AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (696.7 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Potential Role of Hydrogels in Stem Cell Culture and Hepatocyte Differentiation

Ying LuoYingtang Gao( )
Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Institute of Hepatobiliary Disease, Nankai University Affiliated Third Center Hospital, Tianjin 300071, China
Show Author Information

Graphical Abstract

Abstract

Stem cells are cells with self-renewal, reproductive activity, and multiple differentiation potential. The native cell-extracellular matrix (ECM) is a hydrated network of proteins and polysaccharides that provide the surrounding environment to maintain stem cells. Hydrogels possess broadly tunable biofunctions and adequate mechanical strength, as well as ECM-like conditions for better cellular activity. Hydrogels are considered one of the most utilized and relevant options in terms of mimicking the native ECM .This review article summarizes the recent advances in the field of using hydrogels as a stem cell niche to regulate cell fate from design strategies. Furthermore, we also review how hydrogels regulate stem cell fate and differentiate into hepatocytes. Looking forward, we envision that with the adoption of an increasing number of interdisciplinary methods, it is reasonable to combine multiple strategies to prepare hydrogels with enhanced performance. Due to the special arrangement or interaction between multiple scales and multiple components, a new combination of hydrogels has emerged, which provides opportunities for further innovation of hydrogels in regulating cell fate.

References

[1]

P. Gerdes, S.M. Lim, A.D. Ewing, et al. Retrotransposon instability dominates the acquired mutation landscape of mouse induced pluripotent stem cells. Nature Communication, 2022, 13: 7470. https://doi.org/10.1038/s41467-022-35180-x

[2]

M. Segel, B. Neumann, M.F.E. Hill, et al. Niche stiffness underlies the ageing of central nervous system progenitor cells. Nature, 2019, 573: 130−134. https://doi.org/10.1038/s41586-019-1484-9

[3]

G. Lutzweiler, J. Barthes, G. Koenig, et al. Modulation of cellular colonization of porous polyurethane scaffolds via the control of pore interconnection size and nanoscale surface modifications. ACS Applied Materials &Interfaces, 2019, 11: 19819−19829. https://doi.org/10.1021/acsami.9b04625

[4]

M. Alvarez-Paino, M.H. Amer, A. Nasir, et al. Polymer microparticles with defined surface chemistry and topography mediate the formation of stem cell aggregates and cardiomyocyte function. ACS Applied Materials &Interfaces, 2019, 11: 34560−34574. https://doi.org/10.1021/acsami.9b04769

[5]

H.W. Lv, H.P. Wang, Z.J. Zhang, et al. Biomaterial stiffness determines stem cell fate. Life Sciences, 2017, 178: 42−48. https://doi.org/10.1016/j.lfs.2017.04.014

[6]

A. Kumar, J.K. Placone, A.J. Engler. Understanding the extracellular forces that determine cell fate and maintenance. Development, 2017, 144: 4261−4270. https://doi.org/10.1242/dev.158469

[7]
D.K. Patel, K.T. Lim. Biomimetic Polymer-based engineered scaffolds for improved stem cell function. Materials (Basel), 2019, 12.
[8]

W. Chen, C. Wang, W. Liu, et al. A Matrix-metalloproteinase-responsive hydrogel system for modulating the immune microenvironment in myocardial infarction. Advanced Materials, 2023, 35: e2209041. https://doi.org/10.1002/adma.202209041

[9]

A. Jaeschke, N.R. Harvey, M. Tsurkan, et al. Techniques for RNA extraction from cells cultured in starPEG-heparin hydrogels. Open Biology, 2021, 11: 200388. https://doi.org/10.1098/rsob.200388

[10]
G. Liu, R. Wu, B. Yang, et al. A cocktail of growth factors released from heparin hyaluronic-acid hydrogel promotes the myogenic potential of human urine-derived stem cells in vivo. Acta Biomaterialia, 2020, 107: 50–64.
[11]

C.M. Madl, I.A. Flaig, C.A. Holbrook, et al. Biophysical matrix cues from the regenerating niche direct muscle stem cell fate in engineered microenvironments. Biomaterials, 2021, 275: 120973. https://doi.org/10.1016/j.biomaterials.2021.120973

[12]

N. Hu, Z. Cai, X. Jiang, et al. Hypoxia-pretreated ADSC-derived exosome-embedded hydrogels promote angiogenesis and accelerate diabetic wound healing. Acta Biomaterialia, 2023, 157: 175−186. https://doi.org/10.1016/j.actbio.2022.11.057

[13]

H. Shen, H. Lin, A.X. Sun, et al. Acceleration of chondrogenic differentiation of human mesenchymal stem cells by sustained growth factor release in 3D graphene oxide incorporated hydrogels. Acta Biomaterialia, 2020, 105: 44−55. https://doi.org/10.1016/j.actbio.2020.01.048

[14]

G. Garcia-Llorens, T. Martinez-Sena, E. Pareja, et al. A robust reprogramming strategy for generating hepatocyte-like cells usable in pharmaco-toxicological studies. Stem Cell Research Therapy, 2023, 14: 94. https://doi.org/10.1186/s13287-023-03311-w

[15]

M. Hussein, M. Pasqua, U. Pereira, et al. Microencapsulated hepatocytes differentiated from human induced pluripotent stem cells: optimizing 3D culture for tissue engineering applications. Cells, 2023, 12(6): 865. https://doi.org/10.3390/cells12060865

[16]

Z. Yang, R. Huang, B. Zheng, et al. Highly stretchable, adhesive, biocompatible, and antibacterial hydrogel dressings for wound healing. Advanced Science, 2021, 8: 2003627. https://doi.org/10.1002/advs.202003627

[17]

Mohiuddin, B.T. O'Donnell, J.N. Poche, et al. Human adipose-derived hydrogel characterization based on in vitro ASC biocompatibility and differentiation. Stem Cells International, 2019, 2019: 9276398. https://doi.org/10.1155/2019/9276398

[18]

J. Wang, R. Chu, N. Ni, et al. The effect of Matrigel as scaffold material for neural stem cell transplantation for treating spinal cord injury. Scientific Reports, 2020, 10: 2576. https://doi.org/10.1038/s41598-020-59148-3

[19]

S.H. Mao, C.H. Chen, C.T. Chen. Osteogenic potential of induced pluripotent stem cells from human adipose-derived stem cells. Stem Cell Research &Therapy, 2019, 10: 303. https://doi.org/10.1186/s13287-019-1402-y

[20]

K.C. Murphy, J. Whitehead, D. Zhou, et al. Engineering fibrin hydrogels to promote the wound healing potential of mesenchymal stem cell spheroids. Acta Biomaterialia, 2017, 64: 176−186. https://doi.org/10.1016/j.actbio.2017.10.007

[21]

M. Boido, M. Ghibaudi, P. Gentile, et al. Chitosan-based hydrogel to support the paracrine activity of mesenchymal stem cells in spinal cord injury treatment. Scientific Reports, 2019, 9: 6402. https://doi.org/10.1038/s41598-019-42848-w

[22]

M.E. Klontzas, S. Reakasame, R. Silva, et al. Oxidized alginate hydrogels with the GHK peptide enhance cord blood mesenchymal stem cell osteogenesis: A paradigm for metabolomics-based evaluation of biomaterial design. Acta Biomaterialia, 2019, 88: 224−240. https://doi.org/10.1016/j.actbio.2019.02.017

[23]

W. Kim, G. Kim. Collagen/bioceramic-based composite bioink to fabricate a porous 3D hASCs-laden structure for bone tissue regeneration. Biofabrication, 2019, 12: 015007. https://doi.org/10.1088/1758-5090/ab436d

[24]

F. Gao, J. Li, L. Wang, et al. Dual-enzymatically crosslinked hyaluronic acid hydrogel as a long-time 3D stem cell culture system. Biomedical Materials, 2020, 15: 045013. https://doi.org/10.1088/1748-605X/ab712e

[25]

L.H. Chen, T.C. Sung, H.H. Lee, et al. Xeno-free and feeder-free culture and differentiation of human embryonic stem cells on recombinant vitronectin-grafted hydrogels. Biomaterials Science, 2019, 7: 4345−4362. https://doi.org/10.1039/c9bm00418a

[26]

R.H. Dosh, N. Jordan-Mahy, C. Sammon, et al. Use of l-pNIPAM hydrogel as a 3D-scaffold for intestinal crypts and stem cell tissue engineering. Biomaterials Science, 2019, 7: 4310−4324. https://doi.org/10.1039/c9bm00541b

[27]

R.A. Dilla, Y. Xu, Z.K. Zander, et al. Mechanically tunable, human mesenchymal stem cell viable poly(ethylene glycol)-oxime hydrogels with invariant precursor composition, concentration, and stoichiometry. Materials Today Chemistry, 2019, 11: 244−252. https://doi.org/10.1016/j.mtchem.2018.11.003

[28]

Y.-M. Chen, L.-H. Chen, M.-P. Li, et al. Xeno-free culture of human pluripotent stem cells on oligopeptide-grafted hydrogels with various molecular designs. Scientific Reports, 2017, 7: 45146. https://doi.org/10.1038/srep45146

[29]

C. McKee, M. Perez-Cruet, F. Chavez, et al. Simplified three-dimensional culture system for long-term expansion of embryonic stem cells. World Journal of Stem Cells, 2015, 7: 1064−1077. https://doi.org/10.4252/wjsc.v7.i7.1064

[30]

A.Mellati, M.V. Kiamahalleh, S.H. Madani, et al. Poly(N-isopropylacrylamide) hydrogel/chitosan scaffold hybrid for three-dimensional stem cell culture and cartilage tissue engineering. Journal of Biomedical Materials Research, 2016, 104: 2764−2774. https://doi.org/10.1002/jbm.a.35810

[31]

L. Cai, J. Li, S. Quan, et al. Dextran-based hydrogel with enhanced mechanical performance via covalent and non-covalent cross-linking units carrying adipose-derived stem cells toward vascularized bone tissue engineering. Journal of Biomedical Materials Research Part A, 2019, 107: 1120−1131. https://doi.org/10.1002/jbm.a.36580

[32]

J. Tan, Y. Luo, Y. Guo, et al. Development of alginate-based hydrogels: Crosslinking strategies and biomedical applications. International Journal of Biological Macromolecules, 2023, 239: 124275. https://doi.org/10.1016/j.ijbiomac.2023.124275

[33]

Z. Zhang, N. Abidi, L. Lucia, et al. Cellulose/nanocellulose superabsorbent hydrogels as a sustainable platform for materials applications: A mini-review and perspective. Carbohydrate Polymers, 2023, 299: 120140. https://doi.org/10.1016/j.carbpol.2022.120140

[34]

J.R. Clegg, N.A. Peppas. Design of synthetic hydrogel compositions for noncovalent protein recognition. ACS Applied Materials &Interfaces, 2023, 15(44): 50586−50597. https://doi.org/10.1021/acsami.2c20857

[35]

T. Hu, A.C.Y. Lo. Collagen–alginate composite hydrogel: application in tissue engineering and biomedical sciences. Polymers, 2021, 13(11): 1852. https://doi.org/10.3390/polym13111852

[36]

R. Naranjo-Alcazar, S. Bendix, T. Groth, et al. Research progress in enzymatically cross-linked hydrogels as injectable systems for bioprinting and tissue engineering. Gels, 2023, 9(3): 230. https://doi.org/10.3390/gels9030230

[37]

B. Lv, L. Lu, L. Hu, et al. Recent advances in GelMA hydrogel transplantation for musculoskeletal disorders and related disease treatment. Theranostics, 2023, 13(6): 2015−2039. https://doi.org/10.7150/thno.80615

[38]

F. Ali, I. Khan, J. Chen, et al. Emerging fabrication strategies of hydrogels and its applications. Gels, 2022, 8(4): 205. https://doi.org/10.3390/gels8040205

[39]

O. Hasturk, J.A. Smiley, M. Arnett, et al. Cytoprotection of human progenitor and stem cells through encapsulation in alginate templated, dual crosslinked silk and silk-gelatin composite hydrogel microbeads. Advanced Healthcare Materials, 2022, 11: e2200293. https://doi.org/10.1002/adhm.202200293

[40]

A. Paul, M. Stührenberg, S. Chen, et al. Micro- and nano-patterned elastin-like polypeptide hydrogels for stem cell culture. Soft Matter, 2017, 13: 5665−5675. https://doi.org/10.1039/c7sm00487g

[41]

E. Kapyla, S.M. Delgado, A.M. Kasko. Shape-changing photodegradable hydrogels for dynamic 3D cell culture. ACS Applied Materials &Interfaces, 2016, 8: 17885−17893. https://doi.org/10.1021/acsami.6b05527

[42]

S.K. Schmitt, A.W. Xie, R.M. Ghassemi, et al. Polyethylene glycol coatings on plastic substrates for chemically defined stem cell culture. Advanced Healthcare Materials, 2015, 4: 1555−1564. https://doi.org/10.1002/adhm.201500191

[43]

Y. Zhang, N. Ding, T. Zhang, et al. A tetra-PEG hydrogel based aspirin sustained release system exerts beneficial effects on periodontal ligament stem cells mediated bone regeneration. Frontiers in chemistry, 2019, 7: 682. https://doi.org/10.3389/fchem.2019.00682

[44]

G. Yang, Z. Xiao, X. Ren, et al. Enzymatically crosslinked gelatin hydrogel promotes the proliferation of adipose tissue-derived stromal cells. PeerJ, 2016, 4: e2497. https://doi.org/10.7717/peerj.2497

[45]

K. Gwon, E. Kim, G. Tae. Heparin-hyaluronic acid hydrogel in support of cellular activities of 3D encapsulated adipose derived stem cells. Acta Biomaterialia, 2017, 49: 284−295. https://doi.org/10.1016/j.actbio.2016.12.001

[46]

O. Hasturk, K.E. Jordan, J. Choi, et al. Enzymatically crosslinked silk and silk-gelatin hydrogels with tunable gelation kinetics, mechanical properties and bioactivity for cell culture and encapsulation. Biomaterials, 2020, 232: 119720. https://doi.org/10.1016/j.biomaterials.2019.119720

[47]

K. Zhang, L. Song, J. Wang, et al. Strategy for constructing vascularized adipose units in poly(l-glutamic acid) hydrogel porous scaffold through inducing in-situ formation of ASCs spheroids. Acta Biomaterialia, 2017, 51: 246−257. https://doi.org/10.1016/j.actbio.2017.01.043

[48]

N.J. Hogrebe, K.J. Gooch. Direct influence of culture dimensionality on human mesenchymal stem cell differentiation at various matrix stiffnesses using a fibrous self-assembling peptide hydrogel. Journal of Biomedical Materials Research Part A, 2016, 104: 2356−2368. https://doi.org/10.1002/jbm.a.35755

[49]

G. Choe, J. Park, H. Park, et al. Hydrogel biomaterials for stem cell microencapsulation. Polymers, 2018, 10(9): 997. https://doi.org/10.3390/polym10090997

[50]

K. Min, G. Tae. Cellular infiltration in an injectable sulfated cellulose nanocrystal hydrogel and efficient angiogenesis by VEGF loading. Biomaterials Research, 2023, 27: 28. https://doi.org/10.1186/s40824-023-00373-y

[51]

D. Silva, L. Schirmer, T.S. Pinho, et al. Sustained release of human adipose tissue stem cell secretome from star-shaped poly(ethylene glycol) Glycosaminoglycan hydrogels promotes motor improvements after complete transection in spinal cord injury rat model. Advanced Healthcare Materials, 2023, 12(17): e2202803. https://doi.org/10.1002/adhm.202202803

[52]

S. Li, J. Sun, J. Yang, et al. Gelatin methacryloyl (GelMA) loaded with concentrated hypoxic pretreated adipose-derived mesenchymal stem cells (ADSCs) conditioned medium promotes wound healing and vascular regeneration in aged skin. Biomaterials Research, 2023, 27: 11. https://doi.org/10.1186/s40824-023-00352-3

[53]

C. Levinson, M. Lee, L.A. Applegate, et al. An injectable heparin-conjugated hyaluronan scaffold for local delivery of transforming growth factor beta1 promotes successful chondrogenesis. Acta Biomaterialia, 2019, 99: 168−180. https://doi.org/10.1016/j.actbio.2019.09.017

[54]

J. Cui, S. Zhang, S. Cheng, et al. Current and future outlook of loaded components in hydrogel composites for the treatment of chronic diabetic ulcers. Frontiers in Bioengineering and Biotechnology, 2023, 11: 1077490. https://doi.org/10.3389/fbioe.2023.1077490

[55]

P. Ghandforoushan, J. Hanaee, Z. Aghazadeh, et al. Enhancing the function of PLGA-collagen scaffold by incorporating TGF-beta1-loaded PLGA-PEG-PLGA nanoparticles for cartilage tissue engineering using human dental pulp stem cells. Drug Delivery and Translational Research, 2022, 12: 2960−2978. https://doi.org/10.1007/s13346-022-01161-2

[56]

J. Kim, Y.M. Kim, S.C. Song. One-Step Preparation of an injectable hydrogel scaffold system capable of sequential dual-growth factor release to maximize bone regeneration. Advanced Healthcare Materials, 2023, 12: e2202401. https://doi.org/10.1002/adhm.202202401

[57]

B. Divband, M. Aghazadeh, Z.H. Al-Qaim, et al. Bioactive chitosan biguanidine-based injectable hydrogels as a novel BMP-2 and VEGF carrier for osteogenesis of dental pulp stem cells. Carbohydrate Polymers, 2021, 273: 118589. https://doi.org/10.1016/j.carbpol.2021.118589

[58]

P. Pal, Q.C. Nguyen, A.H. Benton, et al. Drug-loaded elastin-like polypeptide-collagen hydrogels with high modulus for bone tissue engineering. Macromolecular Bioscience, 2019, 19: e1900142. https://doi.org/10.1002/mabi.201900142

[59]
D.T. Scadden. The stem-cell niche as an entity of action. Nature, 2006, 441: 1075–1079. https://10.1038/nature04957
[60]

E. Zimran, L. Papa, R. Hoffman. Ex vivo expansion of hematopoietic stem cells: Finally transitioning from the lab to the clinic. Blood Reviews, 2021, 50: 100853. https://doi.org/10.1016/j.blre.2021.100853

[61]
B. Liu, M. Jin, D.A. Wang. In vitro expansion of hematopoietic stem cells in a porous hydrogel-based 3D culture system. Acta Biomaterialia, 2023, 161: 67–79.
[62]

Y. Wang, R. Sugimura. Ex vivo expansion of hematopoietic stem cells. Experimental Cell Research, 2023, 427: 113599. https://doi.org/10.1016/j.yexcr.2023.113599

[63]

M.L. Cuchiara, S. Coşkun, O.A. Banda, et al. Bioactive poly(ethylene glycol) hydrogels to recapitulate the HSC niche and facilitate HSC expansion in culture. Biotechnology and Bioengineering, 2016, 113: 870−881. https://doi.org/10.1002/bit.25848

[64]

T. Maraldi, C. Angeloni, C. Prata, et al. NADPH oxidases: redox regulators of stem cell fate and function. Antioxidants, 2021, 10(6): 973. https://doi.org/10.3390/antiox10060973

[65]

W. Ding, Q. Zhou, Y. Lu, et al. ROS-scavenging hydrogel as protective carrier to regulate stem cells activity and promote osteointegration of 3D printed porous titanium prosthesis in osteoporosis. Frontiers in Bioengineering and Biotechnology, 2023, 11: 1103611. https://doi.org/10.3389/fbioe.2023.1103611

[66]

T. Bai, J. Li, A. Sinclair, et al. Expansion of primitive human hematopoietic stem cells by culture in a zwitterionic hydrogel. Nature medicine, 2019, 25: 1566−1575. https://doi.org/10.1038/s41591-019-0601-5

[67]

S. Higuchi, T.M. Watanabe, K. Kawauchi, et al. Culturing of mouse and human cells on soft substrates promote the expression of stem cell markers. Journal of Bioscience and Bioengineering, 2014, 117: 749−755. https://doi.org/10.1016/j.jbiosc.2013.11.011

[68]

J.S. Choi, B.A.C. Harley. Marrow-inspired matrix cues rapidly affect early fate decisions of hematopoietic stem and progenitor cells. Science advances, 2017, 3: e1600455. https://doi.org/10.1126/sciadv.1600455

[69]

O. Chaudhuri, L. Gu, D. Klumpers, et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nature Materials, 2016, 15: 326−334. https://doi.org/10.1038/nmat4489

[70]

M. Ha, A. Athirasala, A. Tahayeri, et al. Micropatterned hydrogels and cell alignment enhance the odontogenic potential of stem cells from apical papilla in-vitro. Dental Materials, 2020, 36: 88−96. https://doi.org/10.1016/j.dental.2019.10.013

[71]

B.K. Velmurugan, L. Bharathi Priya, P. Poornima, et al. Biomaterial aided differentiation and maturation of induced pluripotent stem cells. Journal of Cellular Physiology, 2019, 234: 8443−8454. https://doi.org/10.1002/jcp.27769

[72]

D. Lü, C. Luo, C. Zhang, et al. Differential regulation of morphology and stemness of mouse embryonic stem cells by substrate stiffness and topography. Biomaterials, 2014, 35: 3945−3955. https://doi.org/10.1016/j.biomaterials.2014.01.066

[73]

P.-Y. Wang, H. Thissen, P. Kingshott. Modulation of human multipotent and pluripotent stem cells using surface nanotopographies and surface-immobilised bioactive signals: A review. Acta Biomaterialia, 2016, 45: 31−59. https://doi.org/10.1016/j.actbio.2016.08.054

[74]

Q. Zhang, Y. Liu, J. Li, et al. Recapitulation of growth factor-enriched microenvironment via BMP receptor activating hydrogel. Bioactive Materials, 2023, 20: 638−650. https://doi.org/10.1016/j.bioactmat.2022.06.012

[75]

C. Black, J.M. Kanczler, M.C. de Andrés, et al. Characterisation and evaluation of the regenerative capacity of Stro-4+ enriched bone marrow mesenchymal stromal cells using bovine extracellular matrix hydrogel and a novel biocompatible melt electro-written medical-grade polycaprolactone scaffold. Biomaterials, 2020, 247: 119998. https://doi.org/10.1016/j.biomaterials.2020.119998

[76]

V.A. Revkova, E.A. Grebenik, V.A. Kalsin, et al. Chitosan-g-oligo (L,L-lactide) copolymer hydrogel potential for neural stem cell differentiation. Tissue Engineering Part A, 2020, 26: 953−963. https://doi.org/10.1089/ten.TEA.2019.0265

[77]

X. Zhou, J. Wang, W. Fang, et al. Genipin cross-linked type II collagen/chondroitin sulfate composite hydrogel-like cell delivery system induces differentiation of adipose-derived stem cells and regenerates degenerated nucleus pulposus. Acta Biomaterialia, 2018, 71: 496−509. https://doi.org/10.1016/j.actbio.2018.03.019

[78]

P. Pal, M.A. Tucci, L.W. Fan, et al. Functionalized collagen/elastin-like polypeptide hydrogels for craniofacial bone regeneration. Advanced Healthcare Materials, 2023, 12: e2202477. https://doi.org/10.1002/adhm.202202477

[79]

C. Pizzolitto, F. Scognamiglio, P. Sacco, et al. Immediate stress dissipation in dual cross-link hydrogels controls osteogenic commitment of mesenchymal stem cells. Carbohydrate Polymers, 2023, 302: 120369. https://doi.org/10.1016/j.carbpol.2022.120369

[80]
A.J. Engler, S. Sen, H.L. Sweeney, et al. Matrix elasticity directs stem cell lineage specification. Cell, 2006, 126: 677–689.
[81]

S. Nam, J. Lee, D.G. Brownfield, et al. Viscoplasticity enables mechanical remodeling of matrix by cells. Biophysical Journal, 2016, 111: 2296−2308. https://doi.org/10.1016/j.bpj.2016.10.002

[82]

Y. Wang, Y. Yang, X. Wang, et al. The varied influences of cell adhesion and spreading on gene transfection of mesenchymal stem cells on a micropatterned substrate. Acta Biomaterialia, 2021, 125: 100−111. https://doi.org/10.1016/j.actbio.2021.01.042

[83]

Y. Hwang, M. Goh, M. Kim, et al. Injectable and detachable heparin-based hydrogel micropatches for hepatic differentiation of hADSCs and their liver targeted delivery. Biomaterials, 2018, 165: 94−104. https://doi.org/10.1016/j.biomaterials.2018.03.001

[84]

S. Zijl, A.S. Vasilevich, P. Viswanathan, et al. Micro-scaled topographies direct differentiation of human epidermal stem cells. Acta Biomaterialia, 2019, 84: 133−145. https://doi.org/10.1016/j.actbio.2018.12.003

[85]

L. Otsuki, A.H. Brand. Cell cycle heterogeneity directs the timing of neural stem cell activation from quiescence. Science, 2018, 360: 99−102. https://doi.org/10.1126/science.aan8795

[86]

M. Quarta, J.O. Brett, R. DiMarco, et al. An artificial niche preserves the quiescence of muscle stem cells and enhances their therapeutic efficacy. Nature Biotechnology, 2016, 34: 752−759. https://doi.org/10.1038/nbt.3576

[87]

V. Moiseeva, A. Cisneros, V. Sica, et al. Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration. Nature, 2023, 613: 169−178. https://doi.org/10.1038/s41586-022-05535-x

[88]

Y. Ge, Y. Miao, S. Gur-Cohen, et al. The aging skin microenvironment dictates stem cell behavior. Proceedings of the National Academy of Sciences of the United States of America, 2020, 117: 5339−5350. https://doi.org/10.1073/pnas.1901720117

[89]

P.M. Gilbert, K.L. Havenstrite, K.E.G. Magnusson, et al. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science), 2010, 329: 1078−1081. https://doi.org/10.1126/science.1191035

[90]

N.A. Terrault, C. Francoz, M. Berenguer, et al. Liver Transplantation 2023: Status Report, Current and Future Challenges. Clinical Gastroenterology and Hepatology, 2023, 21(8): 2150−2166. https://doi.org/10.1016/j.cgh.2023.04.005

[91]

I. Kaur, A. Vasudevan, P. Rawal, et al. Primary Hepatocyte Isolation and Cultures: Technical Aspects, Challenges and Advancements. Bioengineering, 2023, 10(2): 131. https://doi.org/10.3390/bioengineering10020131

[92]

W.C. Ng, Y. Lokanathan, M.M. Baki, et al. Tissue engineering as a promising treatment for glottic insufficiency: a review on biomolecules and cell-laden hydrogel. Biomedicines, 2022, 10(12): 3082. https://doi.org/10.3390/biomedicines10123082

[93]

S. Ye, J.W.B. Boeter, M. Mihajlovic, et al. A chemically defined hydrogel for human liver organoid culture. Advanced Functional Materials, 2020, 30: 2000893. https://doi.org/10.1002/adfm.202000893

[94]

W. Meng, H. Sun, T. Mu, et al. Chitosan-based Pickering emulsion: A comprehensive review on their stabilizers, bioavailability, applications and regulations. Carbohydrate Polymers, 2023, 304: 120491. https://doi.org/10.1016/j.carbpol.2022.120491

[95]

J. Ding, Y. Dun, D. He, et al. RGD-hydrogel improves the therapeutic effect of bone marrow-derived mesenchymal stem cells on phosgene-induced acute lung injury in rats. Computational Intelligence and Neuroscience, 2022, 2022: 2743878. https://doi.org/10.1155/2022/2743878

[96]

M. R. Poorna, R. Jayakumar, J. Chen , et al. Hydrogels: A potential platform for induced pluripotent stem cell culture and differentiation. Colloids and Surfaces B:Biointerfaces, 2021, 207: 111991. https://doi.org/10.1016/j.colsurfb.2021.111991

[97]

J.Vallverdu, G.D.L.T. Martinez, I. Mannaerts, et al. Directed differentiation of human induced pluripotent stem cells to hepatic stellate cells. Nature Protocols, 2021, 16: 2542−2563. https://doi.org/10.1038/s41596-021-00509-1

[98]

J. Kasuya, R. Sudo, T. Mitaka, et al. Hepatic stellate cell-mediated three dimensional hepatocyte and endothelial cell triculture model. Tissue Engineering Part A, 2011, 17: 361−370. https://doi.org/10.1089/ten.tea.2023.0007

[99]

Y. Luo, C. Lou, S. Zhang, et al. Three-dimensional hydrogel culture conditions promote the differentiation of human induced pluripotent stem cells into hepatocytes. Cytotherapy, 2018, 20: 95−107. https://doi.org/10.1016/j.jcyt.2017.08.008

[100]

S. Dupont, L. Morsut, M. Aragona, et al. Role of YAP/TAZ in mechanotransduction. Nature, 2011, 474: 179−183. https://doi.org/10.1038/nature10137

[101]
A.P. Kourouklis, K.B. Kaylan, G. H. Underhill G H. Substrate stiffness and matrix composition coordinately control the differentiation of liver progenitor cells. Biomaterials, 2016, 99: 82–94.
[102]

V. Natarajan, E. J. Berglund, D.X.Chen, et al. Substrate stiffness regulates primary hepatocyte functions. RSC Advances, 2015, 5: 80956−80966. https://doi.org/10.1039/C5RA15208A

Nano Biomedicine and Engineering
Pages 188-202
Cite this article:
Luo Y, Gao Y. Potential Role of Hydrogels in Stem Cell Culture and Hepatocyte Differentiation. Nano Biomedicine and Engineering, 2024, 16(2): 188-202. https://doi.org/10.26599/NBE.2024.9290055

820

Views

97

Downloads

0

Crossref

0

Scopus

Altmetrics

Received: 27 July 2023
Revised: 30 October 2023
Accepted: 21 November 2023
Published: 10 January 2024
© The Author(s) 2024.

This is an open-access article distributed under  the  terms  of  the  Creative  Commons  Attribution  4.0 International  License (CC BY) (http://creativecommons.org/licenses/by/4.0/), which  permits  unrestricted  use,  distribution,  and reproduction in any medium, provided the original author and source are credited.

Return