective

(3)

110

6.1 Discussion & Future perspective

Cell-material interfaces occupy a very important position in tissue engineering and regenerative medicine for regulating cell response and tissue functions1. Especially, the biophysical effect of

materials that determines cell functions in development, physiology, and pathophysiology, remains a central endeavor in tissue engineering2. With the vigorous development and major breakthroughs

of regenerative medicine technology, increasing amounts of information have been generated for understanding bio-interface complexity in regulating cellular responses. It remains a significant challenge to understand biophysical cues like topography to modulate cell development and subcellular behaviors3. The general aim of this thesis is to explore topography-mediated alterations

of cell behaviors and investigate cell-material interface-induced subcellular behaviors like cell morphology alteration, cell migration in wound healing, intracellular macromolecular crowding, and topography-modulated gene delivery of stem cells.

In this thesis, we discussed the role of high-throughput screening (HTS) in regulating cell spreading, proliferation, and migration. Wrinkle-gradient substrates were fabricated with diverse wavelength and amplitude parameters for investigating the topographic cues on the fibroblast cell migration behavior in wound healing approaches. In addition, uniform wrinkle substrates were developed with various wavelength and amplitude parameters to study the topography-induced intracellular macromolecular crowding and identifying subcellular behaviors like cell morphology alterations, mechanical transduction, metabolic activity, and protein expression. Additionally, the uniform wrinkle substrates were also used for investigating the modulation of gene-delivery capacity. The main finding is that topography and its sub-parameters such as direction, wavelength, and amplitude have an important influence on fibroblast migration in wound healing procedure. Different wrinkle features induce different intracellular macromolecular crowding phenomena that is associated with other subcellular activities. In addition, wave-like topography-mediated enhancement of non-viral gene delivery of stem cells was investigated. The obvious influence of topography on cell spreading, proliferation, migration, macromolecular crowding, and gene expression highlights its importance as a design parameter for the application of biomaterials.

Considerable research has been devoted to using high-throughput screening methods to identify how to regulate cell behaviors4–7. Chapter 2 discussed that the high-throughput screening platforms serve

as an important tool for determining cell adhesion8, spreading9, orientation10, proliferation11,

migration12, and cell fate decision13. In this chapter the high-throughput screening platforms with

physical cues (e.g. mechanical properties, topography, wettability), chemical or bio-chemical stimuli (e.g. material composition and proteins) and multiple parameter combinations were discussed for the manipulation of cell behaviors. Unlike the independent substrates or the randomly chosen degrees of biomaterial properties, the high-throughput screening platform combines multiple factors in a single system, which are timesaving, expedite analysis procedures, and minimizes systematic or methodological errors. To take the advantage of high-throughput screening methods as mentioned above, Chapter 3 indicated that the HTS developed enables efficient investigation for the modulation of fibroblasts migration in wound healing procedures12. The PDMS based topographical gradient with

wave-like features were fabricated by decoupling the amplitude and wavelength gradually differ in wavelength and amplitude to explore the role of topographic direction, structure repetition, and feature size of the substrate on fibroblast migration. The approaches developed are combined with multiple parameters to make it possible to investigate cell migration behaviors in a high-throughput way. In addition, the PDMS used have the properties of cost-efficiency, non-toxic and approved by FDA for implantable engineering scaffold14. The results indicated that cell movement was guided by

topographical properties, with a lower wrinkle wavelength (2 μm) eliciting the fastest migration speed, and the migration speed increased with decreasing amplitude15. The wavelength and amplitude both

(4)

111 topographical orientation with respect to the anisotropy of the topography and the cell migration speed is regulated by focal adhesion expression. These results demonstrate that anisotropic gradient platform can serve as an effective system to obtain the optimum parameter for specific cellular behaviors, which could improve regenerative medicine.

Cell movement is also essential for numerous physiological and pathological processes such as embryonic development, angiogenesis, immune surveillance, cancer metastasis, tissue regeneration, and wound healing15. Except for the topography stimuli, the cell migration is regulated by chemical

factors16, stiffness13, growth factors, electrical signals17, molecular signals18, and cell-cell contact19. For

instance, the growth factors regulates cell migration by controling the cell focal adhesion and contractility20. However, some research domestrates that growth factors like PDGF, bFGF, TGF-β2,

and TGF-β3 can stimulate fibroblasts to excessively produce ECM, which may induce scar formation21. The current investigations in this chapter focus more on the biomaterial scaffolds to

mimic the ECM topography enabling cells to be guided by ‘contact guidance’22 or ‘topotaxis’23 leading

to less scar formation24. Compared to the normaly used nano-grooves with right angles and sharp

ridges, the wave-like substrate we used with a semicircular shape, better mimic the ECM fibers and therefore represent a more biologically relevant approach to study the natural wound recovery procedure. Additionaly, the topographic orientation significantly affects the cells migration capacity in the wound healing procedure, which provides important instructions for material design.

This high-throughput screening approach which was used to investigating the fibroblasts migration with various surface feature parameters can provide a superior data collection with fewer experiments. It can be further used to mimic the complexity of in vivo conditions for other tissues or be used as a

model to control tissue properties and cell behaviors. For instance, Zhou et al. prepared directional wrinkle gradients to investigate the osteoblast attachment and cell orientation25. And the authors

translated PDMS-based wrinkle gradients to inorganic surface (SiO2, TiO2, CrO3, and Al2O3) to

investigate the hBM-MSC orientation and focal adhesion assembly26. Furthermore, Yang et al. used

the high-throughput screening approach for investigate the influence of mesenchymal stem cells differentiation towards osteogenic and neuronal lineage, respectively27,28. It should be noted that the

specific anisotropic geometrical organization is essential for the function of tissues such as skin, heart, bone, nerve, muscle, and tendon29. Artificial ECM-mimicking scaffolds that are designed according

to the special features of a tissue (e.g., its composition, mechanical properties, topography, and 3D geometry) have been determined to provide biological activity clues to regulate cell functions for tissue regeneration (Figure 1). What is more, the mechanical properties of the substrate also play a crucial role in modulating various cell behaviors. For example, substrates of about 30−35 kPa are beneficial for osteogenic differentiation, softer substrate (<1 kPa) enhance neurogenic differentiation, and substrate with moderate mechanical properties improve myogenesis or adipogenesis30. Therefore,

it would be a promising strategy for combining several parameters on single substrate and find the promising parameter in an efficient way. Therefore, the HTS platforms can be combined with other bio-factors (like soluble factors31), mechanical stimuli (like stiffness32 and wettability), 3D scaffolds

(like collagen scaffold33 and hydrogels34) and the anisotropic geometry to regulate cell functions of

(5)

112

Figure 1. Materials with tissue-mimetic physical properties (e.g., compositional, mechanical, and structural

features) offer specific stimulations to accelerate tissue regeneration. Reprinted with permission from ref36.

Except for the high-throughput platform, the influence of uniform wrinkle topography on macromolecular crowding and gene delivery were explored. In Chapter 4 the topography induced macromolecular crowding alteration in living cells were discussed. The macromolecular crowding components inside the cytosol has a profound impact on polypeptide and oligomeric proteins generation, folding, diffusion, enzymatic reactions37, and metabolic activity38. For instance, the

addition of some natural or synthetic polymers can enhance the extracellular matrix deposition and metabolic stimulation of MSCs39. It is well established that topography has an impact on various cell

functions, thus investigating the macromolecular crowding of cells that are cultured on topographic surfaces is important for understanding cell-material interfaces and illustrate what occurs naturally inside the cell. The HEK293T cells were transfected with a fluorescence resonance energy transfer (FRET)-based sensor for direct evaluate the macromolecular crowding inside living cells that are stimulated by wave-like surface topographies. The substrates used are uniform wrinkles with different wavelengths that is 0.5 μm, 2 μm, 10 μm, 25 μm and the Flat surface functions as the control. The main findings is that, increased macromolecular crowding was observed for cells cultured on 0.5 μm and 2 µm topographies, and the 2 μm induced a larger cell area and nucleus formation, higher

(6)

113 metabolic activities, proliferation rate, and more protein expression, correlated with increased focal adhesion and myosin tension but not YAP-TAZ transduction.

These findings illustrate that the spatiotemporal readout of crowding is a compelling tool for understanding cell-biomaterials interactions, thereby giving direction to identify specific mechanisms and allow us to investigate the role of macromolecular crowding of the cytoplasm during the cell development. Previous studies demonstrated that topography-induced acceleration of osteogenic differentiation is caused by focal adhesion, RhoA/ROCK signaling pathway40. Some work in our

group determined that topogrphy induced enhancement of stem cell differentiation is correlated with more focal ahesion formation, moyosin tension and YAP-TAZ localization into the nucleus32,41,42.

Chapter 4 shows that higher cowding is correlated with increased focal adhesion and cell contractility but not YAP-TAZ transduction. This is probably due to the different cell type and cell signalling pathways. For instance, osteogenic differentiation is also mediated by other pathways like mitogen-activated protein kinase (MAPK) pathway43, integrin-linked kinase (ILK)/β-catenin pathway32,

FAK/MAPK pathway, and extracellular signal-regulated kinase 1/2 (ERK1/2) pathway44. Therefore,

further investigations are necessary to completely identify the mechanisms for macromolecular crowding enhanced by topography.

It should be noted that this crowding sensor can be used in Escherichia coli and eukaryotic cells. Additionally, it is sensitive only to the excluded volume induced by macromolecular crowding and the sensor readout was reversible45. In other research, different types of sensors were used for in vitro46

and in vivo47 studies. For example, the sensor arrays46 to monitor cell adhesion and spreading,

biomimetic sensor for detecting nitric oxide molecules48, synthetic fluorescent sensors to detect

metals ions49, and metabolic sensor that couples nutritional availability50. Therefore, it would be a

promising strategy to study cell-materials interfaces using crowding sensor correlated with other sensors. Furthermore, as stem cell is essential for tissue engineering, we have tried to import the crowding sensor into hBM-MSCs but due to the low transfection efficiency, the crowding effect was not achieved. Expectantly, in the future, we can further extend this crowding sensor combined with other intracellular sensors into stem cells with high-throughput screening approaches that can provide more information about cell-materials interfaces and many new insights for tissue engineering and regenerative medicine.

In addition to the uniform wrinkle surfaces that modulated macromolecular crowding, in Chapter 5 the aligned nano- and micro- patterned PDMS substrates were used to investigate the topography influence on gene delivery in hBM-MSCs and myoblast cells. Gene delivery on purpose to introduce therapeutic genes or artificially modified genes into cells for modifying the cell function51, which is

essential for applications like biosensors52, cancer therapy53 and tissue regeneration54. In comparison

to the traditional deliver systems that need expensive hardware, time consuming, possibly induce immunogenicity, or have high toxicity problems, substrate-mediated gene delivery system is promising owing to its diverse physicochemical properties and good biocompatibility. We found that a 55% percent improvement of transfection efficiency was identified for hBM-MSCs grown on 2 µm wrinkles as compared to hBM-MSCs cultured on Flat controls. The highest gene-expression efficiency was observed on the 10 µm topography of V49 fibroblasts, which enhanced the transfection efficiency by 64% as compared to the Flat control. The results are in line with our initial hypothesis that mechanical stimuli of topography induced substrate-mediated gene delivery of stem cells. The induced gene transfection efficiency highlights the importance of topography-mediated gene delivery55.

In Chapter 5, the hBM-MSCs spreading area and elongation was altered by topography. Correspondingly, other works of our group have explored the influence of topography on cell morphology change and stem cell differentiation5,56,57. Similarly, others demonstrated well spread area

(7)

114

and elongated morphology promote mesenchymal stem cells gene transfection58. Another study

showed that nanogrooves influence gene transfection by controlling cytoskeleton organization and nuclei morphology59. The gene transfection efficiency of hBM-MSCs was enhanced on W2 more than

on the other wrinkle features. As well as our previously work reported, the wrinkle topography has an influence on single cell stiffness, stem cells growth on the W0.5 and W3 and showed higher stiffness than on the other topographies and stem cells differentiation behaviors concomitantly changed32. However, the reason that induces the diverse of stem cells tansfection are still not clear.

The intracellular processes such as internalization, endosomal escape, cytosolic trafficking and nuclear entry play a central role in gene delivery60. Some researchers showed that cellular uptake of cationic

complexes mainly rely on clathrin-mediated endocytosis, which is modulated by cell division control protein (Cde42) from the Rho family of GTPases60. RhoGTPases activation is essential for focal

adhesion assembly and disassembly, focal adhesions anchor actin stress fibers in turn facilitate intracellular trafficking that presumably improved transfection61. Additionally, the proliferation62 and

cellular metabolism63 is often shown to enhance gene delivery. Therefore, further investigations (e.g.

specific mechanotransduction signal pathways and metabolic activities) are necessary to completely identify the mechanisms for gene delivery enhanced by topography.

Figure 2. Niche interactions known to modulate stem cell phenotype. Reprinted with permission from ref64.

Cell–cell contact

Matrix micro- and nanostructure Cell-adhesive matrix ligands Cell-secreted factors F Matrix mechanics Matrix degradation

(8)

115 As it mentioned in the beginning of this thesis, cells are residing in a highly dynamic and extremely complicated three-dimensional (3D) microenvironment (Figure 2), which provides diverse biochemical and biophysical cues, that regulate cell functions and development65. The interaction with

neighboring cells, soluble factors, extracellular matrix (ECM), and biophysical stimuli (e.g. mechanical property, 2D topography and 3D geometry) strongly influence cell behaviors (e.g. cell adhesion, spreading, proliferation, cell alignment, migrate and the differentiation or self-maintenance of stem cells)36. This thesis provides evidence that topography works as a useful tool for understanding

cell-material interfaces and investigate cellular behaviors like mediated alterations of cell migration, intracellular macromolecular crowding and gene delivery. However, it is demonstrated in other research that the cellular behavior can be obviously different in 2D and in 3D culture models66. For

instance, Burdick et al.67 found that MSCs displayed enhanced cell spreading and more YAP/TAZ

translocated into the nucleus when cells are grown on the surface of stiffer hydrogels; however, the complete reverse trend was detected when cells were seeding within hydrogels with 3D structure, highlighting the important role of 3D structure for in vitro investigations. In the future work, it would

be fascinating to couple topography with 3D micro niches for better mimicking natural tissue structure to explore the influence on cell functions and translation towards commercial uses and clinic application.

(9)

116

6.2 References

(1) Jenkins, T. L.; Little, D. Synthetic Scaffolds for Musculoskeletal Tissue Engineering: Cellular Responses to Fiber Parameters. npj Regen. Med. 2019, 4 (1), 1–14.

(2) Huang, G.; Li, F.; Zhao, X.; Ma, Y.; Li, Y.; Lin, M.; Jin, G.; Lu, T. J.; Genin, G. M.; Xu, F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem. Rev. 2017, 117 (20), 12764–12850.

(3) Gvaramia, D.; Müller, E.; Müller, K.; Atallah, P.; Tsurkan, M.; Freudenberg, U.; Bornhäuser, M.; Werner, C. Combined Influence of Biophysical and Biochemical Cues on Maintenance and Proliferation of Hematopoietic Stem Cells. Biomaterials 2017, 138, 108–117.

(4) Zhou, Q.; Ge, L.; Guimarães, C. F.; Kühn, P. T.; Yang, L.; van Rijn, P. Development of a Novel Orthogonal Double Gradient for High-Throughput Screening of Mesenchymal Stem Cells– Materials Interaction. Advanced Materials Interfaces. 2018, 18 (5), 4-11.

(5) Yang, L.; Ge, L.; Zhou, Q.; Jurczak, K. M.; van Rijn, P. Decoupling the Amplitude and Wavelength of Anisotropic Topography and the Influence on Osteogenic Differentiation of Mesenchymal Stem Cells Using a High-Throughput Screening Approach. ACS Appl. Bio Mater. 2020, 3 (6), 3690–3697.

(6) Amin, Y. Y. I.; Runager, K.; Simoes, F.; Celiz, A.; Taresco, V.; Rossi, R.; Enghild, J. J.; Abildtrup, L. A.; Kraft, D. C. E.; Sutherland, D. S.; et al. Combinatorial Biomolecular Nanopatterning for High-Throughput Screening of Stem-Cell Behavior. Adv. Mater. 2016, 28 (7), 1472–1476.

(7) Yarrow, J. C.; Perlman, Z. E.; Westwood, N. J.; Mitchison, T. J. A High-Throughput Cell Migration Assay Using Scratch Wound Healing, a Comparison of Image-Based Readout Methods. BMC Biotechnol. 2004, 4, 1–9.

(8) Li, X.; MacEwan, M. R.; Xie, J.; Siewe, D.; Yuan, X.; Xia, Y. Fabrication of Density Gradients of Biodegradable Polymer Microparticles and Their Use in Guiding Neurite Outgrowth. Adv. Funct. Mater. 2010, 20 (10), 1632–1637.

(9) Mohan, G.; Gallant, N. D. Surface Chemistry Gradients on Silicone Elastomers for High-Throughput Modulation of Cell-Adhesive Interfaces. J. Biomed. Mater. Res. - Part A 2015, 103 (6),

2066–2076.

(10) Sun, J.; Ding, Y.; Lin, N. J.; Zhou, J.; Ro, H.; Soles, C. L.; Cicerone, M. T.; Lin-gibson, S. Nanogratings. 2010, 3067–3072.

(11) Kim, T. H.; An, D. B.; Oh, S. H.; Kang, M. K.; Song, H. H.; Lee, J. H. Creating Stiffness Gradient Polyvinyl Alcohol Hydrogel Using a Simple Gradual Freezing-Thawing Method to Investigate Stem Cell Differentiation Behaviors. Biomaterials 2015, 40, 51–60.

(12) Ge, L.; Yang, L.; Bron, R.; Burgess, J. K.; Van Rijn, P. Topography-Mediated Fibroblast Cell Migration Is Influenced by Direction, Wavelength, and Amplitude. ACS Appl. Bio Mater. 2020, 3 (4),

2104–2116.

(13) Hadden, W. J.; Young, J. L.; Holle, A. W.; McFetridge, M. L.; Kim, D. Y.; Wijesinghe, P.; Taylor-Weiner, H.; Wen, J. H.; Lee, A. R.; Bieback, K.; et al. Stem Cell Migration and Mechanotransduction on Linear Stiffness Gradient Hydrogels. Proc. Natl. Acad. Sci. 2017, 201618239.

(14) Teo, A. J. T.; Mishra, A.; Park, I.; Kim, Y. J.; Park, W. T.; Yoon, Y. J. Polymeric Biomaterials for Medical Implants and Devices. ACS Biomater. Sci. Eng. 2016, 2 (4), 454–472.

(15) Ge, L.; Yang, L.; Bron, R.; K. Burgess, J.; van Rijn, P. Topography-Mediated Fibroblast Cell Migration Is Influenced by Direction, Wavelength, and Amplitude. ACS Appl. Bio Mater. 2020, 3 (4),

2104–2116.

(16) Hale, N. A.; Yang, Y.; Rajagopalan, P. Cell Migration at the Interface of a Dual Chemical-Mechanical Gradient. ACS Appl. Mater. Interfaces 2010, 2 (8), 2317–2324.

(10)

117 Electrical Currents Stimulation Promotes Both Proliferation and Differentiation of Fetal Neural Stem Cells. 2011, 6 (4).

(18) Ren, T.; Mao, Z.; Guo, J.; Gao, C. Directional Migration of Vascular Smooth Muscle Cells Guided by a Molecule Weight Gradient of Poly(2-Hydroxyethyl Methacrylate) Brushes. Langmuir 2013, 29

(21), 6386–6395.

(19) Han, L.; Mao, Z.; Wu, J.; Guo, Y.; Ren, T.; Gao, C. Directional Cell Migration through Cell-Cell Interaction on Polyelectrolyte Multilayers with Swelling Gradients. Biomaterials 2013, 34 (4), 975–984.

(20) Maheshwari, G.; Wells, A.; Griffith, L. G.; Lauffenburger, D. A. Biophysical Integration of Effects of Epidermal Growth Factor and Fibronectin on Fibroblast Migration. Biophys. J. 1999, 76 (5), 2814–

2823.

(21) Liu, Y.; Li, Y.; Li, N.; Teng, W.; Wang, M.; Zhang, Y.; Xiao, Z. TGF-Β1 Promotes Scar Fibroblasts Proliferation and Transdifferentiation via up-Regulating MicroRNA-21. Sci. Rep. 2016, 6, 32231.

(22) Mörke, C.; Rebl, H.; Finke, B.; Dubs, M.; Nestler, P.; Airoudj, A.; Roucoules, V.; Schnabelrauch, M.; Körtge, A.; Anselme, K.; et al. Abrogated Cell Contact Guidance on Amino-Functionalized Microgrooves. ACS Appl. Mater. Interfaces 2017, 9 (12), 10461–10471.

(23) Mao, Z.; Han, L.; Tan, H.; Wu, J.; Gao, C.; Ren, T. Gradient Biomaterials and Their Influences on Cell Migration. Interface Focus 2012, 2 (3), 337–355.

(24) Kim, H. N.; Hong, Y.; Kim, M. S.; Kim, S. M.; Suh, K. Y. Effect of Orientation and Density of Nanotopography in Dermal Wound Healing. Biomaterials 2012, 33 (34), 8782–8792.

(25) Zhou, Q.; Kuhn, P. T.; Huisman, T.; Nieboer, E.; van Zwol, C.; van Kooten, T. G.; van Rijn, P. Directional Nanotopographic Gradients: A High-Throughput Screening Platform for Cell Contact Guidance. Sci. Rep. 2015, 5 (16240), 16240.

(26) Zhou, Q.; Guimarães, C. F.; Kühn, P. T.; Van Kooten, T. G.; Van Rijn, P.; Castañeda Ocampo, O. Screening Platform for Cell Contact Guidance Based on Inorganic Biomaterial Micro/Nanotopographical Gradients. ACS Appl. Mater. Interfaces 2017, 9 (37), 31433–31445.

(27) L. Yang, K. M. Jurczak, L. Ge, P. van Rijn*. High throughput screening and hierarchical topography-mediated neural differentiation of mesenchymal stem cells. Adv. Healthcare Mater. 2020, 2000117.

(28) Yang, L.; Ge, L.; Zhou, Q.; Jurczak, K. M.; Van Rijn, P. Decoupling the Amplitude and Wavelength of Anisotropic Topography and the Influence on Osteogenic Differentiation of Mesenchymal Stem Cells Using a High-Throughput Screening Approach. ACS Appl. Bio Mater. 2020, 3 (6), 3690–3697.

(29) Kim, J.; Bae, W. G.; Kim, Y. J.; Seonwoo, H.; Choung, H. W.; Jang, K. J.; Park, S.; Kim, B. H.; Kim, H. N.; Choi, K. S.; et al. Directional Matrix Nanotopography with Varied Sizes for Engineering Wound Healing. Adv. Healthc. Mater. 2017, 6 (19), 1–10.

(30) Engler, A. J.; Sen, S.; Sweeney, H. L.; Discher, D. E. Matrix Elasticity Directs Stem Cell Lineage Specification. Cell 2006, 126 (4), 677–689.

(31) Teixeira, S. P. B.; Domingues, R. M. A.; Shevchuk, M.; Gomes, M. E.; Peppas, N. A.; Reis, R. L. Biomaterials for Sequestration of Growth Factors and Modulation of Cell Behavior. Adv. Funct. Mater. 2020, 1909011, 1–26.

(32) Yang, L.; Gao, Q.; Ge, L.; Zhou, Q.; Warszawik, E. M.; Bron, R.; Lai, K. W. C.; van Rijn, P. Topography Induced Stiffness Alteration of Stem Cells Influences Osteogenic Differentiation.

Biomater. Sci. 2020, 8, 2638–2652.

(33) Fan, C.; Li, X.; Xiao, Z.; Zhao, Y.; Liang, H.; Wang, B.; Han, S.; Li, X.; Xu, B.; Wang, N.; et al. A Modified Collagen Scaffold Facilitates Endogenous Neurogenesis for Acute Spinal Cord Injury Repair. Acta Biomater. 2017, 51, 304–316.

(34) Xue, Y.; Liu, D.; Wang, C.; Bao, C.; Wang, X.; Zhu, H.; Mao, H.; Cai, Z.; Lin, Q.; Zhu, L. Photo and Reduction Dual-Responsive Hydrogel for Regulating Cell Adhesion and Cell Sheet Harvest. ACS Appl. Bio Mater. 2020, 3 (4), 2410–2418.

(11)

118

(35) Lee, H.; Kim, W. J.; Lee, J. U.; Yoo, J. J.; Kim, G. H.; Lee, S. J. Effect of Hierarchical Scaffold Consisting of Aligned DECM Nanofibers and Poly(Lactide- Co-Glycolide) Struts on the Orientation and Maturation of Human Muscle Progenitor Cells. ACS Appl. Mater. Interfaces 2019, 11 (43), 39449–

39458.

(36) Li, Y.; Xiao, Y.; Liu, C. The Horizon of Materiobiology: A Perspective on Material-Guided Cell Behaviors and Tissue Engineering. Chem. Rev. 2017, 117 (5), 4376–4421.

(37) Zhou, H.-X.; Rivas, G.; Minton, A. P. Macromolecular Crowding and Confinement: Biochemical, Biophysical, and Potential Physiological Consequences. Annu. Rev. Biophys. 2008, 37 (1), 375–397.

(38) Lindner, R. A.; Ralston, G. B. Macromolecular Crowding: Effects on Actin Polymerisation.

Biophysical Chemistry. 1997, 57–66.

(39) Prewitz, M. C.; Stißel, A.; Friedrichs, J.; Träber, N.; Vogler, S.; Bornhäuser, M.; Werner, C. Extracellular Matrix Deposition of Bone Marrow Stroma Enhanced by Macromolecular Crowding.

Biomaterials 2015, 73, 60–69.

(40) Seo, C. H.; Jeong, H.; Feng, Y.; Montagne, K.; Ushida, T.; Suzuki, Y.; Furukawa, K. S. Micropit Surfaces Designed for Accelerating Osteogenic Differentiation of Murine Mesenchymal Stem Cells via Enhancing Focal Adhesion and Actin Polymerization. Biomaterials 2014, 35 (7), 2245–2252.

(41) Yang, L.; Yang, L.; Ge, L.; Ge, L.; Van Rijn, P.; Van Rijn, P. Synergistic Effect of Cell-Derived Extracellular Matrices and Topography on Osteogenesis of Mesenchymal Stem Cells. ACS Appl. Mater. Interfaces 2020, 12 (23), 25591–25603.

(42) Yang, L.; Ge, L.; Zhou, Q.; Mokabber, T.; Pei, Y.; Bron, R.; van Rijn, P. Biomimetic Multiscale Hierarchical Topography Enhances Osteogenic Differentiation of Human Mesenchymal Stem Cells.

Adv. Mater. Interfaces 2020, 7 (14), 1–12.

(43) Wang, Q.; Chen, B.; Cao, M.; Sun, J.; Wu, H.; Zhao, P.; Xing, J.; Yang, Y.; Zhang, X.; Ji, M.; et al. Response of MAPK Pathway to Iron Oxide Nanoparticles in Vitro Treatment Promotes Osteogenic Differentiation of HBMSCs. Biomaterials 2016, 86, 11–20.

(44) Liu, X.; Shi, S.; Feng, Q.; Bachhuka, A.; He, W.; Huang, Q.; Zhang, R.; Yang, X.; Vasilev, K. Surface Chemical Gradient Affects the Differentiation of Human Adipose-Derived Stem Cells via ERK1/2 Signaling Pathway. ACS Appl. Mater. Interfaces 2015, 7 (33), 18473–18482.

(45) Boersma, A. J.; Zuhorn, I. S.; Poolman, B. A Sensor for Quantification of Macromolecular Crowding in Living Cells. Nat. Methods 2015, 12 (3), 227–229.

(46) Atienza, J. M.; Zhu, J.; Wang, X.; Xu, X.; Abassi, Y. Dynamic Monitoring of Cell Adhesion and Spreading on Microelectronic Sensor Arrays. J. Biomol. Screen. 2005, 10 (8), 795–805.

(47) Xu, Z.; Baek, K. H.; Kim, H. N.; Cui, J.; Qian, X.; Spring, D. R.; Shin, I.; Yoon, J. Zn2+-Triggered Amide Tautomerization Produces a Highly Zn 2+-Selective, Cell-Permeable, and Ratiometric Fluorescent Sensor. J. Am. Chem. Soc. 2010, 132 (2), 601–610.

(48) Guo, C. X.; Ng, S. R.; Khoo, S. Y.; Zheng, X.; Chen, P.; Li, C. M. RGD-Peptide Functionalized Graphene Biomimetic Live-Cell Sensor for Real-Time Detection of Nitric Oxide Molecules. ACS Nano 2012, 6 (8), 6944–6951.

(49) Domaille, D. W.; Que, E. L.; Chang, C. J. Synthetic Fluorescent Sensors for Studying the Cell Biology of Metals. Nat. Chem. Biol. 2008, 4 (3), 168–175.

(50) Weart, R. B.; Lee, A. H.; Chien, A. C.; Haeusser, D. P.; Hill, N. S.; Levin, P. A. A Metabolic Sensor Governing Cell Size in Bacteria. Cell 2007, 130 (2), 335–347.

(51) Dimde, M.; Neumann, F.; Reisbeck, F.; Ehrmann, S.; Cuellar-Camacho, J. L.; Steinhilber, D.; Ma, N.; Haag, R. Defined PH-Sensitive Nanogels as Gene Delivery Platform for SiRNA Mediated: In Vitro Gene Silencing. Biomater. Sci. 2017, 5 (11), 2328–2336.

(52) Elnathan, R.; Kwiat, M.; Patolsky, F.; Voelcker, N. H. Engineering Vertically Aligned Semiconductor Nanowire Arrays for Applications in the Life Sciences. Nano Today 2014, 9 (2), 172–196.

(12)

119 (53) Senapati, S.; Sarkar, T.; Das, P.; Maiti, P. Layered Double Hydroxide Nanoparticles for Efficient

Gene Delivery for Cancer Treatment. Bioconjug. Chem. 2019, 30 (10), 2544–2554.

(54) Shekhar, S.; Lee, B.; Roy, A.; Candiello, J.; Kumta, P. N. Surface Mediated Non-Viral Gene Transfection on Titanium Substrates Using Polymer Electrolyte and Nanostructured Silicate Substituted Calcium Phosphate PDNA (NanoSiCaPs) Composites. Mater. Today Commun. 2018, 16

(March), 169–173.

(55) Ong, W.; Lin, J.; Bechler, M. E.; Wang, K.; Wang, M.; ffrench-Constant, C.; Chew, S. Y. Microfiber Drug/Gene Delivery Platform for Study of Myelination. Acta Biomater. 2018, 75, 152–160.

(56) Liangliang Yang, Klaudia Malgorzata Jurczak, Lu Ge, P. van R. High Throughput Screening and Hierarchical Topography-Mediated Neural Differentiation of Mesenchymal Stem Cells. Adv. Healthc. Mater. 2020,11(9), 2000117.

(57) Yang, L.; Ge, L.; Zhou, Q.; Mokabber, T.; Pei, Y.; Bron, R.; van Rijn, P. Biomimetic Multiscale Hierarchical Topography Enhances Osteogenic Differentiation of Human Mesenchymal Stem Cells.

Adv. Mater. Interfaces. 2020, 14 (7), 1-12.

(58) Yang, Y.; Wang, X.; Hu, X.; Kawazoe, N.; Yang, Y.; Chen, G. Influence of Cell Morphology on Mesenchymal Stem Cell Transfection. ACS Appl. Mater. Interfaces 2019, 11 (2), 1932–1941.

(59) Wang, P. Y.; Lian, Y. S.; Chang, R.; Liao, W. H.; Chen, W. S.; Tsai, W. B. Modulation of PEI-Mediated Gene Transfection through Controlling Cytoskeleton Organization and Nuclear Morphology via Nanogrooved Topographies. ACS Biomater. Sci. Eng. 2017, 3 (12), 3283–3291.

(60) Mantz, A.; Pannier, A. K. Biomaterial Substrate Modifications That Influence Cell-Material Interactions to Prime Cellular Responses to Nonviral Gene Delivery. Exp. Biol. Med. 2019, 244 (2),

100–113.

(61) Nobes, C. D.; Hall, A. Rho, Rac, and Cdc42 GTPases Regulate the Assembly of Multimolecular Focal Complexes Associated with Actin Stress Fibers, Lamellipodia, and Filopodia. Cell 1995, 81 (1),

53–62.

(62) Kong, H. J.; Liu, J.; Riddle, K.; Matsumoto, T.; Leach, K.; Mooney, D. J. Non-Viral Gene Delivery Regulated by Stiffness of Cell Adhesion Substrates. Nat. Mater. 2005, 4 (6), 460–464.

(63) Kelly, A. M.; Plautz, S. A.; Zempleni, J.; Pannier, A. K. Glucocorticoid Cell Priming Enhances Transfection Outcomes in Adult Human Mesenchymal Stem Cells. Mol. Ther. 2016, 24 (2), 331–341.

(64) Madl, C. M.; Heilshorn, S. C. Engineering Hydrogel Microenvironments to Recapitulate the Stem Cell Niche. Annu. Rev. Biomed. Eng. 2018, 20, 21–47.

(65) Humphrey, J. D.; Dufresne, E. R.; Schwartz, M. A. Mechanotransduction and Extracellular Matrix Homeostasis. Nat. Rev. Mol. Cell Biol. 2014, 15 (12), 802–812.

(66) Baker, B. M.; Chen, C. S. Deconstructing the Third Dimension-How 3D Culture Microenvironments Alter Cellular Cues. Journal of Cell Science. 2012, 3015–3024.

(67) Caliari, S. R.; Vega, S. L.; Kwon, M.; Soulas, E. M.; Burdick, J. A. Dimensionality and Spreading Influence MSC YAP/TAZ Signaling in Hydrogel Environments. Biomaterials 2016, 103, 314–323.

(13)

Referenties

GERELATEERDE DOCUMENTEN

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright

The general aim of this thesis is to explore topography-mediated alterations to cell behavior using cell-materials interfaces, and investigate subcellular behaviors like

Except for the wrinkle wavelength and amplitude, another essential factor investigated was the cell migration with respect to the orientation of the topography as it has

Overall, the work presented in this thesis is to explore topography-mediated alterations of cell behaviors and investigate cell-material interface-induced subcellular behaviors

In dit proefschrift, om topografie-gemedieerde alteraties te verkennen en om sub- cellulair gedrag te onderzoeken, demonstreren we de belangrijke rol van topografie in het

Topographic stimuli are essential for cell function modulation and play an important role in regulating cell migration, intracellular crowding and gene-delivery. Topography gradient

In the randomized case, linear convergence is established when the cost function is strongly convex, yet with no convexity requirements on the individual functions in the sum.. For

In recent years, researchers have developed high-throughput (HT) approaches that enable the analysis of hundreds to thousands of interactions among biomaterials, biomolecules, and