• 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!
17
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)

1

CHAPTER 1

General Introduction & Aim of this

Thesis

(3)

2

1.1

Cell and material interfaces

The acceleration of aging population and human pursuit of health and longevity have stimulated

society's demand for the development of tissue engineering and regenerative medicine

1

. With the

vigorous development and major breakthroughs of regenerative medicine technology, biomaterials

occupy a very important position for providing a suitable therapeutic option with a reduced risk of

disease transmission, infection, and immunogenicity, and limitless availability

2,3

. Although

biochemical factors (e.g., growth factors

4

, hormones) regulate cells and tissue functions, the

cell-materials interactions, especially the biophysical effect of cell-materials, determines cell functions in

development, physiology, and pathophysiology and remains a central endeavor in tissue engineering

5.

1.1.1 The cellular microenvironment

Cells are known to reside in a highly dynamic and extremely complicated three-dimensional (3D)

microenvironment, which not only serves as structural support but also provides diverse biochemical

and biophysical cues, that regulate cell functions and development

6

. The cell microenvironment

(

Figure 1) are normally including neighboring cells with highly structured and heterogeneous mix,

soluble factors, extracellular matrix (ECM), and biophysical stimuli, e.g. mechanical property, 2D

topography, and 3D geometry strongly influence cell behaviors such as cell adhesion, spreading,

proliferation, cell alignment, migration, and the differentiation or self-maintenance of stem cells

7

.

Figure 1. Schematic illustration of the mean components of cellular microenvironment. Adapted with permission from ref5.

The interaction between neighboring cells plays an important role to determine the physiology and

cell behaviors of living cells

8

. For instance, Ding et al. have demonstrated that matrix stiffness and

cell-cell contact interplay in the mesenchymal stem cells (MSC) differentiation decision

9

. In addition,

others illustrate that co-culture of MSC and osteoblasts could change the osteogenic differentiation

10

.

What’s more, some recent work has used peptide nanofibers to mimic the cell-cell interaction

(4)

3

indicator N-cadherin, which can facilitate MSC differentiation into chondrogenic lineage

11

. Nowadays,

ECM has attracted extensive interest for the regulation of cell behaviors

12

. As it is well known that

cells can sense and respond to surrounding ECM in which they reside, the components of ECM and

ECM-like components such as collagen

13

, laminin

14

, gelatin

15

and fibronectin

16

are crucial for the

regulation of cell spreading, proliferation, migration, and differentiation

17,18

. For instance, fibronectin

and laminin are binding to their neighboring cells and other ECM proteins on the nanoscale, thus

initiate various of intracellular signaling pathways

19

. Some interesting work has shown that mechanical

properties such as changing ECM stiffness is needed for reprogramming normal cells into tumor

precursors

20

.

It is generally accepted that chemical or biochemical factors like –COOH

21

, −NH2

22

, growth factors

23

,

or hormones are critical for a number of biological activities. For instance, the platelet-derived growth

factors can recruit dermal fibroblasts crusting to the wound site of injury

24

. And some studies

demonstrate that collagen hydrogel incorporated with graphene oxide (GO) absorbed transforming

growth factor β3 (TGF-β3) regulating MSCs differentiation into chondrogenic lineage

25

. Except for

the chemical factors, physical stimuli like strain and stress, magnetic, electrical

26

, thermal

27

, light

28

,

stiffness, and topography also serve as an important signal for regulating cell shape, elongation

29

,

migration

30

, proliferation, and stem cell differentiation. In this section, we will mainly focus on

cell-materials interactions especially the topography influence of cell-materials determines cell behaviors, like

migration, macromolecular crowding, and gene-deliver efficiency.

1.1.2 Topography stimuli

Researchers increasingly highlight the essential role of nano-/micro- scale topographic structure on

the profound regulation of cell behaviors

31–33

. Cells in vivo experience, sense, and respond to their

surrounding physical cues from few nanometers to hundreds of micrometers by contact guidance,

and translate these physical stimuli into intracellular signals through mechanical transduction

34

. The

mechanical signals modulated mainly through direct interactions of integrin clustering, activates focal

adhesion and RhoA/ROCK

35,36

pathway, further induce cell skeleton tension and cell morphology

change, thereby altering relative gene expression to regulate cell functions (e.g., cell adhesion,

alignment, proliferate, migrate and differentiation)

37

.

The natural tissues like bone, tendon

38

, and nerve

39

have anisotropic hierarchical structures with

nano-/micro- sized features. To better mimic the natural structure of extracellular matrix and prepare

substrate with different topographic cues, numerous fabrication methods have been developed. For

example, microcontact printing

40

, photolithography

41

, photopatterning

42

, electrospinning

43

,

microfluidics-assisted patterning

44

, plasma oxidation

45

for the purpose to developed the 2D substrates

or hydrogels

46

for mimicking the more natural 3D geometry, etc. Understanding interaction between

topographic cues and cells, as well as the modulation of specific biological functions is still a challenge

for materiobiology and tissue engineering

47

.

Extensive research has fabricated different kinds of structures, normally categorized into two classes:

isotropic structure (e.g., roughness

48

, porosity

49

) and anisotropic ones (e.g., grating

50

, pillar

51

, fibers

52

,

wrinkle

53

), as well as 3D geometry cues. The mechanical properties of substrates can regulate the

mechanical transduction of cells and further have an influence on the cell adhesion, cell shape and

cytoskeletal architecture that are altered by micro/nanopatterns on the surface of a substrate. The

nano- and micro- sized architecture is crucial for cell functions (adhesion, migration, proliferate and

differentiation) modulation

53,54

. For instance, previous studies have indicated that the anisotropic

architectures of grooves endow cell alignment to the substrate, and the alignment of nanofibers can

promote neuron and myoblasts maturation and differentiation

55

. In

Figure 2 some typical cell

adhesion distribution and cell skeletal alteration are shown that have been identified previously.

(5)

4

In addition, in heart tissue engineering, the nanoscale cues of hydrogels can induce anisotropic cell

behavior and contractility characteristics as observed by cells in their native environment, which gives

guidance information for heart tissue repair

56

. Others works have shown that in an

in vitro wound

healing procedure the fibroblasts showed elongated shape along the nanogroove and cell migration

rate was regulated by the substrate features, thus nano-groove size and density are important

considerations for tissue engineering scaffold design

57

. What’s more, the 3D geometry developed by

changing the pores or cross-link ratio the geometry is able to regulate stem cell differentiation

behaviors

58

. Taken together, the topography is essential for cell function modulation, learning cell and

materials-interfaces is the key point for tissue engineering and disease therapy. In this thesis, we

mainly focus on the topography influence on cell behaviors, like migration, macromolecular crowding,

and gene-deliver efficiency.

Figure 2. (A)(B) Micro-/nanopatterned substrates regulate the distribution of integrin-mediated adhesions, cell shape, cytoskeletal architecture and multicellular organization, (C, D) Further controlled by adjusting the material stiffness, (E, F) Nano-topological features to regulate cell–matrix adhesions for the manipulation of the size and geometry of cells cultured on them. Adapted with permission from ref7.

1.1.3 Cell migration

Recent studies focus more on the cell migration behaviors, for the reason that cell movement is

essential for numerous physiological and pathological processes such as embryonic development,

angiogenesis, immune surveillance, cancer metastasis, tissue regeneration, and wound healing

59

. The

topography plays a key role in affecting cell behaviors like cell adhesion, alignment

45

, proliferation,

migration

53

, and differentiation

54

. The interfaces between cell and topographic cues is essential for the

modulation of cell migration in wound healing procedure. Wound healing is a complex biological

(6)

5

process and the typical wound healing procedure mainly including four phases as shown in

Figure 3,

including: (1) Hemostasis phase, (2) Inflammation phase, (3) Cell migration/proliferation, and (4)

Remodeling phase. In the hemostasis phase, the platelets modulate the process by interacting with

subendothelial matrix proteins and tissue factor-bearing cells to move fast to the wound site, and

clustering to the platelet plug and releasing various growth factors and matrix remolding enzymes

60

.

In the inflammation phase, the quick movement of immune cells like macrophages or neutrophils is

the key factor for the clean-up of dead cells and bacteria and then release growth factors promoting

the fibroblasts cell migration. In the cell migration/proliferation procedure, the fibroblasts move to

the wound site and proliferate in the wound site to remodel the tissues, afterward, epithelial cells

migrate on the wound edge to cover the defect

61

.

Figure 3. A schematic depicting the process of wound healing, including four continuous phases-homeostasis, inflammation, proliferation, and remodeling. Adapted with permission from ref61.

Learning about cell migration is critical for the wound healing procedure. The cell migration has been

shown to be directed by chemical factors

62

, cell density

63

, molecular signals

64

, stiffness

65

, and

topography

66

. The design and manipulation of topographic biomaterials play a pivotal role in

controlling cell migration and avoid scar formation. For instance, C2C12 cells grown on suspended

fiber networks showed higher migration speed on the attached and aligned adhesion site

67

. Others

showed that fibroblast migration speed is influenced by the density of the nano-topographic pattern

as well as on the width or depth of the groove

68

. In addition, the collective migration of

(7)

osteoblast-6

like cells (MG-63) and human mesenchymal stem cells were modulated by smaller groove depth in

an

in vitro fracture healing model

69

. What’s more, some interesting work demonstrate that not only

the topographical density but also the orientation of the nanogroove distinctly regulate NIH-3T3 cell

migration speed, cell division, and ECM production in dermal wound healing procedure

70

. Taken

together, the design of topographic material is crucial for understanding cell-matrix interactions and

provide guiding information for tissue repair.

1.1.4 Macromolecular crowding

Learning about the cell-material interfaces is critical for many processes and the interface greatly

influences intracellular mechanisms and phenomena. The cytoplasm is always heterogeneous and

highly volume-occupied with biomolecules, like various nutrients, proteins, nucleic acids, enzymes,

intermediate metabolites, and other macromolecular monomers/components. The concentration of

macromolecular components that can be reached as high as 50-400 mg ml

−1

in the crowded and

confined spaces

71

. Accordingly, the high concentrations of macromolecular components inside the

cell affects the function of molecular chaperones, polypeptide chains and oligomeric proteins

folding

72

, improve enzyme reaction rate

73

, and metabolic activity

74

. Recently, numerous studies focus

on the addition of some natural or synthetic polymers like Ficoll

75

, dextran, poly (N-vinylpyrrolidone)

(PVP)

76

, polyethylene glycol (PEG)

77

or bovine serum album in the culture media to artificially control

and enhance the macromolecular crowding inside the cell and study how it affects cell behaviors. For

instance, the addition of carbohydrate-based macromolecules can dramatically enhance stem cells

extracellular matrix production and further influence the cell skeleton alignment, proliferation and

differentiation

78

. Also, the macromolecular crowding is applied to material synthesis, and molecular

self-assembly

79

.

For the purpose of exploring macromolecular crowding inside the cell, some excellent work has been

done on developing a sensor to directly determine the crowding inside the cell

80,81

. For instance,

Boersma and coworkers developed a FRET-based sensor, which has the FRET pair m-Cerulean (cyan

fluorescent protein) and m-Citrine (yellow fluorescent protein) positioned at the N terminus and C

terminus, respectively, for directly measuring the crowding inside living cells. The schematic picture

of the crowding sensor is shown in

Figure 4. The changes in fluorescence are a result from FRET

efficiency adjustment, which increases with the increase of macromolecular crowding. The sensor

readout is reversible, and only sensitive to the macromolecular crowding induced excluded volume

82

.

Topography has an impact on various cell behaviors like cell adhesion, alignment, proliferation,

migration, and stem cell differentiation. Therefore, we believe that the spatiotemporal readout of

crowding is a compelling tool for better understanding cell and biomaterials interactions and allow us

to investigate the role of macromolecular crowding of the cytoplasm during the cell development

stages.

(8)

7

Figure 4. Design and characterization of macromolecular crowding sensor inside cells. Adapted with permission from ref82.

1.1.5 Gene delivery

Nowadays, gene delivery has gained much attention for the purpose of delivery therapeutic genes or

introduce artificial modified genes into cells for modifying the cell function

83

or for specific clinical

purposes

84

. In recent decades, efforts have mostly focused on applications of biosensors

85

, diagnostic

devices, cancer therapy

86

, tissue regeneration

87

, and vaccine development therapy

88

. It is well

established that traditional gene delivery systems like electroporation

89

, magnetofection

90

, ultrasound

91

,

or viral vectors

92,93

need expensive hardware, are time consuming, have high toxicity problems or

possibly induce immunogenicity. Exploring high-efficiency gene carriers with low toxicity is still a

challenge for gene delivery applications.

Due to the remarkable development in nanotechnology, considerable excellent works have been

devoted to constructing non-toxic delivery systems with the basic concepts of low toxicity and high

transfection efficiency. For instance, gene carriers like polyethyleneimine (PEI)

94

, lipofectamine

95

,

poly-amidoamine (PAMAM)

96

, silica-based nanoparticles (SNPs)

97–99

, and poly-lysine (PLL)

100

are

commonly employed for systemic administration. For instance, spikey nanoparticles demonstrate

higher transfection efficiency than the hemisphere- and bowl-type subunit nanoparticles and avoids

enzymatic cleavage

101

. The vehicles enter the cell membrane by mimicking functions of viral agents

that enable stronger binding affinity but avoid the immune potential and toxicity risks of viral

vectors

101

. It remains challenging to develop highly active polymers to achieved high transfection

efficiency with lower toxicity.

It is important to highlight that present research suggest that substrate-mediated gene delivery plays

a critical role in gene delivery systems owing to diverse physiochemical properties and good

biocompatibility. For instance, vertical silicon nanowires

102,103

, silicon nano- to microscale pillars

104

,

aligned hollow carbon nanotubes

105

, and nano-grooves

106

play critical roles in gene delivery. For

example, some work observed that 200 nm nanopillars can improve human mesenchymal stem cells

(hMSCs) transfection efficiency

107

. Some other interesting work found that different shape of pillars

showed different gene transfection behaviors and the pillars can penetrate into cell membrane while

maintaining cell viability

108

. In another study it was found that nanogrooves influence gene

transfection by controlling cytoskeleton organization and nuclei morphology

106

. Taken together, the

substrate-mediated gene delivery is promising for non-viral gene transfer in which topography as

physicochemical stimulus is able to drive and influence cell function, which is crucial for

understanding how material interfaces can be applied to enhance gene delivery.

(9)

8

1.2 Aim of this thesis

The general aim of this thesis is to explore topography-mediated alterations to cell behavior using

cell-materials interfaces, and investigate subcellular behaviors like cell morphology alteration, cell

migration in wound healing procedure, inner cell macromolecular crowding induced by

nano-/micro- patterns, and topography modulated gene delivery of stem cells. For this purpose,

wrinkle-gradient substrates were fabricated with diverse wavelength and amplitude parameters to make it

possible for investigating the topographic cues as well as direction of wave-like topography on the

cell migration behavior in wound healing. In addition, uniform wrinkle surfaces were developed

with diverse wavelength and amplitude parameters to study the topography influenced cell

macromolecular crowding and identifying the possible related signaling of cell shape alterations,

mechanical transduction, metabolic activity and protein expression. Furthermore, the uniform

wrinkle substrate were investigated as possible stimulation to control or even enhance gene delivery

capacity of stem cells. Topography-induced improvement of transfection efficiency and endocytic

capacity were investigated.

1.3 Outline of this thesis

Chapter 1 gives a basic introduction of the cell micro environment and the essential factors for

cell modulation, like ECM, chemical signals, cell-cell contact, physical stimuli (stiffness and

topography). And the cell behaviors like migration, macromolecular crowding and gene delivery.

In

Chapter 2, we report on recent progress of the gradient platforms, as also partly used in this

thesis, to study bio-interfaces and the effects of physicochemical stimuli on e.g., cell adhesion, cell

morphology, and migration.

In

Chapter 3, wrinkle topography gradients were developed where especially the wavelength and

amplitude were decoupled as such that the topographies, the wavelength and amplitude, both

function as separate parameter and direction-induced cell migration, proliferation and adhesion

was investigated.

In

Chapter 4, uniform wrinkle surfaces were developed to investigate the role of topography on

cell macromolecular crowding and identifying the possible mechanisms responsible for altered

macromolecular crowding inside the cell.

In

Chapter 5, topography-mediated gene delivery of stem cells is explored and it is investigated

how topography may induce improvement of transfection efficiency and endocytic capacity.

This thesis finalizes with a general conclusion and discussion on how this thesis impacts the current

scientific knowledge (

Chapter 6).

(10)

9

1.4 References

(1) Chen, F. M.; Liu, X. Advancing Biomaterials of Human Origin for Tissue Engineering. Prog. Polym. Sci. 2016, 53, 86–168.

(2) Zhu, M.; Li, W.; Dong, X.; Yuan, X.; Midgley, A. C.; Chang, H.; Wang, Y.; Wang, H.; Wang, K.; Ma, P. X.; et al. In Vivo Engineered Extracellular Matrix Scaffolds with Instructive Niches for Oriented Tissue Regeneration. Nat. Commun. 2019, 10 (1).

(3) Giannoudis, P. V.; Capanna, R. Bone tissue engineering and bone regeneration Tissue. Indury. 2006, 37(3), 917-936.

(4) 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.

(5) 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.

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

(7) 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.

(8) Mao, A. S.; Shin, J. W.; Mooney, D. J. Effects of Substrate Stiffness and Cell-Cell Contact on Mesenchymal Stem Cell Differentiation. Biomaterials 2016, 98, 184–191.

(9) Ye, K.; Cao, L.; Li, S.; Yu, L.; Ding, J. Interplay of Matrix Stiffness and Cell-Cell Contact in Regulating Differentiation of Stem Cells. ACS Appl. Mater. Interfaces 2016, 8 (34), 21903–21913. (10) Csaki, C.; Matis, U.; Mobasheri, A.; Shakibaei, M. Co-Culture of Canine Mesenchymal Stem Cells

with Primary Bone-Derived Osteoblasts Promotes Osteogenic Differentiation. Histochem. Cell Biol. 2009, 131 (2), 251–266.

(11) Eren Cimenci, C.; Kurtulus, G. U.; Caliskan, O. S.; Guler, M. O.; Tekinay, A. B. N-Cadherin Mimetic Peptide Nanofiber System Induces Chondrogenic Differentiation of Mesenchymal Stem Cells. Bioconjug. Chem. 2019, 30 (9), 2417–2426.

(12) Park, J.; Kim, D.-H.; Levchenko, A. Topotaxis: A New Mechanism of Directed Cell Migration in Topographic ECM Gradients. Biophys. J. 2018, 114 (6), 1257–1263.

(13) Brown, J. H.; Das, P.; DiVito, M. D.; Ivancic, D.; Poh Tan, L.; Wertheim, J. A. Nanofibrous PLGA Electrospun Scaffolds Modified with Type I Collagen Influence Hepatocyte Function and Support Viability In Vitro. Acta Biomater. 2018, 73, 217–227.

(14) Motta, C. M. M.; Endres, K. J.; Wesdemiotis, C.; Willits, R. K.; Becker, M. L. Enhancing Schwann Cell Migration Using Concentration Gradients of Laminin Derived-Peptides. Biomaterials. 2019, 218, 119335.

(15) Kang, H.; Shih, Y. R. V.; Hwang, Y.; Wen, C.; Rao, V.; Seo, T.; Varghese, S. Mineralized Gelatin Methacrylate-Based Matrices Induce Osteogenic Differentiation of Human Induced Pluripotent Stem Cells. Acta Biomater. 2014, 10 (12), 4961–4970.

(16) Dhaliwal, A.; Maldonado, M.; Han, Z.; Segura, T. Differential Uptake of DNA-Poly(Ethylenimine) Polyplexes in Cells Cultured on Collagen and Fibronectin Surfaces. Acta Biomater. 2010, 6 (9), 3436– 3447.

(17) Levental, K. R.; Yu, H.; Kass, L.; Lakins, J. N.; Egeblad, M.; Erler, J. T.; Fong, S. F. T.; Csiszar, K.; Giaccia, A.; Weninger, W.; et al. Matrix Crosslinking Forces Tumor Progression by Enhancing Integrin Signaling. Cell 2009, 139 (5), 891–906.

(11)

10

Small Molecule Promotes Cartilage Extracellular Matrix Generation and Inhibits Osteoarthritis Development. Nat. Commun. 2019, 10 (1).

(19) Rico, P.; Mnatsakanyan, H.; Dalby, M. J.; Salmerón-Sánchez, M. Material-Driven Fibronectin Assembly Promotes Maintenance of Mesenchymal Stem Cell Phenotypes. Adv. Funct. Mater. 2016, 26 (36), 6563–6573.

(20) Panciera, T.; Citron, A.; Di Biagio, D.; Battilana, G.; Gandin, A.; Giulitti, S.; Forcato, M.; Bicciato, S.; Panzetta, V.; Fusco, S.; et al. Reprogramming Normal Cells into Tumour Precursors Requires ECM Stiffness and Oncogene-Mediated Changes of Cell Mechanical Properties. Nat. Mater. 2020, 19 (July).

(21) Li, B.; Ma, Y.; Wang, S.; Moran, P. M. Influence of Carboxyl Group Density on Neuron Cell Attachment and Differentiation Behavior: Gradient-Guided Neurite Outgrowth. Biomaterials 2005, 26 (24), 4956–4963.

(22) 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.

(23) Shen, H.; Lin, H.; Sun, A. X.; Song, S.; Wang, B.; Yang, Y.; Dai, J.; Tuan, R. S. Acceleration of Chondrogenic Differentiation of Human Mesenchymal Stem Cells by Sustained Growth Factor Release in 3D Graphene Oxide Incorporated Hydrogels. Acta Biomater. 2020, 105, 44–55.

(24) Helfand, B. T.; Mendez, M. G.; Pugh, J.; Delsert, C.; Goldman, R. D. Maintaining the Shape of Nerve Cells. Mol. Biol. Cell 2003, 14 (December), 5069–5081.

(25) Zhou, M.; Lozano, N.; Wychowaniec, J. K.; Hodgkinson, T.; Richardson, S. M.; Kostarelos, K.; Hoyland, J. A. Graphene Oxide: A Growth Factor Delivery Carrier to Enhance Chondrogenic Differentiation of Human Mesenchymal Stem Cells in 3D Hydrogels. Acta Biomater. 2019, 96, 271– 280.

(26) Du, J.; Zhen, G.; Chen, H.; Zhang, S.; Qing, L.; Yang, X.; Lee, G.; Mao, H. Q.; Jia, X. Optimal Electrical Stimulation Boosts Stem Cell Therapy in Nerve Regeneration. Biomaterials 2018, 181, 347– 359.

(27) Abandansari, H. S.; Ghanian, M. H.; Varzideh, F.; Mahmoudi, E.; Rajabi, S.; Taheri, P.; Nabid, M. R.; Baharvand, H. In Situ Formation of Interpenetrating Polymer Network Using Sequential Thermal and Click Crosslinking for Enhanced Retention of Transplanted Cells. Biomaterials 2018, 170, 12–25.

(28) Dorgau, B.; Felemban, M.; Hilgen, G.; Kiening, M.; Zerti, D.; Hunt, N. C.; Doherty, M.; Whitfield, P.; Hallam, D.; White, K.; et al. Decellularised Extracellular Matrix-Derived Peptides from Neural Retina and Retinal Pigment Epithelium Enhance the Expression of Synaptic Markers and Light Responsiveness of Human Pluripotent Stem Cell Derived Retinal Organoids. Biomaterials 2019, 199 (October 2018), 63–75.

(29) Yip, A. K.; Nguyen, A. T.; Rizwan, M.; Wong, S. T.; Chiam, K. H.; Yim, E. K. F. Anisotropic Traction Stresses and Focal Adhesion Polarization Mediates Topography-Induced Cell Elongation. Biomaterials 2018, 181 (July), 103–112.

(30) Campbell, J. J.; Husmann, A.; Hume, R. D.; Watson, C. J.; Cameron, R. E. Development of Three-Dimensional Collagen Scaffolds with Controlled Architecture for Cell Migration Studies Using Breast Cancer Cell Lines. Biomaterials 2017, 114, 34–43.

(31) Abagnale, G.; Steger, M.; Nguyen, V. H.; Hersch, N.; Sechi, A.; Joussen, S.; Denecke, B.; Merkel, R.; Hoffmann, B.; Dreser, A.; et al. Surface Topography Enhances Differentiation of Mesenchymal Stem Cells towards Osteogenic and Adipogenic Lineages. Biomaterials 2015, 61, 316–326.

(32) Dalby, M. J.; Gadegaard, N.; Tare, R.; Andar, A.; Riehle, M. O.; Herzyk, P.; Wilkinson, C. D. W.; Oreffo, R. O. C. The Control of Human Mesenchymal Cell Differentiation Using Nanoscale Symmetry and Disorder. Nat. Mater. 2007, 6 (12), 997–1003.

(12)

11

(33) Cutiongco, M. F. A.; Goh, S. H.; Aid-Launais, R.; Le Visage, C.; Low, H. Y.; Yim, E. K. F. Planar and Tubular Patterning of Micro and Nano-Topographies on Poly(Vinyl Alcohol) Hydrogel for Improved Endothelial Cell Responses. Biomaterials 2016, 84, 184–195.

(34) Ray, A.; Lee, O.; Win, Z.; Edwards, R. M.; Alford, P. W.; Kim, D. H.; Provenzano, P. P. Anisotropic Forces from Spatially Constrained Focal Adhesions Mediate Contact Guidance Directed Cell Migration. Nat. Commun. 2017, 8, 1–17.

(35) Seo, C. H.; Furukawa, K.; Montagne, K.; Jeong, H.; Ushida, T. The Effect of Substrate Microtopography on Focal Adhesion Maturation and Actin Organization via the RhoA/ROCK Pathway. Biomaterials 2011, 32 (36), 9568–9575.

(36) 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.

(37) Roca-Cusachs, P.; Iskratsch, T.; Sheetz, M. P. Finding the Weakest Link-Exploring Integrin-Mediated Mechanical Molecular Pathways. J. Cell Sci. 2012, 125 (13), 3025–3038.

(38) Domingues, R. M. A.; Chiera, S.; Gershovich, P.; Motta, A.; Reis, R. L.; Gomes, M. E. Enhancing the Biomechanical Performance of Anisotropic Nanofibrous Scaffolds in Tendon Tissue Engineering: Reinforcement with Cellulose Nanocrystals. Adv. Healthc. Mater. 2016, 5 (11), 1364– 1375.

(39) Zhang, Z.; Jørgensen, M. L.; Wang, Z.; Amagat, J.; Wang, Y.; Li, Q.; Dong, M.; Chen, M. 3D Anisotropic Photocatalytic Architectures as Bioactive Nerve Guidance Conduits for Peripheral Neural Regeneration. Biomaterials 2020, 253 (April), 120108.

(40) Wilbur, J. L.; Kumar, A.; Kim, E.; Whitesides, G. M. Microfabrication by Microcontact Printing of Self‐assembled Monolayers. Adv. Mater. 1994, 6 (7–8), 600–604.

(41) Yao, Y.; Zhang, L.; Leydecker, T.; Samorì, P. Direct Photolithography on Molecular Crystals for High Performance Organic Optoelectronic Devices. J. Am. Chem. Soc. 2018, 140 (22), 6984–6990. (42) Cells, L. Hybrid Bio / Arti ® Cial Microdevices : Three-Dimensional Photopatterning of Hydrogels

Containing Living Cells. Biomed. Microdevices 2002, 257–266.

(43) Xue, J.; Wu, T.; Dai, Y.; Xia, Y. Electrospinning and Electrospun Nanofibers: Methods, Materials, and Applications. Chem. Rev. 2019, 119 (8), 5298–5415.

(44) Wang, J.; Le-The, H.; Wang, Z.; Li, H.; Jin, M.; Van Den Berg, A.; Zhou, G.; Segerink, L. I.; Shui, L.; Eijkel, J. C. T. Microfluidics Assisted Fabrication of Three-Tier Hierarchical Microparticles for Constructing Bioinspired Surfaces. ACS Nano 2019, 13 (3), 3638–3648.

(45) 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.

(46) Deng, Y.; Ren, J.; Chen, G.; Li, G.; Wu, X.; Wang, G.; Gu, G.; Li, J. Injectable in Situ Cross-Linking Chitosan-Hyaluronic Acid Based Hydrogels for Abdominal Tissue Regeneration. Sci. Rep. 2017, 7 (1), 1–13.

(47) Bettinger, C. J.; Langer, R.; Borenstein, J. T. Minireviews Engineering Substrate Topography at the Micro- and Nanoscale to Control Cell Function. 2009, 5406–5415.

(48) Boyan, B. D.; Lotz, E. M.; Schwartz, Z. Roughness and Hydrophilicity as Osteogenic Biomimetic Surface Properties. Tissue Eng. - Part A 2017, 23 (23–24), 1479–1489.

(49) Jakus, A. E.; Geisendorfer, N. R.; Lewis, P. L.; Shah, R. N. 3D-Printing Porosity: A New Approach to Creating Elevated Porosity Materials and Structures. Acta Biomater. 2018, 72, 94–109.

(50) Sonam, S.; Sathe, S. R.; Yim, E. K. F.; Sheetz, M. P.; Lim, C. T. Cell Contractility Arising from Topography and Shear Flow Determines Human Mesenchymal Stem Cell Fate. Sci. Rep. 2016, 6 (October 2015), 1–12.

(13)

12

(51) Zhang, S.; Ma, B.; Liu, F.; Duan, J.; Wang, S.; Qiu, J.; Li, D.; Sang, Y.; Liu, C.; Liu, D.; et al. Polylactic Acid Nanopillar Array-Driven Osteogenic Differentiation of Human Adipose-Derived Stem Cells Determined by Pillar Diameter. Nano Lett. 2018, 18 (4), 2243–2253.

(52) Ahmad, T.; Lee, J.; Shin, Y. M.; Shin, H. J.; Madhurakat Perikamana, S. K.; Park, S. H.; Kim, S. W.; Shin, H. Hybrid-Spheroids Incorporating ECM like Engineered Fragmented Fibers Potentiate Stem Cell Function by Improved Cell/Cell and Cell/ECM Interactions. Acta Biomater. 2017, 64, 161–175. (53) 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.

(54) 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 (9), 2638–2652.

(55) Nicholas Dias, Yung Peng, R. K. Anisotropic Materials for Skeletal Muscle Tissue Engineering. Physiol. Behav. 2017, 176 (3), 139–148.

(56) Kim, D.-H.; Lipke, E. A.; Kim, P.; Cheong, R.; Thompson, S.; Delannoy, M.; Suh, K.-Y.; Tung, L.; Levchenko, A. Nanoscale Cues Regulate the Structure and Function of Macroscopic Cardiac Tissue Constructs. Proc. Natl. Acad. Sci. 2009, 107 (2), 565–570.

(57) 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. Advanced Healthcare Materials. 2017.

(58) Wen, Y.; Xun, S.; Haoye, M.; Baichuan, S.; Peng, C.; Xuejian, L.; Kaihong, Z.; Xuan, Y.; Jiang, P.; Shibi, L. 3D Printed Porous Ceramic Scaffolds for Bone Tissue Engineering: A Review. Biomater. Sci. 2017, 5 (9), 1690–1698.

(59) 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. (60) Sekhon, U. D. S.; Sen Gupta, A. Platelets and Platelet-Inspired Biomaterials Technologies in Wound

Healing Applications. ACS Biomater. Sci. Eng. 2018, 4 (4), 1176–1192.

(61) Tavakoli, S.; Klar, A. S. Advanced Hydrogels as Wound Dressings. Biomolecules 2020, 10 (8), 1–20. (62) 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.

(63) 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. (64) Won, Y.-W.; Patel, A. N.; Bull, D. A. Cell Surface Engineering to Enhance Mesenchymal Stem Cell

Migration toward an SDF-1 Gradient. Biomaterials 2014, 35 (21), 5627–5635.

(65) 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.

(66) Deng, J.; Zhao, C.; Spatz, J. P.; Wei, Q. Nanopatterned Adhesive, Stretchable Hydrogel to Control Ligand Spacing and Regulate Cell Spreading and Migration. ACS Nano 2017, 11 (8), 8282–8291. (67) Sheets, K.; Wunsch, S.; Ng, C.; Nain, A. S. Shape-Dependent Cell Migration and Focal Adhesion

Organization on Suspended and Aligned Nanofiber Scaffolds. Acta Biomater. 2013, 9 (7), 7169–7177. (68) Kim, D. H.; Han, K.; Gupta, K.; Kwon, K. W.; Suh, K. Y.; Levchenko, A. Mechanosensitivity of Fibroblast Cell Shape and Movement to Anisotropic Substratum Topography Gradients. Biomaterials 2009, 30 (29), 5433–5444.

(69) Zhang, Q.; Dong, H.; Li, Y.; Zhu, Y.; Zeng, L.; Gao, H.; Yuan, B.; Chen, X.; Mao, C. Microgrooved Polymer Substrates Promote Collective Cell Migration to Accelerate Fracture Healing in an in Vitro

(14)

13

Model. ACS Appl. Mater. Interfaces 2015, 7 (41), 23336–23345.

(70) 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.

(71) Ellis, R. J. Macromolecular Crowding: Obvious but Underappreciated. Trends Biochem. Sci. 2001, 26 (10), 597–604.

(72) 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. (73) Chapanian, R.; Kwan, D. H.; Constantinescu, I.; Shaikh, F. A.; Rossi, N. A. A.; Withers, S. G.;

Kizhakkedathu, J. N. Enhancement of Biological Reactions on Cell Surfaces via Macromolecular Crowding. Nat. Commun. 2014, 5.

(74) Lindner, R. A.; Ralston, G. B. Macromolecular Crowding: Effects on Actin Polymerisation. Biophysical Chemistry. 1997, pp 57–66.

(75) Van Den Berg, B.; Wain, R.; Dobson, C. M.; Ellis, R. J. Macromolecular Crowding Perturbs Protein Refolding Kinetics: Implications for Folding inside the Cell. EMBO J. 2000, 19 (15), 3870–3875. (76) Wu, T.; Xue, J.; Xia, Y. Engraving the Surface of Electrospun Microfibers with Nanoscale Grooves

Promotes the Outgrowth of Neurites and the Migration of Schwann Cells. Angew. Chemie - Int. Ed. 2020.

(77) Ge, X.; Luo, D.; Xu, J. Cell-Free Protein Expression under Macromolecular Crowding Conditions. PLoS One 2011, 6 (12).

(78) Chen, C.; Loe, F.; Blocki, A.; Peng, Y.; Raghunath, M. Applying Macromolecular Crowding to Enhance Extracellular Matrix Deposition and Its Remodeling in Vitro for Tissue Engineering and Cell-Based Therapies. Adv. Drug Deliv. Rev. 2011, 63 (4), 277–290.

(79) Hata, Y.; Sawada, T.; Serizawa, T. Macromolecular Crowding for Materials-Directed Controlled Self-Assembly. J. Mater. Chem. B 2018, 6 (40), 6344–6359.

(80) Burg, M. B. Macromolecular Crowding as a Cell Volume Sensor. Cell. Physiol. Biochem. 2000, 10 (5– 6), 251–256.

(81) Al-Habori, M. Macromolecular Crowding and Its Role as Intracellular Signalling of Cell Volume Regulation. Int. J. Biochem. Cell Biol. 2001, 33 (9), 844–864.

(82) 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.

(83) 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.

(84) Xiang, Y.; Oo, N. N. L.; Lee, J. P.; Li, Z.; Loh, X. J. Recent Development of Synthetic Nonviral Systems for Sustained Gene Delivery. Drug Discov. Today 2017, 22 (9), 1318–1335.

(85) 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.

(86) 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.

(87) 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.

(88) Han, H.; Yang, J.; Chen, W.; Li, Q.; Yang, Y.; Li, Q. A Comprehensive Review on Histone-Mediated Transfection for Gene Therapy. Biotechnol. Adv. 2019, 37 (1), 132–144.

(15)

14

Electropermeabilization: Mechanisms and Models. Annu. Rev. Biophys. 2019, 48, 63–91.

(90) Zhao, X.; Meng, Z.; Wang, Y.; Chen, W.; Sun, C.; Cui, B.; Cui, J.; Yu, M.; Zeng, Z.; Guo, S.; et al. Pollen Magnetofection for Genetic Modification with Magnetic Nanoparticles as Gene Carriers. Nat. Plants 2017, 3 (12), 956–964.

(91) 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.

(92) Sun, X.; Li, M.; Yang, Y.; Jia, H.; Liu, W. Carrier-Free Nanodrug-Based Virus-Surface-Mimicking Nanosystems for Efficient Drug/Gene Co-Delivery. Biomater. Sci. 2018, 6 (12), 3300–3308.

(93) Xiao, X.; Guo, P.; Shiota, C.; Zhang, T.; Coudriet, G. M.; Fischbach, S.; Prasadan, K.; Fusco, J.; Ramachandran, S.; Witkowski, P.; et al. Endogenous Reprogramming of Alpha Cells into Beta Cells, Induced by Viral Gene Therapy, Reverses Autoimmune Diabetes. Cell Stem Cell 2018, 22 (1), 78-90.e4.

(94) Schulze, J.; Kuhn, S.; Hendrikx, S.; Schulz-Siegmund, M.; Polte, T.; Aigner, A. Spray-Dried Nanoparticle-in-Microparticle Delivery Systems (NiMDS) for Gene Delivery, Comprising Polyethylenimine (PEI)-Based Nanoparticles in a Poly(Vinyl Alcohol) Matrix. Small 2018, 14 (12), 1–8.

(95) Chang, R.; Yan, Q.; Kingshott, P.; Tsai, W. B.; Wang, P. Y. Harnessing the Perinuclear Actin Cap (PnAC) to Influence Nanocarrier Trafficking and Gene Transfection Efficiency in Skeletal Myoblasts Using Nanopillars. Acta Biomater. 2020, 111, 221–231.

(96) Fang, H.; Guo, Z.; Lin, L.; Chen, J.; Sun, P.; Wu, J.; Xu, C.; Tian, H.; Chen, X. Molecular Strings Significantly Improved the Gene Transfection Efficiency of Polycations. J. Am. Chem. Soc. 2018, 140 (38), 11992–12000.

(97) Singh, R. K.; Knowles, J. C.; Kim, H. W. Advances in Nanoparticle Development for Improved Therapeutics Delivery: Nanoscale Topographical Aspect. J. Tissue Eng. 2019, 10.

(98) Stephen, Z. R.; Dayringer, C. J.; Lim, J. J.; Revia, R. A.; Halbert, M. V.; Jeon, M.; Bakthavatsalam, A.; Ellenbogen, R. G.; Zhang, M. Approach to Rapid Synthesis and Functionalization of Iron Oxide Nanoparticles for High Gene Transfection. ACS Appl. Mater. Interfaces 2016, 8 (10), 6320–6328. (99) Chen, W.; Li, H.; Liu, Z.; Yuan, W. Lipopolyplex for Therapeutic Gene Delivery and Its Application

for the Treatment of Parkinson’s Disease. Front. Aging Neurosci. 2016, 8 (APR), 1–10.

(100) Zhao, B. wei; Zhou, Z.; Shen, Y. Effects of Chirality on Gene Delivery Efficiency of Polylysine. Chinese J. Polym. Sci. (English Ed. 2016, 34 (1), 94–103.

(101) Song, H.; Yu, M.; Lu, Y.; Gu, Z.; Yang, Y.; Zhang, M.; Fu, J.; Yu, C. Plasmid DNA Delivery: Nanotopography Matters. J. Am. Chem. Soc. 2017, 139 (50), 18247–18254.

(102) Shalek, A. K.; Robinson, J. T.; Karp, E. S.; Lee, J. S.; Ahn, D. R.; Yoon, M. H.; Sutton, A.; Jorgolli, M.; Gertner, R. S.; Gujral, T. S.; et al. Vertical Silicon Nanowires as a Universal Platform for Delivering Biomolecules into Living Cells. Proc. Natl. Acad. Sci. U. S. A. 2010, 107 (5), 1870–1875. (103) Shalek, A. K.; Gaublomme, J. T.; Wang, L.; Yosef, N.; Chevrier, N.; Andersen, M. S.; Robinson, J.

T.; Pochet, N.; Neuberg, D.; Gertner, R. S.; et al. Nanowire-Mediated Delivery Enables Functional Interrogation of Primary Immune Cells: Application to the Analysis of Chronic Lymphocytic Leukemia. Nano Lett. 2012, 12 (12), 6498–6504.

(104) Wang, K.; Bruce, A.; Mezan, R.; Kadiyala, A.; Wang, L.; Dawson, J.; Rojanasakul, Y.; Yang, Y. Nanotopographical Modulation of Cell Function through Nuclear Deformation. ACS Appl. Mater. Interfaces 2016, 8 (8), 5082–5092.

(105) Ryoo, S. R.; Kim, Y. K.; Kim, M. H.; Min, D. H. Behaviors of NIH-3T3 Fibroblasts on Graphene/Carbon Nanotubes: Proliferation, Focal Adhesion, and Gene Transfection Studies. ACS

(16)

15

(106) 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. (107) Teo, B. K. K.; Goh, S. H.; Kustandi, T. S.; Loh, W. W.; Low, H. Y.; Yim, E. K. F. The Effect of

Micro and Nanotopography on Endocytosis in Drug and Gene Delivery Systems. Biomaterials 2011, 32 (36), 9866–9875.

(108) Harding, F. J.; Surdo, S.; Delalat, B.; Cozzi, C.; Elnathan, R.; Gronthos, S.; Voelcker, N. H.; Barillaro, G. Ordered Silicon Pillar Arrays Prepared by Electrochemical Micromachining: Substrates for High-Efficiency Cell Transfection. ACS Appl. Mater. Interfaces 2016, 8 (43), 29197–29202.

(17)

Referenties

GERELATEERDE DOCUMENTEN

Chapter 4: Topography-mediated myotube and endothelial alignment, differentiation, and extracellular matrix organization for skeletal muscle engineering ...83.

The aim of this thesis is to identify which topography is linked to the alignment, proliferation, and differentiation of human myoblasts in conjunction with endothelial

In our directional topography gradient, topographies larger than those in section 1 augmented cell attachment, proliferation, and differentiation because the nuclei were pushed to

Collective migration of endothelial cell aggregates affected by directional topography Chapter 3 showed how endothelial cells aggregate, and then migrate around the substrate;

In chapter 2 we investigated the optimum alignment of human myoblasts (muscle stem cells) and hypothesized that myoblasts have a preferred directional topography

Verder hebben we laten zien dat topografie en topografie-gestructureerde myoblasten – myotubes – capillairvorming kunnen sturen maar dat hiervoor hulpcellen voor nodig

All great moments that made my mind go away from the stress of a PhD and gave me a sense of family in the Netherlands and energies to continue forward.. Marloes: I always looked up

Topography enhances alignment of cells only above a critical minimal size threshold (this thesis). Myotubes support attachment and alignment of endothelial cells but do not