1
|
Elvitigala KCML, Mohan L, Mubarok W, Sakai S. Phototuning of Hyaluronic-Acid-Based Hydrogel Properties to Control Network Formation in Human Vascular Endothelial Cells. Adv Healthc Mater 2024; 13:e2303787. [PMID: 38684108 PMCID: PMC11468695 DOI: 10.1002/adhm.202303787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/06/2024] [Indexed: 05/02/2024]
Abstract
In vitro network formation by endothelial cells serves as a fundamental model for studies aimed at understanding angiogenesis. The morphogenesis of these cells to form a network is intricately regulated by the mechanical and biochemical properties of the extracellular matrix. Here the effects of modulating these properties in hydrogels derived from phenolated hyaluronic acid (HA-Ph) and phenolated gelatin (Gelatin-Ph) are presented. Visible-light irradiation in the presence of tris(2,2'-bipyridyl)ruthenium(II) chloride hexahydrate and sodium persulfate induces the crosslinking of these polymers, thereby forming a hydrogel and degrading HA-Ph. Human vascular endothelial cells form networks on the hydrogel prepared by visible-light irradiation for 45 min (42 W cm-2 at 450 nm) but not on the hydrogels prepared by irradiation for 15, 30, or 60 min. The irradiation time-dependent degradation of HA-Ph and the changes in the mechanical stiffness of the hydrogels, coupled with the expressions of RhoA and β-actin genes and CD44 receptors in the cells, reveal that the network formation is synergistically influenced by the hydrogel stiffness and HA-Ph degradation. These findings highlight the potential of tailoring HA-based hydrogel properties to modulate human vascular endothelial cell responses, which is critical for advancing their application in vascular tissue engineering.
Collapse
Affiliation(s)
| | - Lakshmi Mohan
- Department of BioengineeringHenry Samueli School of EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Wildan Mubarok
- Department of Materials Engineering ScienceGraduate School of Engineering ScienceOsaka UniversityToyonakaOsaka560‐8531Japan
| | - Shinji Sakai
- Department of Materials Engineering ScienceGraduate School of Engineering ScienceOsaka UniversityToyonakaOsaka560‐8531Japan
| |
Collapse
|
2
|
Qin W, Xing T, Qin S, Tang B, Chen W. BMSCs-driven graphite oxide-grafted-carbon fibers reinforced polyetheretherketone composites as functional implants: in vivo biosafety and osteogenesis. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1343-1358. [PMID: 38493406 DOI: 10.1080/09205063.2024.2328877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 01/08/2024] [Indexed: 03/18/2024]
Abstract
Mesenchymal stem cells (MSCs) are increasingly becoming a potential treatment approach for bone injuries due to the multi-lineage differentiation potential, ability to recognize damaged tissue sites and secrete bioactive factors that can enhance tissue repair. The aim of this work was to improve osteogenesis of carbon fibers reinforced polyetheretherketone (CF/PEEK) implants through bone marrow mesenchymal stem cells (BMSCs)-based therapy. Moreover, bioactive graphene oxide (GO) was introduced into CF/PEEK by grafting GO onto CF to boost the osteogenic efficiency of BMSCs. Subsequently, CF/PEEK was implanted into the symmetrical skull defect models of SD rats. Then in vivo biosafety and osteogenesis were evaluated. The results indicated that surface wettability of CF/PEEK was effectively improved by GO, which was beneficial for the adhesion of BMSCs. The pathological tissue sections stained with H&E showed no significant pathological change in the main organs including heart, liver, spleen, lung and kidney, which indicated no acute systemic toxicity. Furthermore, bone mineralization deposition rate of CF/PEEK containing GO was 2.2 times that of pure CF/PEEK. The X-ray test showed that the surface of CF/PEEK containing GO was obviously covered by more newly formed bone tissue than pure CF/PEEK after 8 weeks of implantation. This work demonstrated that GO effectively enhanced surface bioactivity of CF/PEEK and assisted BMSCs in accelerating differentiation into bone tissue, providing a feasible strategy for improving osteogenesis of PEEK and CF/PEEK.
Collapse
Affiliation(s)
- Wen Qin
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Tong Xing
- Engineering Research Center of Heavy Mechanical, Ministry of Education, Taiyuan University of Science and Technology, Taiyuan, China
| | - Shengnan Qin
- Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Bin Tang
- College of Material Science and Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Weiyi Chen
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, China
| |
Collapse
|
3
|
Lin S, He X, Wang Y, Chen Y, Lin A. Emerging role of lncRNAs as mechanical signaling molecules in mechanotransduction and their association with Hippo-YAP signaling: a review. J Zhejiang Univ Sci B 2024; 25:280-292. [PMID: 38584091 PMCID: PMC11009445 DOI: 10.1631/jzus.b2300497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/11/2023] [Indexed: 04/09/2024]
Abstract
Cells within tissues are subject to various mechanical forces, including hydrostatic pressure, shear stress, compression, and tension. These mechanical stimuli can be converted into biochemical signals through mechanoreceptors or cytoskeleton-dependent response processes, shaping the microenvironment and maintaining cellular physiological balance. Several studies have demonstrated the roles of Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ) as mechanotransducers, exerting dynamic influence on cellular phenotypes including differentiation and disease pathogenesis. This regulatory function entails the involvement of the cytoskeleton, nucleoskeleton, integrin, focal adhesions (FAs), and the integration of multiple signaling pathways, including extracellular signal-regulated kinase (ERK), wingless/integrated (WNT), and Hippo signaling. Furthermore, emerging evidence substantiates the implication of long non-coding RNAs (lncRNAs) as mechanosensitive molecules in cellular mechanotransduction. In this review, we discuss the mechanisms through which YAP/TAZ and lncRNAs serve as effectors in responding to mechanical stimuli. Additionally, we summarize and elaborate on the crucial signal molecules involved in mechanotransduction.
Collapse
Affiliation(s)
- Siyi Lin
- College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyu He
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Ying Wang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Yu Chen
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Cancer Center, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China.
- International School of Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Hangzhou 310058, China.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
4
|
Wiebe-Ben Zakour KE, Kaya S, Matros JC, Hacker MC, Cheikh-Rouhou A, Spaniol K, Geerling G, Witt J. Enhancement of lacrimal gland cell function by decellularized lacrimal gland derived hydrogel. Biofabrication 2024; 16:025008. [PMID: 38241707 DOI: 10.1088/1758-5090/ad2082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/19/2024] [Indexed: 01/21/2024]
Abstract
Sustainable treatment of aqueous deficient dry eye (ADDE) represents an unmet medical need and therefore requires new curative and regenerative approaches based on appropriatein vitromodels. Tissue specific hydrogels retain the individual biochemical composition of the extracellular matrix and thus promote the inherent cell´s physiological function. Hence, we created a decellularized lacrimal gland (LG) hydrogel (dLG-HG) meeting the requirements for a bioink as the basis of a LG model with potential forin vitroADDE studies. Varying hydrolysis durations were compared to obtain dLG-HG with best possible physical and ultrastructural properties while preserving the original biochemical composition. A particular focus was placed on dLG-HG´s impact on viability and functionality of LG associated cell types with relevance for a futurein vitromodel in comparison to the unspecific single component hydrogel collagen type-I (Col) and the common cell culture substrate Matrigel. Proliferation of LG epithelial cells (EpC), LG mesenchymal stem cells, and endothelial cells cultured on dLG-HG was enhanced compared to culture on Matrigel. Most importantly with respect to a functionalin vitromodel, the secretion capacity of EpC cultured on dLG-HG was higher than that of EpC cultured on Col or Matrigel. In addition to these promising cell related properties, a rapid matrix metalloproteinase-dependent biodegradation was observed, which on the one hand suggests a lively cell-matrix interaction, but on the other hand limits the cultivation period. Concluding, dLG-HG possesses decisive properties for the tissue engineering of a LGin vitromodel such as cytocompatibility and promotion of secretion, making it superior to unspecific cell culture substrates. However, deceleration of biodegradation should be addressed in future experiments.
Collapse
Affiliation(s)
- Katharina E Wiebe-Ben Zakour
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Sema Kaya
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Julia C Matros
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Michael C Hacker
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Amina Cheikh-Rouhou
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Kristina Spaniol
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Gerd Geerling
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Joana Witt
- Department of Ophthalmology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
5
|
White SE, Karbasion N, Snider JC, Florian-Rodriguez M, Bersi MR, Miller KS. Remodeling of murine vaginal smooth muscle function with reproductive age and elastic fiber disruption. Acta Biomater 2024; 175:186-198. [PMID: 38151068 DOI: 10.1016/j.actbio.2023.12.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
Advanced maternal age during pregnancy is associated with increased risk of vaginal tearing during delivery and maladaptive postpartum healing. Although the underlying mechanisms of age-related vaginal injuries are not fully elucidated, changes in vaginal microstructure may contribute. Smooth muscle cells promote the contractile nature of the vagina and contribute to pelvic floor stability. While menopause is associated with decreased vaginal smooth muscle content, whether contractile changes occur before the onset of menopause remains unknown. Therefore, the first objective of this study was to quantify the active mechanical behavior of the murine vagina with age. Further, aging is associated with decreased vaginal elastin content. As such, the second objective was to determine if elastic fiber disruption alters vaginal contractility. Vaginal samples from mice aged 2-14 months were used in maximum contractility experiments and biaxial extension-inflation protocols. To evaluate the role of elastic fibers with age, half of the vaginal samples were randomly allocated to enzymatic elastic fiber disruption. Contractile potential decreased and vaginal material stiffness increased with age. These age-related changes in smooth muscle function may be due, in part, to changes in microstructural composition or contractile gene expression. Furthermore, elastic fiber disruption had a diminished effect on smooth muscle contractility in older mice. This suggests a decreased functional role of elastic fibers with age. Quantifying the age-dependent mechanical contribution of smooth muscle cells and elastic fibers to vaginal properties provides a first step towards better understanding how age-related changes in vaginal structure may contribute to tissue integrity and healing. STATEMENT OF SIGNIFICANCE: Advanced maternal age at the time of pregnancy is linked to increased risks of vaginal tearing during delivery, postpartum hemorrhaging, and the development of pelvic floor disorders. While the underlying causes of increased vaginal injuries with age and associated pathologies remain unclear, changes in vaginal microstructure, such as elastic fibers and smooth muscle cells, may contribute. Menopause is associated with fragmented elastic fibers and decreased smooth muscle content; however, how reproductive aging affects changes in the vaginal composition and the mechanical properties remains unknown. Quantifying the mechanical contribution of smooth muscle cells and elastic fibers to vaginal properties with age will advance understanding of the potential structural causes of age-related changes to tissue integrity and healing.
Collapse
Affiliation(s)
- Shelby E White
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Niyousha Karbasion
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - J Caleb Snider
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Maria Florian-Rodriguez
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthew R Bersi
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Kristin S Miller
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, USA; Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
6
|
Kawecki NS, Norris SCP, Xu Y, Wu Y, Davis AR, Fridman E, Chen KK, Crosbie RH, Garmyn AJ, Li S, Mason TG, Rowat AC. Engineering multicomponent tissue by spontaneous adhesion of myogenic and adipogenic microtissues cultured with customized scaffolds. Food Res Int 2023; 172:113080. [PMID: 37689860 DOI: 10.1016/j.foodres.2023.113080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 09/11/2023]
Abstract
The integration of intramuscular fat-or marbling-into cultured meat will be critical for meat texture, mouthfeel, flavor, and thus consumer appeal. However, culturing muscle tissue with marbling is challenging since myocytes and adipocytes have different media and scaffold requirements for optimal growth and differentiation. Here, we present an approach to engineer multicomponent tissue using myogenic and adipogenic microtissues. The key innovation in our approach is the engineering of myogenic and adipogenic microtissues using scaffolds with customized physical properties; we use these microtissues as building blocks that spontaneously adhere to produce multicomponent tissue, or marbled cultured meat. Myocytes are grown and differentiated on gelatin nanofiber scaffolds with aligned topology that mimic the aligned structure of skeletal muscle and promotes the formation of myotubes in both primary rabbit skeletal muscle and murine C2C12 cells. Pre-adipocytes are cultured and differentiated on edible gelatin microbead scaffolds, which are customized to have a physiologically-relevant stiffness, and promote lipid accumulation in both primary rabbit and murine 3T3-L1 pre-adipocytes. After harvesting and stacking the individual myogenic and adipogenic microtissues, we find that the resultant multicomponent tissues adhere into intact structures within 6-12 h in culture. The resultant multicomponent 3D tissue constructs show behavior of a solid material with a Young's modulus of ∼ 2 ± 0.4 kPa and an ultimate tensile strength of ∼ 23 ± 7 kPa without the use of additional crosslinkers. Using this approach, we generate marbled cultured meat with ∼ mm to ∼ cm thickness, which has a protein content of ∼ 4 ± 2 g/100 g that is comparable to a conventionally produced Wagyu steak with a protein content of ∼ 9 ± 4 g/100 g. We show the translatability of this layer-by-layer assembly approach for microtissues across primary rabbit cells, murine cell lines, as well as for gelatin and plant-based scaffolds, which demonstrates a strategy to generate edible marbled meats derived from different species and scaffold materials.
Collapse
Affiliation(s)
- N Stephanie Kawecki
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sam C P Norris
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yixuan Xu
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yifan Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ashton R Davis
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ester Fridman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kathleen K Chen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, University of California LA, USA; Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea J Garmyn
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas G Mason
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Physics and Astronomy, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
7
|
Mielnicka A, Kołodziej T, Dziob D, Lasota S, Sroka J, Rajfur Z. Impact of elastic substrate on the dynamic heterogeneity of WC256 Walker carcinosarcoma cells. Sci Rep 2023; 13:15743. [PMID: 37735532 PMCID: PMC10514059 DOI: 10.1038/s41598-023-35313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 05/16/2023] [Indexed: 09/23/2023] Open
Abstract
Cellular heterogeneity is a phenomenon in which cell populations are composed of subpopulations that vary in their behavior. Heterogeneity is particularly pronounced in cancer cells and can affect the efficacy of oncological therapies. Previous studies have considered heterogeneity dynamics to be indicative of evolutionary changes within subpopulations; however, these studies do not consider the short-time morphological plasticity of cells. Physical properties of the microenvironment elasticity have also been poorly investigated within the context of cellular heterogeneity, despite its role in determining cellular behavior. This article demonstrates that cellular heterogeneity can be highly dynamic and dependent on the micromechanical properties of the substrate. During observation, migrating Walker carcinosarcoma WC256 cells were observed to belong to different subpopulations, in which their morphologies and migration strategies differed. Furthermore, the application of an elastic substrate (E = 40 kPa) modified three aspects of cellular heterogeneity: the occurrence of subpopulations, the occurrence of transitions between subpopulations, and cellular migration and morphology. These findings provide a new perspective in the analysis of cellular heterogeneity, whereby it may not be a static feature of cancer cell populations, instead varying over time. This helps further the understanding of cancer cell behavior, including their phenotype and migration strategy, which may help to improve cancer therapies by extending their suitability to investigate tumor heterogeneity.
Collapse
Affiliation(s)
- Aleksandra Mielnicka
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, ul. Lojasiewicza 11, 30-348, Kraków, Poland
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, PAS, ul. Ludwika Pasteura 3, 02-093, Warsaw, Poland
| | - Tomasz Kołodziej
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, ul. Lojasiewicza 11, 30-348, Kraków, Poland
- Department of Pharmaceutical Biophysics, Faculty of Pharmacy, Jagiellonian University Medical College, ul. Medyczna 9, 30-688, Kraków, Poland
| | - Daniel Dziob
- Department of Pharmaceutical Biophysics, Faculty of Pharmacy, Jagiellonian University Medical College, ul. Medyczna 9, 30-688, Kraków, Poland
| | - Sławomir Lasota
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387, Kraków, Poland
| | - Jolanta Sroka
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387, Kraków, Poland
| | - Zenon Rajfur
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, ul. Lojasiewicza 11, 30-348, Kraków, Poland.
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348, Kraków, Poland.
| |
Collapse
|
8
|
Umer A, Ghouri MD, Muyizere T, Aqib RM, Muhaymin A, Cai R, Chen C. Engineered Nano-Bio Interfaces for Stem Cell Therapy. PRECISION CHEMISTRY 2023; 1:341-356. [PMID: 37654807 PMCID: PMC10466455 DOI: 10.1021/prechem.3c00056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 09/02/2023]
Abstract
Engineered nanomaterials (ENMs) with different topographies provide effective nano-bio interfaces for controlling the differentiation of stem cells. The interaction of stem cells with nanoscale topographies and chemical cues in their microenvironment at the nano-bio interface can guide their fate. The use of nanotopographical cues, in particular nanorods, nanopillars, nanogrooves, nanofibers, and nanopits, as well as biochemical forces mediated factors, including growth factors, cytokines, and extracellular matrix proteins, can significantly impact stem cell differentiation. These factors were seen as very effective in determining the proliferation and spreading of stem cells. The specific outgrowth of stem cells can be decided with size variation of topographic nanomaterial along with variation in matrix stiffness and surface structure like a special arrangement. The precision chemistry enabled controlled design, synthesis, and chemical composition of ENMs can regulate stem cell behaviors. The parameters of size such as aspect ratio, diameter, and pore size of nanotopographic structures are the main factors for specific termination of stem cells. Protein corona nanoparticles (NPs) have shown a powerful facet in stem cell therapy, where combining specific proteins could facilitate a certain stem cell differentiation and cellular proliferation. Nano-bio reactions implicate the interaction between biological entities and nanoparticles, which can be used to tailor the stem cells' culmination. The ion release can also be a parameter to enhance cellular proliferation and to commit the early differentiation of stem cells. Further research is needed to fully understand the mechanisms underlying the interactions between engineered nano-bio interfaces and stem cells and to develop optimized regenerative medicine and tissue engineering designs.
Collapse
Affiliation(s)
- Arsalan Umer
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
- University
of Chinese Academy of Sciences, Beijing100049, China
| | - Muhammad Daniyal Ghouri
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
- University
of Chinese Academy of Sciences, Beijing100049, China
| | - Theoneste Muyizere
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Raja Muhammad Aqib
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Abdul Muhaymin
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Rong Cai
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
| | - Chunying Chen
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
& CAS Center for Excellence in Nanoscience and Technology of China, Chinese Academy of Sciences (CAS), Beijing100190, China
- University
of Chinese Academy of Sciences, Beijing100049, China
- GBA
National Institute for Nanotechnology Innovation, Guangdong 5110700, China
| |
Collapse
|
9
|
Wang F, Guo K, Nan L, Wang S, Lu J, Wang Q, Ba Z, Huang Y, Wu D. Kartogenin-loaded hydrogel promotes intervertebral disc repair via protecting MSCs against reactive oxygen species microenvironment by Nrf2/TXNIP/NLRP3 axis. Free Radic Biol Med 2023; 204:128-150. [PMID: 37149010 DOI: 10.1016/j.freeradbiomed.2023.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023]
Abstract
Intervertebral disc (IVD) degeneration (IDD) and the consequent low back pain present a major medical challenge. Stem cell-based tissue engineering is promising for the treatment of IDD. However, stem cell-based treatment is severely impaired by the increased generation of reactive oxygen species (ROS) in degenerative disc, which can lead to a high level of cell dysfunction and even death. In this study, a kartogenin (KGN)@PLGA-GelMA/PRP composite hydrogel was designed and used as a carrier of ADSCs-based therapies in disc repair. Injectable composite hydrogel act as a carrier for controlled release of KGN and deliver ADSCs to the degenerative disc. The released KGN can stimulate the differentiation of ADSCs into a nucleus pulposus (NP) -like phenotype and boost antioxidant capacity of ADSCs via activating Nrf2/TXNIP/NLRP3 axis. Furthermore, the composite hydrogel combined with ADSCs attenuated the in vivo degeneration of rat IVDs, maintained IVD tissue integrity and accelerated the synthesis of NP-like extracellular matrix. Therefore, the KGN@PLGA-GelMA/PRP composite hydrogel is a promising strategy for stem cell-based therapies of IDD.
Collapse
Affiliation(s)
- Feng Wang
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Kai Guo
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Liping Nan
- Center for Orthopaedic Science and Translational Medicine, Department of Orthopedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Shuguang Wang
- Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Jiawei Lu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qiang Wang
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhaoyu Ba
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yufeng Huang
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Desheng Wu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
10
|
Holiel AA, Mustafa HM, Sedek EM. Biodegradation of an injectable treated dentin matrix hydrogel as a novel pulp capping agent for dentin regeneration. BMC Oral Health 2023; 23:126. [PMID: 36841767 PMCID: PMC9960635 DOI: 10.1186/s12903-023-02831-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 02/22/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND A novel injectable mixture termed treated dentin matrix hydrogel (TDMH) has been introduced for restoring dentin defect in DPC. However, no study evaluated its physiological biodegradation. Therefore, the present study aimed to assess scaffold homogeneity, mechanical properties and biodegradability in vitro and in vivo and the regenerated dentin induced by TDMH as a novel pulp capping agent in human permanent teeth. METHODS Three TDMH discs were weighted, and dry/wet ratios were calculated in four slices from each disc to evaluate homogeneity. Hydrogel discs were also analyzed in triplicate to measure the compressive strength using a universal testing machine. The in vitro degradation behavior of hydrogel in PBS at 37 °C for 2 months was also investigated by monitoring the percent weight change. Moreover, 20 intact fully erupted premolars were included for assessment of TDMH in vivo biodegradation when used as a novel injectable pulp capping agent. The capped teeth were divided into four equal groups according to extraction interval after 2-, 8-, 12- and 16-weeks, stained with hematoxylin-eosin for histological and histomorphometric evaluation. Statistical analysis was performed using F test (ANOVA) and post hoc test (p = 0.05). RESULTS No statistical differences among hydrogel slices were detected with (p = 0.192) according to homogeneity. TDMH compression modulus was (30.45 ± 1.11 kPa). Hydrogel retained its shape well up to 4 weeks and after 8 weeks completely degraded. Histological analysis after 16 weeks showed a significant reduction in TDMH area and a simultaneous significant increase in the new dentin area. The mean values of TDMH were 58.8% ± 5.9 and 9.8% ± 3.3 at 2 and 16 weeks, while the new dentin occupied 9.5% ± 2.8 at 2 weeks and 82.9% ± 3.8 at 16 weeks. CONCLUSIONS TDMH was homogenous and exhibited significant stability and almost completely recovered after excessive compression. TDMH generally maintained their bulk geometry throughout 7 weeks. The in vivo response to TDMH was characterized by extensive degradation of the hydrogel and dentin matrix particles and abundant formation of new dentin. The degradation rate of TDMH matched the rate of new dentin formation. TRIAL REGISTRATION PACTR201901866476410: 30/1/2019.
Collapse
Affiliation(s)
- Ahmed A Holiel
- Conservative Dentistry Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.
| | - Hossam M Mustafa
- Oral Biology Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Eman M Sedek
- Dental Biomaterials Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| |
Collapse
|
11
|
Understanding How Cells Probe the World: A Preliminary Step towards Modeling Cell Behavior? Int J Mol Sci 2023; 24:ijms24032266. [PMID: 36768586 PMCID: PMC9916635 DOI: 10.3390/ijms24032266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Cell biologists have long aimed at quantitatively modeling cell function. Recently, the outstanding progress of high-throughput measurement methods and data processing tools has made this a realistic goal. The aim of this paper is twofold: First, to suggest that, while much progress has been done in modeling cell states and transitions, current accounts of environmental cues driving these transitions remain insufficient. There is a need to provide an integrated view of the biochemical, topographical and mechanical information processed by cells to take decisions. It might be rewarding in the near future to try to connect cell environmental cues to physiologically relevant outcomes rather than modeling relationships between these cues and internal signaling networks. The second aim of this paper is to review exogenous signals that are sensed by living cells and significantly influence fate decisions. Indeed, in addition to the composition of the surrounding medium, cells are highly sensitive to the properties of neighboring surfaces, including the spatial organization of anchored molecules and substrate mechanical and topographical properties. These properties should thus be included in models of cell behavior. It is also suggested that attempts at cell modeling could strongly benefit from two research lines: (i) trying to decipher the way cells encode the information they retrieve from environment analysis, and (ii) developing more standardized means of assessing the quality of proposed models, as was done in other research domains such as protein structure prediction.
Collapse
|
12
|
Bai M, Zhang Z, Chen H, Liu X, Xie J. Paxillin tunes the relationship between cell-matrix and cell-cell adhesions to regulate stiffness-dependent dentinogenesis. Regen Biomater 2022; 10:rbac100. [PMID: 36683745 PMCID: PMC9847533 DOI: 10.1093/rb/rbac100] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Mechanical stiffness is recognized as a key physical factor and directs cell function via a mechanotransduction process, from extracellular physical cues to intracellular signaling cascades that affect transcriptional activity. Cells continually receive mechanical signals from both the surrounding matrix and adjacent cells. However, how mechanical stiffness cue at cell-substrate interfaces coordinates cell-cell junctions in guiding mesenchymal stem cell behaviors is poorly understood. Here, polydimethylsiloxane substrates with different stiffnesses were used to study mechanosensation/transduction mechanisms in controlling odontogenic differentiation of dental papilla cells (DPCs). DPC phenotypes (morphology and differentiation) changed in response to the applied force derived from stiff substrates. Significantly, higher expression of paxillin on stiffer substrates promoted DPC dentinogenesis. Upon treatment with siRNA to knockdown paxillin, N-cadherin increased mainly in the cytomembrane at the area of cell-cell contacts, whereas β-catenin decreased in the nuclei. The result of a double luciferase reporter assay showed that stiffness promoted β-catenin binding to TCF, which could coactivate the target genes associated with odontogenic differentiation, as evidenced by bioinformatics analysis. Finally, we determined that the addition of a β-catenin inhibitor suppressed DPC mineralization in all the stiffness groups. Thus, our results indicated that a mechanotransduction process from cell-substrate interactions to cell-cell adhesions was required for DPC odontogenic differentiation under the stimulation of substrate stiffness. This finding suggests that stem cell fate specification under the stimulus of stiffness at the substrates is based on crosstalk between substrate interactions and adherens junctions, which provides an essential mechanism for cell-based tissue engineering.
Collapse
Affiliation(s)
- Mingru Bai
- Correspondence address. E-mail: (M.B.); (J.X.)
| | - Zhaowei Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Huiyu Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoyu Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- Correspondence address. E-mail: (M.B.); (J.X.)
| |
Collapse
|
13
|
Effects of sodium hypochlorite and ethylenediaminetetraacetic acid on proliferation, osteogenic/odontogenic differentiation, and mechanosensitive gene expression of human dental pulp stem cells. Tissue Cell 2022; 79:101955. [DOI: 10.1016/j.tice.2022.101955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
|
14
|
Han M, Yang H, Lu X, Li Y, Liu Z, Li F, Shang Z, Wang X, Li X, Li J, Liu H, Xin T. Three-Dimensional-Cultured MSC-Derived Exosome-Hydrogel Hybrid Microneedle Array Patch for Spinal Cord Repair. NANO LETTERS 2022; 22:6391-6401. [PMID: 35876503 DOI: 10.1021/acs.nanolett.2c02259] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Exosomes derived from mesenchymal stem cells (MSCs) have been proven to exhibit great potentials in spinal cord injury (SCI) therapy. However, conventional two-dimensional (2D) culture will inevitably lead to the loss of stemness of MSCs, which substantially limits the therapeutic potency of MSCs exosomes (2D-Exo). Exosomes derived from three-dimensional culture (3D-Exo) possess higher therapeutic efficiency which have wide applications in spinal cord therapy. Typically, conventional exosome therapy that relies on local repeated injection results in secondary injury and low efficiency. It is urgent to develop a more reliable, convenient, and effective exosome delivery method to achieve constant in situ exosomes release. Herein, we proposed a controlled 3D-exohydrogel hybrid microneedle array patch to achieve SCI repair in situ. Our studies suggested that MSCs with 3D-culturing could maintain their stemness, and consequently, 3D-Exo effectively reduced SCI-induced inflammation and glial scarring. Thus, it is a promising therapeutic strategy for the treatment of SCI.
Collapse
Affiliation(s)
- Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P.R. China
| | - Hongru Yang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, P.R. China
| | - Xiangdong Lu
- Department of Neurosurgery, People's Hospital Affiliated to Shandong First Medical University, Jinan 250117, P.R. China
| | - Yuming Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, P.R. China
| | - Feng Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Zehan Shang
- Department of Neurosurgery, Shangdong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, P.R. China
| | - Xiaofeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Xuze Li
- Department of Neurosurgery, Shangdong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250014, P.R. China
| | - Junliang Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, P.R. China
- Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan 250022, P.R. China
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250014, P.R. China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P.R. China
| |
Collapse
|
15
|
Selvaraj S, Rupert S, Nandabalan SK, Anbalagan C, Rajaram PS, Satyanesan J, Vennila R, Rajagopal S. Effect of Cell-Derived Matrices on Growth and Differentiation of Human Wharton's Jelly-Derived Mesenchymal Stem Cells. Cells Tissues Organs 2022; 213:67-78. [PMID: 35908543 DOI: 10.1159/000526153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/17/2022] [Indexed: 02/18/2024] Open
Abstract
Cell-derived matrices (CDMs) are scaffolds constructed by decellularization of cellular matrices from different tissues and organs. Since CDMs mimic the extracellular matrices (ECMs) of native tissues, it plays an essential role in the preparation of bioscaffolds. CDM scaffolds from mesenchymal stem cells (MSCs) have been reported to support cell adhesion and proliferation of its own cells. Therefore, in this study we aimed to test if growth of human Wharton's jelly-derived MSCs may be enhanced when cultured on their own CDMs. To do this, MSCs were induced to generate ECM using ascorbic acid. Thus, obtained matrices were decellularized and characterized quantitatively for changes in their biochemical components (total protein, collagen, glycosaminoglycans) and qualitatively for fibronectin, laminin, and collagen (I & IV) by immunostaining. Our results show the retention of essential ECM components in the decellularized WJ-MSC-derived matrix (WJ-CDM). The influence of WJ-CDM on proliferation and differentiation of WJ-MSCs were evaluated by comparing their growth on collagen and fibronectin-only coated plates. A non-coated tissue culture polystyrene plate (TCPS) served as control. Our cell proliferation results show that no significant changes were observed in the proliferation of MSCs when cultured on WJ-CDM as compared to the bio-coated and non-coated cultures. However, gene expression analysis of the differentiation process showed that osteogenic and adipogenic differentiation potential of the WJ-MSCs was significantly increased upon culturing them on WJ-CDM. In conclusion, the present study reveals that the WJ-MSCs cultured on WJ-CDM may augment osteogenic and adipogenic differentiation.
Collapse
Affiliation(s)
- Sakthivel Selvaraj
- Stem Cell Research Centre, Government Stanley Medical College and Hospital, Chennai, India,
| | - Secunda Rupert
- Stem Cell Research Centre, Government Stanley Medical College and Hospital, Chennai, India
| | | | - Charumathi Anbalagan
- Stem Cell Research Centre, Government Stanley Medical College and Hospital, Chennai, India
| | | | - Jeswanth Satyanesan
- Stem Cell Research Centre, Government Stanley Medical College and Hospital, Chennai, India
| | - Rosy Vennila
- Karur Government Medical College and Hospital, Karur, India
| | - Surendran Rajagopal
- Hepato-Pancreato-Biliary Centre for Surgery & Transplantation, MIOT International, Chennai, India
| |
Collapse
|
16
|
Anggraini D, Ota N, Shen Y, Tang T, Tanaka Y, Hosokawa Y, Li M, Yalikun Y. Recent advances in microfluidic devices for single-cell cultivation: methods and applications. LAB ON A CHIP 2022; 22:1438-1468. [PMID: 35274649 DOI: 10.1039/d1lc01030a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Single-cell analysis is essential to improve our understanding of cell functionality from cellular and subcellular aspects for diagnosis and therapy. Single-cell cultivation is one of the most important processes in single-cell analysis, which allows the monitoring of actual information of individual cells and provides sufficient single-cell clones and cell-derived products for further analysis. The microfluidic device is a fast-rising system that offers efficient, effective, and sensitive single-cell cultivation and real-time single-cell analysis conducted either on-chip or off-chip. Here, we introduce the importance of single-cell cultivation from the aspects of cellular and subcellular studies. We highlight the materials and structures utilized in microfluidic devices for single-cell cultivation. We further discuss biological applications utilizing single-cell cultivation-based microfluidics, such as cellular phenotyping, cell-cell interactions, and omics profiling. Finally, present limitations and future prospects of microfluidics for single-cell cultivation are also discussed.
Collapse
Affiliation(s)
- Dian Anggraini
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan.
| | - Nobutoshi Ota
- Center for Biosystems Dynamics Research (BDR), RIKEN, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yigang Shen
- Center for Biosystems Dynamics Research (BDR), RIKEN, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Tao Tang
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan.
| | - Yo Tanaka
- Center for Biosystems Dynamics Research (BDR), RIKEN, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoichiroh Hosokawa
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan.
| | - Ming Li
- School of Engineering, Macquarie University, Sydney 2122, Australia.
| | - Yaxiaer Yalikun
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan.
- Center for Biosystems Dynamics Research (BDR), RIKEN, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Impact of Microenvironmental Changes during Degeneration on Intervertebral Disc Progenitor Cells: A Comparison with Mesenchymal Stem Cells. Bioengineering (Basel) 2022; 9:bioengineering9040148. [PMID: 35447707 PMCID: PMC9025850 DOI: 10.3390/bioengineering9040148] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
Intervertebral disc (IVD) degeneration occurs with natural ageing and is linked to low back pain, a common disease. As an avascular tissue, the microenvironment inside the IVD is harsh. During degeneration, the condition becomes even more compromised, presenting a significant challenge to the survival and function of the resident cells, as well as to any regeneration attempts using cell implantation. Mesenchymal stem cells (MSCs) have been proposed as a candidate stem cell tool for IVD regeneration. Recently, endogenous IVD progenitor cells have been identified inside the IVD, highlighting their potential for self-repair. IVD progenitor cells have properties similar to MSCs, with minor differences in potency and surface marker expression. Currently, it is unclear how IVD progenitor cells react to microenvironmental factors and in what ways they possibly behave differently to MSCs. Here, we first summarized the microenvironmental factors presented in the IVD and their changes during degeneration. Then, we analyzed the available studies on the responses of IVD progenitor cells and MSCs to these factors, and made comparisons between these two types of cells, when possible, in an attempt to achieve a clear understanding of the characteristics of IVD progenitor cells when compared to MSCs; as well as, to provide possible clues to cell fate after implantation, which may facilitate future manipulation and design of IVD regeneration studies.
Collapse
|
18
|
Subbiah R, Balbinot GDS, Athirasala A, Collares FM, Sereda G, Bertassoni LE. Nanoscale mineralization of cell-laden methacrylated gelatin hydrogels using calcium carbonate-calcium citrate core-shell microparticles. J Mater Chem B 2021; 9:9583-9593. [PMID: 34779469 DOI: 10.1039/d1tb01673c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Conventional biomaterials developed for bone regeneration fail to fully recapitulate the nanoscale structural organization and complex composition of the native bone microenvironment. Therefore, despite promoting osteogenic differentiation of stem cells, they fall short of providing the structural, biochemical, and mechanical stimuli necessary to drive osteogenesis for bone regeneration and function. To address this, we have recently developed a novel strategy to engineer bone-like tissue using a biomimetic approach to achieve rapid and controlled nanoscale mineralization of a cell-laden matrix in the presence of osteopontin, a non-collagenous protein, and a supersaturated solution of calcium and phosphate medium. Here, we build on this approach to engineer bone regeneration scaffolds comprising methacrylated gelatin (GelMA) hydrogels incorporated with calcium citrate core-shell microparticles as a sustained and reliable source of calcium ions for in situ mineralization. We demonstrate successful biomineralization of GelMA hydrogels by embedded calcium carbonate-calcium citrate core-shell microparticles with the resultant mineral chemistry, structure, and organization reminiscent of that of native bone. The biomimetic mineralization was further shown to promote osteogenic differentiation of encapsulated human mesenchymal stem cells even in the absence of other exogenous osteogenic induction factors. Ultimately, by combining the superior biological response engendered by biomimetic mineralization with the intrinsic tissue engineering advantages offered by GelMA, such as biocompatibility, biodegradability, and printability, we envision that our system offers great potential for bone regeneration efforts.
Collapse
Affiliation(s)
- Ramesh Subbiah
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
| | - Gabriela de Souza Balbinot
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Avathamsa Athirasala
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
| | - Fabricio Mezzomo Collares
- Department of Dental Materials, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Grigoriy Sereda
- Department of Chemistry, University of South Dakota, Vermillion, SD 57069, USA.
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA.
| |
Collapse
|
19
|
Lin C, He Y, Feng Q, Xu K, Chen Z, Tao B, Li X, Xia Z, Jiang H, Cai K. Self-renewal or quiescence? Orchestrating the fate of mesenchymal stem cells by matrix viscoelasticity via PI3K/Akt-CDK1 pathway. Biomaterials 2021; 279:121235. [PMID: 34749070 DOI: 10.1016/j.biomaterials.2021.121235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/11/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
To control the fate of mesenchymal stem cells (MSCs) in a 3D environment by adjusting the mechanical parameters of MSC-loading scaffolds, is one of the hot topics in the field of regenerative biomaterials. However, a thorough understanding of the relevant MSCs behaviors affected by viscoelasticity, a dynamic physical parameter of scaffolds, is still lacking. Herein, we established an alginate hydrogel system with constant stiffness and tunable stress relaxation rate, which is a key parameter for the viscoelastic property of material. MSCs were cultured inside three groups of alginate hydrogels with various stress relaxation rates, and then RNA-seq analysis of cells was performed. Results showed that the change of stress relaxation rates of hydrogels regulated the most of the different expression genes of MSCs, which were enriched in cell proliferation-related pathways. MSCs cultured in hydrogels with fast stress relaxation rate presented a high self-renewal proliferation profile via activating phosphatidylinositol 3- kinase (PI3K)/protein kinase B (Akt) pathway. In contrast, a slow stress relaxation rate of hydrogels induced MSCs to enter a reversible quiescence state due to the weakened PI3K/Akt activation. Combined with a further finite element analysis, we speculated that the quiescence of MSCs could be served as a positive strategy for MSCs to deal with the matrix with a low deformation to keep stemness. Based on the results, we identified that stress relaxation rate of hydrogel was a potential physical factor of hydrogel to regulate the self-renewal or quiescence of MSCs. Thus, our findings provide a significant guiding principle for the design of MSCs-encapsulated biomaterials.
Collapse
Affiliation(s)
- Chuanchuan Lin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ye He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, 27708, USA
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kun Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Zhe Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Bailong Tao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xuemin Li
- Innovation Drug Research Centre, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Zengzilu Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Hong Jiang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
20
|
Puech PH, Bongrand P. Mechanotransduction as a major driver of cell behaviour: mechanisms, and relevance to cell organization and future research. Open Biol 2021; 11:210256. [PMID: 34753321 PMCID: PMC8586914 DOI: 10.1098/rsob.210256] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023] Open
Abstract
How do cells process environmental cues to make decisions? This simple question is still generating much experimental and theoretical work, at the border of physics, chemistry and biology, with strong implications in medicine. The purpose of mechanobiology is to understand how biochemical and physical cues are turned into signals through mechanotransduction. Here, we review recent evidence showing that (i) mechanotransduction plays a major role in triggering signalling cascades following cell-neighbourhood interaction; (ii) the cell capacity to continually generate forces, and biomolecule properties to undergo conformational changes in response to piconewton forces, provide a molecular basis for understanding mechanotransduction; and (iii) mechanotransduction shapes the guidance cues retrieved by living cells and the information flow they generate. This includes the temporal and spatial properties of intracellular signalling cascades. In conclusion, it is suggested that the described concepts may provide guidelines to define experimentally accessible parameters to describe cell structure and dynamics, as a prerequisite to take advantage of recent progress in high-throughput data gathering, computer simulation and artificial intelligence, in order to build a workable, hopefully predictive, account of cell signalling networks.
Collapse
Affiliation(s)
- Pierre-Henri Puech
- Lab Adhesion and Inflammation (LAI), Inserm UMR 1067, CNRS UMR 7333, Aix-Marseille Université UM61, Marseille, France
| | - Pierre Bongrand
- Lab Adhesion and Inflammation (LAI), Inserm UMR 1067, CNRS UMR 7333, Aix-Marseille Université UM61, Marseille, France
| |
Collapse
|
21
|
Rogers RE, Haskell A, White BP, Dalal S, Lopez M, Tahan D, Pan S, Kaur G, Kim H, Barreda H, Woodard SL, Benavides OR, Dai J, Zhao Q, Maitland KC, Han A, Nikolov ZL, Liu F, Lee RH, Gregory CA, Kaunas R. A scalable system for generation of mesenchymal stem cells derived from induced pluripotent cells employing bioreactors and degradable microcarriers. Stem Cells Transl Med 2021; 10:1650-1665. [PMID: 34505405 PMCID: PMC8641084 DOI: 10.1002/sctm.21-0151] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are effective in treating disorders resulting from an inflammatory or heightened immune response. The hMSCs derived from induced pluripotent stem cells (ihMSCs) share the characteristics of tissue derived hMSCs but lack challenges associated with limited tissue sources and donor variation. To meet the expected future demand for ihMSCs, there is a need to develop scalable methods for their production at clinical yields while retaining immunomodulatory efficacy. Herein, we describe a platform for the scalable expansion and rapid harvest of ihMSCs with robust immunomodulatory activity using degradable gelatin methacryloyl (GelMA) microcarriers. GelMA microcarriers were rapidly and reproducibly fabricated using a custom microfluidic step emulsification device at relatively low cost. Using vertical wheel bioreactors, 8.8 to 16.3‐fold expansion of ihMSCs was achieved over 8 days. Complete recovery by 5‐minute digestion of the microcarriers with standard cell dissociation reagents resulted in >95% viability. The ihMSCs matched or exceeded immunomodulatory potential in vitro when compared with ihMSCs expanded on monolayers. This is the first description of a robust, scalable, and cost‐effective method for generation of immunomodulatory ihMSCs, representing a significant contribution to their translational potential.
Collapse
Affiliation(s)
- Robert E Rogers
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Andrew Haskell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Berkley P White
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| | - Sujata Dalal
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Megan Lopez
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Daniel Tahan
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Simin Pan
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Gagandeep Kaur
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Hyemee Kim
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Heather Barreda
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Susan L Woodard
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA
| | - Oscar R Benavides
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| | - Jing Dai
- Department of Electrical and Computer Engineering, Texas A&M University, Wisenbaker Engineering Building, College Station, Texas, USA
| | - Qingguo Zhao
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Kristen C Maitland
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| | - Arum Han
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA.,Department of Electrical and Computer Engineering, Texas A&M University, Wisenbaker Engineering Building, College Station, Texas, USA
| | - Zivko L Nikolov
- National Center for Therapeutics Manufacturing, Texas A&M University, College Station, Texas, USA.,Biological and Agricultural Engineering, Texas A&M University, Scoates Hall, College Station, Texas, USA
| | - Fei Liu
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Ryang Hwa Lee
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Carl A Gregory
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, Bryan, Texas, USA
| | - Roland Kaunas
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, College Station, Texas, USA
| |
Collapse
|
22
|
El-Rashidy AA, El Moshy S, Radwan IA, Rady D, Abbass MMS, Dörfer CE, Fawzy El-Sayed KM. Effect of Polymeric Matrix Stiffness on Osteogenic Differentiation of Mesenchymal Stem/Progenitor Cells: Concise Review. Polymers (Basel) 2021; 13:2950. [PMID: 34502988 PMCID: PMC8434088 DOI: 10.3390/polym13172950] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 01/23/2023] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) have a multi-differentiation potential into specialized cell types, with remarkable regenerative and therapeutic results. Several factors could trigger the differentiation of MSCs into specific lineages, among them the biophysical and chemical characteristics of the extracellular matrix (ECM), including its stiffness, composition, topography, and mechanical properties. MSCs can sense and assess the stiffness of extracellular substrates through the process of mechanotransduction. Through this process, the extracellular matrix can govern and direct MSCs' lineage commitment through complex intracellular pathways. Hence, various biomimetic natural and synthetic polymeric matrices of tunable stiffness were developed and further investigated to mimic the MSCs' native tissues. Customizing scaffold materials to mimic cells' natural environment is of utmost importance during the process of tissue engineering. This review aims to highlight the regulatory role of matrix stiffness in directing the osteogenic differentiation of MSCs, addressing how MSCs sense and respond to their ECM, in addition to listing different polymeric biomaterials and methods used to alter their stiffness to dictate MSCs' differentiation towards the osteogenic lineage.
Collapse
Affiliation(s)
- Aiah A. El-Rashidy
- Biomaterials Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt;
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
| | - Sara El Moshy
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Israa Ahmed Radwan
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Dina Rady
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Marwa M. S. Abbass
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
23
|
He C, Wang T, Wang Y, Xu T, Zhao S, Shi H, Zou R. ILK regulates osteogenic differentiation of Human Periodontal Ligament Stem Cells through YAP-mediated Mechanical Memory. Oral Dis 2021; 29:274-284. [PMID: 34370371 DOI: 10.1111/odi.13997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022]
Abstract
Mechanical memory meant the mechanical properties of the matrix could influence the cell fate even after the matrix was changed and has been justified in many kinds of cells. To utilize the phenomenon to improve periodontal tissue engineering, we studied whether mechanical memory existed in human periodontal ligament stem cells and testified if ILK plays a role in this process. The substrate of different stiffness was fabricated by gelatin methacrylate hydrogel. Two groups of hPDLSCs with stiff (St) and soft (So) matrix respectively were cultivated. Then half of the cells exchanged their matrix stiffness in the fourth passage and therefore So, St, So-St and St-So were formed. Morphology of hPDLSCs and intracellular location of YAP was observed via fluorescence staining, osteogenic differentiation of hPDLSCs was assessed by Real-Time PCR, ALP staining and western blot. Then all these were reassessed after the ILK gene had been knocked down. The results showed that morphology and YAP location of hPDLSCs were different between matrix changed and unchanged groups; osteogenic genes expression, ALP staining and western blot also varied. After the ILK gene had been knocked down, the YAP location and osteogenic activity of hPDLSCs were significantly influenced. Thus, it could be concluded that mechanical memory exists in hPDLSCs; ILK is involved in this process.
Collapse
Affiliation(s)
- Chuan He
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Tairan Wang
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Yijie Wang
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Tongtong Xu
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Shuyang Zhao
- School of medicine, Xi'an Jiaotong University, Xi'an, China
| | - Haoyu Shi
- School of medicine, Xi'an Jiaotong University, Xi'an, China
| | - Rui Zou
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
Stiffness Regulates the Morphology, Adhesion, Proliferation, and Osteogenic Differentiation of Maxillary Schneiderian Sinus Membrane-Derived Stem Cells. Stem Cells Int 2021; 2021:8868004. [PMID: 34306097 PMCID: PMC8285206 DOI: 10.1155/2021/8868004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 05/06/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022] Open
Abstract
Recent studies, which aim to optimize maxillary sinus augmentation, have paid significant attention exploring osteogenic potential of maxillary Schneiderian sinus membrane-derived cells (MSSM-derived cells). However, it remains unclear that how MSSM-derived cells could respond to niche's biomechanical properties. Herein, this study investigated the possible effects of substrate stiffness on rMSSM-derived stem cell fate. Initially, rMSSM-derived stem cells with multiple differentiation potential were successfully obtained. We then fabricated polyacrylamide substrates with varied stiffness ranging from 13 to 68 kPa to modulate the mechanical environment of rMSSM-derived stem cells. A larger cell spreading area and increased proliferation of rMSSM-derived stem cells were found on the stiffer substrates. Similarly, cells became more adhesive as their stiffness increased. Furthermore, the higher stiffness facilitated osteogenic differentiation of rMSSM-derived stem cells. Overall, our results indicated that increase in stiffness could mediate behaviors of rMSSM-derived stem cells, which may serve as a guide in future research to design novel biomaterials for maxillary sinus augmentation.
Collapse
|
25
|
Yin S, Cao Y. Hydrogels for Large-Scale Expansion of Stem Cells. Acta Biomater 2021; 128:1-20. [PMID: 33746032 DOI: 10.1016/j.actbio.2021.03.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
Stem cells demonstrate considerable promise for various preclinical and clinical applications, including drug screening, disease treatments, and regenerative medicine. Producing high-quality and large amounts of stem cells is in demand for these applications. Despite challenges, as hydrogel-based cell culture technology has developed, tremendous progress has been made in stem cell expansion and directed differentiation. Hydrogels are soft materials with abundant water. Many hydrogel properties, including biodegradability, mechanical strength, and porosity, have been shown to play essential roles in regulating stem cell proliferation and differentiation. The biochemical and physical properties of hydrogels can be specifically tailored to mimic the native microenvironment that various stem cells reside in vivo. A few hydrogel-based systems have been developed for successful stem cell cultures and expansion in vitro. In this review, we summarize various types of hydrogels that have been designed to effectively enhance the proliferation of hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs), respectively. According to each stem cell type's preference, we also discuss strategies for fabricating hydrogels with biochemical and mechanical cues and other characteristics representing microenvironments of stem cells in vivo. STATEMENT OF SIGNIFICANCE: In this review article we summarize current progress on the construction of hydrogel systems for the culture and expansion of various stem cells, including hematopoietic stem cells (HSCs), mesenchymal stem/stromal cells (MSCs), and pluripotent stem cells (PSCs). The Significance includes: (1) Provide detailed discussion on the stem cell niches that should be considered for stem cell in vitro expansion. (2) Summarize various strategies to construct hydrogels that can largely recapture the microenvironment of native stem cells. (3) Suggest a few future directions that can be implemented to improve current in vitro stem cell expansion systems.
Collapse
Affiliation(s)
- Sheng Yin
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057
| | - Yi Cao
- National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China; Chemistry and Biomedicine innovation center, Nanjing University, Nanjing, 210093, China; Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China; Shenzhen Research Institute of Nanjing University, Shenzhen, China, 518057.
| |
Collapse
|
26
|
Osmond MJ, Krebs MD. Tunable chitosan-calcium phosphate composites as cell-instructive dental pulp capping agents. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1450-1465. [PMID: 33941040 DOI: 10.1080/09205063.2021.1925390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Dental cavities are the most prevalent, preventable disease worldwide providing a need for robust treatment options to restore both the form and function of decaying teeth. Here is a presentation of a possible regenerative pulp capping agent that can serve to restore tooth function while regenerating healthy dentin tissue over a long period of time. To achieve this goal a material needs to crosslink quickly, be structurally rigid, and support the proliferation and differentiation of stem cells contained within the dental pulp. In this study, calcium phosphate nanoparticles were embedded in polymer hydrogels of carboxymethyl-chitosan and a diglycidyl ether. The particle size, surface, and mechanical properties of these materials were characterized. These composites have moduli up to 3 MPa, support the culture of dental pulp stem cells more than 3 weeks and exhibit osteogenic potential even without osteogenic media. These composites demonstrate a promising potential as the next generation of pulp capping materials.
Collapse
Affiliation(s)
- Matthew J Osmond
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO, USA
| | - Melissa D Krebs
- Department of Chemical & Biological Engineering, Colorado School of Mines, Golden, CO, USA
| |
Collapse
|
27
|
Mubarok W, Qu Y, Sakai S. Influence of Hydrogen Peroxide-Mediated Cross-Linking and Degradation on Cell-Adhesive Gelatin Hydrogels. ACS APPLIED BIO MATERIALS 2021; 4:4184-4190. [PMID: 35006831 DOI: 10.1021/acsabm.0c01675] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Hydrogen peroxide (H2O2) is widely used for the gelation of aqueous solutions of gelatin derivatives with phenolic hydroxyl groups (Gelatin-Ph) catalyzed by horseradish peroxidase (HRP). Apart from this, H2O2 is known to cause degradation/depolymerization of various polymers. Here, we prepared Gelatin-Ph hydrogels from solutions containing Gelatin-Ph and HRP by continuously supplying H2O2 from the gas phase and investigated the mechanical properties of resultant hydrogels and the behaviors of rat fibroblast and human adipose-derived stem cells on them. Young's modulus of the hydrogel obtained from 5 w/v % Gelatin-Ph and 1 and 5 U/mL HRP increased when the exposure time to air containing H2O2 (16 ppm) was extended from 15 to 30 min. However, further prolonging the exposure time to 60 min reduced Young's modulus to the same magnitude as for the hydrogels exposed to air containing H2O2 for 15 min. Interestingly, the cell length and aspect ratio of the cells continued to increase, as the exposure time was extended, without reflecting the decrease in Young's modulus. These results indicate that when preparing Gelatin-Ph hydrogels through HRP/H2O2-mediated gelation, it is necessary to consider the effect of the degradation of Gelatin-Ph caused by H2O2 on the mechanical properties of the resultant hydrogels and the behaviors of cells on them.
Collapse
Affiliation(s)
- Wildan Mubarok
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| | - Yanfei Qu
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| | - Shinji Sakai
- Division of Chemical Engineering, Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, Osaka 560-8531, Japan
| |
Collapse
|
28
|
Modulating the physico-mechanical properties of polyacrylamide/gelatin hydrogels for tissue engineering application. Polym Bull (Berl) 2021. [DOI: 10.1007/s00289-021-03592-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
29
|
Wang H, Xu X. Continuum elastic models for force transmission in biopolymer gels. SOFT MATTER 2020; 16:10781-10808. [PMID: 33289764 DOI: 10.1039/d0sm01451f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We review continuum elastic models for the transmission of both external forces and internal active cellular forces in biopolymer gels, and relate them to recent experiments. Rather than being exhaustive, we focus on continuum elastic models for small affine deformations and intend to provide a systematic continuum method and some analytical perspectives on the study of force transmission in biopolymer gels. We start from a very brief review of the nonlinear mechanics of individual biopolymers and a summary of constitutive models for the nonlinear elasticity of biopolymer gels. We next show that the simple 3-chain model can give predictions that fit well the shear experiments of some biopolymer gels, including the effects of strain-stiffening and negative normal stress. We then review continuum models for the transmission of internal active forces that are induced by a spherically contracting cell embedded in a three-dimensional biopolymer gel. Various scaling regimes for the decay of cell-induced displacements are identified for linear isotropic and anisotropic materials, and for biopolymer gels with nonlinear compressive-softening and strain-stiffening elasticity, respectively. After that, we present (using an energetic approach) the generic and unified continuum theory proposed in [D. Ben-Yaakov et al., Soft Matter, 2015, 11, 1412] about how the transmission of forces in the biogel matrix can mediate long-range interactions between cells with mechanical homeostasis. We show the predictions of the theory in a special hexagonal multicellular array, and relate them to recent experiments. Finally, we conclude this paper with comments on the limitations and outlook of continuum modeling, and highlight the need for complementary theoretical approaches, such as discrete network simulations, to force transmission in biopolymer gels and phenomenological active gel theories for multicellular systems.
Collapse
Affiliation(s)
- Haiqin Wang
- Technion - Israel Institute of Technology, Haifa, 32000, Israel.
| | | |
Collapse
|
30
|
Sun Y, Liu J, Xu Z, Lin X, Zhang X, Li L, Li Y. Matrix stiffness regulates myocardial differentiation of human umbilical cord mesenchymal stem cells. Aging (Albany NY) 2020; 13:2231-2250. [PMID: 33318310 PMCID: PMC7880396 DOI: 10.18632/aging.202244] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/20/2020] [Indexed: 05/07/2023]
Abstract
Myocardial infarction is a cardiovascular disease with high mortality. Human umbilical cord mesenchymal stem cells (hUC-MSCs) with strong self-renewal capacity and multipotency, provide the possibility of replacing injured cardiomyocytes. hUC-MSCs were cultured on polyacrylamide hydrogels with stiffnesses corresponding to Young's modulus of 13-16kPa and 62-68kPa which mimic the stiffnesses of healthy heart tissue and fibrotic myocardium. The expression of early myocardial markers Nkx2.5, GATA4, Mesp1 and the mature myocardial markers cTnT, cTnI, α-actin were detected by RT-PCR and Western Blot, which showed that soft matrix (13-16 kPa) tended to induce the differentiation of hUC-MSCs into myocardium, compared with stiff matrix (62-68 kPa). Piezos are mechanically sensitive non-selective cation channels. The expression of Piezo1 increased with the stiffness gradient of 1-10kPa, 13-16kPa, 35-38kPa and 62-68kPa on the 1st day, but Piezo2 expression was irregular. The expression of integrin β1 and calcium ions were also higher on stiff substrate than on soft substrate. hUC-MSCs tend to differentiate into myocardium on the matrix stiffness of 13-16 kPa. The relationship among matrix stiffness, Piezo1 and myocardial differentiation needs further validation.
Collapse
Affiliation(s)
- Yingying Sun
- Department of Stomatology, The First Hospital of Jilin University, Jilin University, Changchun, China
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jingwei Liu
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoxuan Lin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
31
|
Ejeian F, Razmjou A, Nasr-Esfahani MH, Mohammad M, Karamali F, Ebrahimi Warkiani M, Asadnia M, Chen V. ZIF-8 Modified Polypropylene Membrane: A Biomimetic Cell Culture Platform with a View to the Improvement of Guided Bone Regeneration. Int J Nanomedicine 2020; 15:10029-10043. [PMID: 33335393 PMCID: PMC7737945 DOI: 10.2147/ijn.s269169] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Despite the significant advances in modeling of biomechanical aspects of cell microenvironment, it remains a major challenge to precisely mimic the physiological condition of the particular cell niche. Here, the metal-organic frameworks (MOFs) have been introduced as a feasible platform for multifactorial control of cell-substrate interaction, given the wide range of physical and mechanical properties of MOF materials and their structural flexibility. RESULTS In situ crystallization of zeolitic imidazolate framework-8 (ZIF-8) on the polydopamine (PDA)-modified membrane significantly raised surface energy, wettability, roughness, and stiffness of the substrate. This modulation led to an almost twofold increment in the primary attachment of dental pulp stem cells (DPSCs) compare to conventional plastic culture dishes. The findings indicate that polypropylene (PP) membrane modified by PDA/ZIF-8 coating effectively supports the growth and proliferation of DPSCs at a substantial rate. Further analysis also displayed the exaggerated multilineage differentiation of DPSCs with amplified level of autocrine cell fate determination signals, like BSP1, BMP2, PPARG, FABP4, ACAN, and COL2A. Notably, osteogenic markers were dramatically overexpressed (more than 100-folds rather than tissue culture plate) in response to biomechanical characteristics of the ZIF-8 layer. CONCLUSION Hence, surface modification of cell culture platforms with MOF nanostructures proposed as a powerful nanomedical approach for selectively guiding stem cells for tissue regeneration. In particular, PP/PDA/ZIF-8 membrane presented ideal characteristics for using as a barrier membrane for guided bone regeneration (GBR) in periodontal tissue engineering.
Collapse
Affiliation(s)
- Fatemeh Ejeian
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan73441-81746, Iran
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Amir Razmjou
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan73441-81746, Iran
- UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Munirah Mohammad
- UNESCO Center for Membrane Technology, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Mohsen Asadnia
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia
| | - Vicki Chen
- School of Chemical Engineering, University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
32
|
3D Bioprinting of Human Adipose-Derived Stem Cells and Their Tenogenic Differentiation in Clinical-Grade Medium. Int J Mol Sci 2020; 21:ijms21228694. [PMID: 33218011 PMCID: PMC7698777 DOI: 10.3390/ijms21228694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Defining the best combination of cells and biomaterials is a key challenge for the development of tendon tissue engineering (TE) strategies. Adipose-derived stem cells (ASCs) are ideal candidates for this purpose. In addition, controlled cell-based products adherent to good manufacturing practice (GMP) are required for their clinical scale-up. With this aim, in this study, ASC 3D bioprinting and GMP-compliant tenogenic differentiation were investigated. In detail, primary human ASCs were embedded within a nanofibrillar-cellulose/alginate bioink and 3D-bioprinted into multi-layered square-grid matrices. Bioink viscoelastic properties and scaffold ultrastructural morphology were analyzed by rheology and scanning electron microscopy (SEM). The optimal cell concentration for printing among 3, 6 and 9 × 106 ASC/mL was evaluated in terms of cell viability. ASC morphology was characterized by SEM and F-actin immunostaining. Tenogenic differentiation ability was then evaluated in terms of cell viability, morphology and expression of scleraxis and collagen type III by biochemical induction using BMP-12, TGF-β3, CTGF and ascorbic acid supplementation (TENO). Pro-inflammatory cytokine release was also assessed. Bioprinted ASCs showed high viability and survival and exhibited a tenocyte-like phenotype after biochemical induction, with no inflammatory response to the bioink. In conclusion, we report a first proof of concept for the clinical scale-up of ASC 3D bioprinting for tendon TE.
Collapse
|
33
|
Han S, Kim J, Lee G, Kim D. Mechanical Properties of Materials for Stem Cell Differentiation. ACTA ACUST UNITED AC 2020; 4:e2000247. [DOI: 10.1002/adbi.202000247] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Seong‐Beom Han
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Jeong‐Ki Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Geonhui Lee
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| | - Dong‐Hwee Kim
- KU‐KIST Graduate School of Converging Science and Technology Korea University 145, Anam‐ro, Seongbuk‐gu Seoul 02841 Republic of Korea
| |
Collapse
|
34
|
Ruan D, Wu C, Deng S, Zhang Y, Guan G. The Anatase Phase of Nanotopography Titania with Higher Roughness Has Better Biocompatibility in Osteoblast Cell Morphology and Proliferation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8032718. [PMID: 33029524 PMCID: PMC7527892 DOI: 10.1155/2020/8032718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/01/2020] [Accepted: 09/11/2020] [Indexed: 01/19/2023]
Abstract
Previous studies have concluded that surface-modified titanium oxide (titania, TiO2) surface properties promote osteoblast cell morphology and proliferation. To screen a suitable structured titania coating with the best biocompatibility to be used in dental implants, five titania films (two amorphous, one rutile, and two anatases) with different surfaces were successfully synthesized on polished titanium by radio frequency (RF) magnetron sputtering. We applied atomic force microscopy (AFM) and X-ray diffraction (XRD) to depict the formulations. Furthermore, MC3T3-E1, the mouse osteoblast precursor cell, was used to assess cell proliferation and observe morphologic changes at the film surface. The data indicated that the overall number of MC3T3-E1 cells on anatase films was significantly higher as compared with cells on rutile and amorphous films. Meanwhile, the actin filaments of the cells grown on the anatase phase films were well defined and fully spread. In addition, the film with higher roughness had enhanced biocompatibility than that with lower roughness. The results showed that the crystal phase and titania coated roughness had a greater influence on the biocompatibility of nanostructured titania film. The higher the roughness of the anatase phase was, the better bioactivity for the morphology and proliferation of osteoblast. This is a good surface-modified biological material and may have a good application prospect in dental implants.
Collapse
Affiliation(s)
- Danping Ruan
- Minhang Branch, Zhongshan Hospital, Fudan University, China
| | - Chunyun Wu
- Minhang Branch, Zhongshan Hospital, Fudan University, China
| | - Sinan Deng
- Minhang Branch, Zhongshan Hospital, Fudan University, China
| | - Yu Zhang
- Minhang Branch, Zhongshan Hospital, Fudan University, China
| | - Guoling Guan
- Minhang Branch, Zhongshan Hospital, Fudan University, China
| |
Collapse
|
35
|
Sun Y, Xu Z, Wang M, Lv S, Wu H, Chi G, Li L, Li Y. Soft Matrix Combined With BMPR Inhibition Regulates Neurogenic Differentiation of Human Umbilical Cord Mesenchymal Stem Cells. Front Bioeng Biotechnol 2020; 8:791. [PMID: 32760710 PMCID: PMC7372119 DOI: 10.3389/fbioe.2020.00791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/22/2020] [Indexed: 11/25/2022] Open
Abstract
Stem cells constantly encounter as well as respond to a variety of signals in their microenvironment. Although the role of biochemical factors has always been emphasized, the significance of biophysical signals has not been studied until recently. Additionally, biophysical elements, like extracellular matrix (ECM) stiffness, can regulate functions of stem cells. In this study, we demonstrated that soft matrix with 1-10 kPa can induce neural differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs). Importantly, we used a combination of soft matrix and bone morphogenetic protein receptor (BMPR) inhibition to promote neurogenic differentiation of hUC-MSCs. Furthermore, BMPR/SMADs occurs in crosstalk with the integrinβ1 downstream signaling pathway. In addition, BMPR inhibition plays a positive role in maintaining the undifferentiated state of hUC-MSCs on the hydrogel substrate. The results provide further evidence for the molecular mechanisms via which stem cells convert mechanical inputs into fateful decisions.
Collapse
Affiliation(s)
- Yingying Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
- Department of Stomatology, First Hospital of Jilin University, Changchun, China
| | - Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Meijing Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Haitao Wu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
36
|
Rahmati M, Silva EA, Reseland JE, A Heyward C, Haugen HJ. Biological responses to physicochemical properties of biomaterial surface. Chem Soc Rev 2020; 49:5178-5224. [PMID: 32642749 DOI: 10.1039/d0cs00103a] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Biomedical scientists use chemistry-driven processes found in nature as an inspiration to design biomaterials as promising diagnostic tools, therapeutic solutions, or tissue substitutes. While substantial consideration is devoted to the design and validation of biomaterials, the nature of their interactions with the surrounding biological microenvironment is commonly neglected. This gap of knowledge could be owing to our poor understanding of biochemical signaling pathways, lack of reliable techniques for designing biomaterials with optimal physicochemical properties, and/or poor stability of biomaterial properties after implantation. The success of host responses to biomaterials, known as biocompatibility, depends on chemical principles as the root of both cell signaling pathways in the body and how the biomaterial surface is designed. Most of the current review papers have discussed chemical engineering and biological principles of designing biomaterials as separate topics, which has resulted in neglecting the main role of chemistry in this field. In this review, we discuss biocompatibility in the context of chemistry, what it is and how to assess it, while describing contributions from both biochemical cues and biomaterials as well as the means of harmonizing them. We address both biochemical signal-transduction pathways and engineering principles of designing a biomaterial with an emphasis on its surface physicochemistry. As we aim to show the role of chemistry in the crosstalk between the surface physicochemical properties and body responses, we concisely highlight the main biochemical signal-transduction pathways involved in the biocompatibility complex. Finally, we discuss the progress and challenges associated with the current strategies used for improving the chemical and physical interactions between cells and biomaterial surface.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway. h.j.haugen.odont.uio.no
| | | | | | | | | |
Collapse
|
37
|
Wang L, Dong S, Liu Y, Ma Y, Zhang J, Yang Z, Jiang W, Yuan Y. Fabrication of Injectable, Porous Hyaluronic Acid Hydrogel Based on an In-Situ Bubble-Forming Hydrogel Entrapment Process. Polymers (Basel) 2020; 12:E1138. [PMID: 32429363 PMCID: PMC7284757 DOI: 10.3390/polym12051138] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 01/07/2023] Open
Abstract
Injectable hydrogels have been widely applied in the field of regenerative medicine. However, current techniques for injectable hydrogels are facing a challenge when trying to generate a biomimetic, porous architecture that is well-acknowledged to facilitate cell behaviors. In this study, an injectable, interconnected, porous hyaluronic acid (HA) hydrogel based on an in-situ bubble self-generation and entrapment process was developed. Through an amide reaction between HA and cystamine dihydrochloride activated by EDC/NHS, CO2 bubbles were generated and were subsequently entrapped inside the substrate due to a rapid gelation-induced retention effect. HA hydrogels with different molecular weights and concentrations were prepared and the effects of the hydrogel precursor solution's concentration and viscosity on the properties of hydrogels were investigated. The results showed that HA10-10 (10 wt.%, MW 100,000 Da) and HA20-2.5 (2.5 wt.%, MW 200,000 Da) exhibited desirable gelation and obvious porous structure. Moreover, HA10-10 represented a high elastic modulus (32 kPa). According to the further in vitro and in vivo studies, all the hydrogels prepared in this study show favorable biocompatibility for desirable cell behaviors and mild host response. Overall, such an in-situ hydrogel with a self-forming bubble and entrapment strategy is believed to provide a robust and versatile platform to engineer injectable hydrogels for a variety of applications in tissue engineering, regenerative medicine, and personalized therapeutics.
Collapse
Affiliation(s)
- Lixuan Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China; (L.W.); (Y.L.)
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Yutong Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China; (L.W.); (Y.L.)
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Yifan Ma
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; (Y.M.); (J.Z.)
| | - Jingjing Zhang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; (Y.M.); (J.Z.)
| | - Zhaogang Yang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education, and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China; (L.W.); (Y.L.)
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
38
|
Li ZL, Wang ZJ, Wei GH, Yang Y, Wang XW. Changes in extracellular matrix in different stages of colorectal cancer and their effects on proliferation of cancer cells. World J Gastrointest Oncol 2020; 12:267-275. [PMID: 32206177 PMCID: PMC7081112 DOI: 10.4251/wjgo.v12.i3.267] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/12/2020] [Accepted: 02/07/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The extracellular matrix is the main component of the tumor microenvironment. Extracellular matrix remodels with the oncogenesis and development of tumors. Previous studies usually focused on the changes of proteins in normal colorectal tissues and colorectal cancers. Little is known about the changes in the extracellular matrix in different stages of colorectal cancer and the effects of these changes on the development of this cancer. AIM To test the changes of type I collagen, type IV collagen, matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), and tissue inhibitor of metalloproteinase-3 (TIMP-3) in different stages of colorectal cancer and the effects of these changes on the proliferation of cancer cells. METHODS The extracellular matrix from various stages of colorectal cancer and normal colon tissue was obtained by using acellular technology. We used proteomics to detect the differential expression of proteins between normal colon tissues and colorectal cancer tissues, and then we used Western blot to observe their expression in each stage of colorectal cancer and in normal colon tissue. By co-culturing the extracellular matrix and HT29 colon cancer cells in vivo and in vitro, we tested the cancer cell proliferation rate in vitro by methyl thiazolyl tetrazolium (MTT) assay and in vivo by measuring the tumor volume. RESULTS The expression of type I collagen and MMP-2 increased with increased tumor stage. The expression of MMP-9 was higher in colorectal cancer tissues and was highest in stage III cancer. The expression of type IV collagen and TIMP-3 decreased with increased tumor stage. The proliferation rate of cancer cells in the extracellular matrix of colorectal cancer was higher than that in the extracellular matrix of the normal colon. CONCLUSION These data suggest that the extracellular matrix structure and composition become disorganized during the development of tumors, which is more conducive for the growth of cancer cells.
Collapse
Affiliation(s)
- Zhu-Lin Li
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhen-Jun Wang
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Guang-Hui Wei
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yong Yang
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiao-Wan Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
39
|
Bai M, Cai L, Li X, Ye L, Xie J. Stiffness and topography of biomaterials dictate cell-matrix interaction in musculoskeletal cells at the bio-interface: A concise progress review. J Biomed Mater Res B Appl Biomater 2020; 108:2426-2440. [PMID: 32027091 DOI: 10.1002/jbm.b.34575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/30/2019] [Accepted: 01/19/2020] [Indexed: 02/05/2023]
Abstract
Mutually interacted musculoskeletal tissues work together within the physiological environment full of varieties of external stimulus. Consistent with the locomotive function of the tissues, musculoskeletal cells are remarkably mechanosensitive to the physical cues. Signals like extracellular matrix (ECM) stiffness, topography, and geometry can be sensed and transduced into intracellular signaling cascades to trigger a series of cell responses, including cell adhesion, cell phenotype maintenance, cytoskeletal reconstruction, and stem cell differentiation (Du et al., 2011; Murphy et al., 2014; Lv et al., 2015; Kim et al., 2016; Kumar et al., 2017). With the development of tissue engineering and regenerative medicine, the potent effects of ECM physical properties on cell behaviors at the cell-matrix interface are drawing much attention. To mimic the interaction between cell and its ECM physical properties, developing advanced biomaterials with desired characteristics which could achieve the biointerface between cells and the surrounded matrix close to the physiological conditions becomes a great hotspot. In this review, based on the current publications in the field of biointerfaces, we systematically summarized the significant roles of stiffness and topography on musculoskeletal cell behaviors. We hope to shed light on the importance of physical cues in musculoskeletal tissue engineering and provide up to date strategies towards the natural or artificial replication of physiological microenvironment.
Collapse
Affiliation(s)
- Mingru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Elastic Anisotropy Governs the Range of Cell-Induced Displacements. Biophys J 2020; 118:1152-1164. [PMID: 31995739 DOI: 10.1016/j.bpj.2019.12.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/22/2019] [Accepted: 12/27/2019] [Indexed: 12/20/2022] Open
Abstract
The unique nonlinear mechanics of the fibrous extracellular matrix (ECM) facilitates long-range cell-cell mechanical communications that would be impossible for linear elastic substrates. Past research has described the contribution of two separated effects on the range of force transmission, including ECM elastic nonlinearity and fiber alignment. However, the relation between these different effects is unclear, and how they combine to dictate force transmission range is still elusive. Here, we combine discrete fiber simulations with continuum modeling to study the decay of displacements induced by a contractile cell in fibrous networks. We demonstrate that fiber nonlinearity and fiber reorientation both contribute to the strain-induced elastic anisotropy of the cell's local environment. This elastic anisotropy is a "lumped" parameter that governs the slow decay of displacements, and it depends on the magnitude of applied strain, either an external tension or an internal contraction, as a model of the cell. Furthermore, we show that accounting for artificially prescribed elastic anisotropy dictates the decay of displacements induced by a contracting cell. Our findings unify previous single effects into a mechanical theory that explains force transmission in fibrous networks. This work may provide insights into biological processes that involve communication of distant cells mediated by the ECM, such as those occurring in morphogenesis, wound healing, angiogenesis, and cancer metastasis. It may also provide design parameters for biomaterials to control force transmission between cells as a way to guide morphogenesis in tissue engineering.
Collapse
|
41
|
Yang L, Zhai Y, Hao Y, Zhu Z, Cheng G. The Regulatory Functionality of Exosomes Derived from hUMSCs in 3D Culture for Alzheimer's Disease Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906273. [PMID: 31840420 DOI: 10.1002/smll.201906273] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Indexed: 06/10/2023]
Abstract
Reducing amyloid-β (Aβ) accumulation could be a potential therapeutic approach for Alzheimer's disease (AD). Particular functional biomolecules in exosomes vested by the microenvironment in which the original cells resided can be transferred to recipient cells to improve pathological conditions. However, there are few reports addressing whether exosomes derived from cells cultured on scaffolds with varying dimension can reduce Aβ deposition or ameliorate cognitive decline for AD therapy. Herein, both 3D graphene scaffold and 2D graphene film are used as the matrix for human umbilical cord mesenchymal stem cell culture, from which the supernatants are obtained to isolate exosomes. The levels of 195 kinds of miRNAs and proteins, including neprilysin, insulin-degrading enzyme and heat shock protein 70, in 3D-cultured exosomes (3D-Exo) are dramatically different from those obtained from 2D culture. Hence, 3D-Exo could up-regulate the expression of α-secretase and down-regulate the β-secretase to reduce Aβ production in both AD pathology cells and transgenic mice, through their special cargo. With rescuing Aβ pathology, 3D-Exo exerts enhanced therapeutic effects on ameliorating the memory and cognitive deficits in AD mice. These findings provide a novel clinical application for scaffold materials and functional exosomes derived from stem cells.
Collapse
Affiliation(s)
- Lingyan Yang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Yuanxin Zhai
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Ying Hao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| | - Zhanchi Zhu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Guosheng Cheng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, Anhui, 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
42
|
Zhong J, Yang Y, Liao L, Zhang C. Matrix stiffness-regulated cellular functions under different dimensionalities. Biomater Sci 2020; 8:2734-2755. [DOI: 10.1039/c9bm01809c] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The microenvironments that cells encounter with in vitro.
Collapse
Affiliation(s)
- Jiajun Zhong
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yuexiong Yang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Liqiong Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Biomaterials Research Center
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
| | - Chao Zhang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| |
Collapse
|
43
|
Alabi BR, LaRanger R, Shay JW. Decellularized mice colons as models to study the contribution of the extracellular matrix to cell behavior and colon cancer progression. Acta Biomater 2019; 100:213-222. [PMID: 31562987 DOI: 10.1016/j.actbio.2019.09.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/11/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022]
Abstract
Current 3D culture models to study colorectal cancer lack architectural support and signaling proteins provided by the tissue extracellular matrix (ECM) which may influence cell behavior and cancer progression. Therefore, the ability to study cancer cells in the context of a matrix that is physiologically more relevant and to understand how the ECM affects cancer progression has been understudied. To address this, we developed an ex-vivo 3D system, provided by intact wild type (WT) and colon cancer susceptible decellularized mouse colons (DMC), to support the growth of human cancer cells. DMC are free of viable cells but still contain extracellular matrix proteins including subsets of collagens. Stiffness, an important mechanical property, is also maintained in DMCs. Importantly, we observed that the DMC is permissive for cell proliferation and differentiation of a human colon cancer cell line (HT-29). Notably, the ability of cells in the WT DMC to differentiate was also greater when compared to Matrigel™, an extracellular matrix extract from a mouse tumor cell line. Additionally, we observed in invasion assays that DMC obtained from polyps from a colon cancer susceptible mouse model facilitated increased cell migration/invasion of colorectal cancer cells and immortalized non-tumor colonic epithelial cells compared to DMC from WT mice. Finally, using mass spectrometry, we identified extracellular matrix proteins that are more abundant in DMC from a colorectal cancer mouse model compared to age and sex-matched WT mice. We propose that these abundantly expressed proteins in the tumor microenvironment are potentially involved in colorectal cancer progression. STATEMENT OF SIGNIFICANCE: Decellularized matrices, when properly produced, are attractive biomaterials for tissue regeneration and replacement. We show here that the mouse decellularized matrices can also be repurposed to elucidate how the extracellular matrix influences human cell behavior and cancer progression. To do this we produce decellularized matrices, from mice colonic tissue, that have preserved tissue mechanical and structural properties. We demonstrate that the matrix better supports the differentiation of HT-29 cells, a colonic cancer cell line, compared to Matrigel™. Additionally, we show that the extracellular matrix contributes to colon cancer progression via invasion assays using extracellular matrix extracts. Finally, we use mass spectrometry to identify ECM proteins that are more abundant in colonic polyps compared to adjacent tissue regions. This model system may have therapeutic implications for colorectal cancer patients.
Collapse
Affiliation(s)
- Busola R Alabi
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, United States
| | - Ryan LaRanger
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, United States
| | - Jerry W Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, United States.
| |
Collapse
|
44
|
Xu Z, Orkwis JA, DeVine BM, Harris GM. Extracellular matrix cues modulate Schwann cell morphology, proliferation, and protein expression. J Tissue Eng Regen Med 2019; 14:229-242. [PMID: 31702874 DOI: 10.1002/term.2987] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 09/17/2019] [Accepted: 10/17/2019] [Indexed: 01/07/2023]
Abstract
Peripheral nerve injuries require a complex set of signals from cells, macrophages, and the extracellular matrix (ECM) to induce regeneration across injury sites and achieve functional recovery. Schwann cells (SCs), the major glial cell in the peripheral nervous system (PNS), are critical to nerve regeneration due to their inherent capacity for altering phenotype postinjury to facilitate wound healing. The ECM plays a vital role in wound healing as well as regulating cell phenotype during tissue repair. To examine the underlying mechanisms between the ECM and SCs, this work sought to determine how specific ECM cues regulate the phenotype of SCs. To address this, SCs were cultured on polydimethylsiloxane substrates of a variable Young's modulus coated with ECM proteins. Cells were analyzed for spreading area, proliferation, cell and nuclear shape, and c-Jun expression. It was found that substrates with a stiffness of 8.67 kPa coated with laminin promoted the highest expression of c-Jun, a marker signifying a "regenerative" SC. Microcontact printed, cell adhesive areas were then utilized to precisely control the geometry and spreading of SCs and by controlling spreading area and cellular elongation; expression of c-Jun was either promoted or downregulated. These results begin to address the significant interplay between ECM cues and phenotype of SCs, while offering a potential means to enhance PNS regeneration through cellular therapies.
Collapse
Affiliation(s)
- Zhenyuan Xu
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Jacob A Orkwis
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Braden M DeVine
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| | - Greg M Harris
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, Ohio.,Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
45
|
Moghaddam MM, Bonakdar S, Shariatpanahi MR, Shokrgozar MA, Faghihi S. The Effect of Physical Cues on the Stem Cell Differentiation. Curr Stem Cell Res Ther 2019; 14:268-277. [DOI: 10.2174/1574888x14666181227120706] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/05/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022]
Abstract
Development of multicellular organisms is a very complex and organized process during which cells respond to various factors and features in extracellular environments. It has been demonstrated that during embryonic evolvement, under certain physiological or experimental conditions, unspecialized cells or stem cells can be induced to become tissue or organ-specific cells with special functions. Considering the importance of physical cues in stem cell fate, the present study reviews the role of physical factors in stem cells differentiation and discusses the molecular mechanisms associated with these factors.
Collapse
Affiliation(s)
- Mehrdad M. Moghaddam
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, 14965/161, Iran
| | - Shahin Bonakdar
- National Cell Bank, Pasteur Institute of Iran, Tehran 3159915111, Iran
| | | | | | - Shahab Faghihi
- Stem Cell and Regenerative Medicine Group, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, 14965/161, Iran
| |
Collapse
|
46
|
Speer JE, Gunasekara DB, Wang Y, Fallon JK, Attayek PJ, Smith PC, Sims CE, Allbritton NL. Molecular transport through primary human small intestinal monolayers by culture on a collagen scaffold with a gradient of chemical cross-linking. J Biol Eng 2019; 13:36. [PMID: 31061676 PMCID: PMC6487070 DOI: 10.1186/s13036-019-0165-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The luminal surface of the small intestine is composed of a monolayer of cells overlying a lamina propria comprised of extracellular matrix (ECM) proteins. The ECM provides a porous substrate critical for nutrient exchange and cellular adhesion. The enterocytes within the epithelial monolayer possess proteins such as transporters, carriers, pumps and channels that participate in the movement of drugs, metabolites, ions and amino acids and whose function can be regulated or altered by the properties of the ECM. Here, we characterized expression and function of proteins involved in transport across the human small intestinal epithelium grown on two different culture platforms. One strategy employs a conventional scaffolding method comprised of a thin ECM film overlaying a porous membrane while the other utilizes a thick ECM hydrogel placed on a porous membrane. The thick hydrogel possesses a gradient of chemical cross-linking along its length to provide a softer substrate than that of the ECM film-coated membrane while maintaining mechanical stability. RESULTS The monolayers on both platforms possessed goblet cells and abundant enterocytes and were impermeable to Lucifer yellow and fluorescein-dextran (70 kD) indicating high barrier integrity. Multiple transporter proteins were present in both primary-cell culture formats at levels similar to those present in freshly isolated crypts/villi; however, expression of breast cancer resistance protein (BCRP) and multidrug resistance protein 2 (MRP2) in the monolayers on the conventional scaffold was substantially less than that on the gradient cross-linked scaffold and freshly isolated crypts/villi. Monolayers on the conventional scaffold failed to transport the BCRP substrate prazosin while cells on the gradient cross-linked scaffold successfully transported this drug to better mimic the properties of in vivo small intestine. CONCLUSIONS The results of this comparison highlight the need to create in vitro intestinal transport platforms whose characteristics mimic the in vivo lamina propria in order to accurately recapitulate epithelial function. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Jennifer E. Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Dulan B. Gunasekara
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Peter J. Attayek
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| |
Collapse
|
47
|
Mattioli-Belmonte M, Montemurro F, Licini C, Iezzi I, Dicarlo M, Cerqueni G, Coro F, Vozzi G. Cell-Free Demineralized Bone Matrix for Mesenchymal Stem Cells Survival and Colonization. MATERIALS 2019; 12:ma12091360. [PMID: 31027339 PMCID: PMC6538993 DOI: 10.3390/ma12091360] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/09/2019] [Accepted: 04/24/2019] [Indexed: 11/25/2022]
Abstract
Decellularized bone matrix is receiving much attention as biological scaffolds and implantable biomaterials for bone tissue regeneration. Here, we evaluated the efficacy of a cell-free demineralized bone matrix on mesenchymal stem cells (MSCs) survival and differentiation in vitro. The seeding of human umbilical cord-derived MSCs (hUC-SCs) on decellularized bone matrices up to 14 days was exploited, assessing their capability of scaffold colonization and evaluating gene expression of bone markers. Light and Scanning Electron Microscopies were used. The obtained cell-free decalcified structures showed elastic moduli attributable to both topology and biochemical composition. Morphological observation evidenced an almost complete colonization of the scaffolds after 14 days of culture. Moreover, in hUC-SCs cultured on decalcified scaffolds, without the addition of any osteoinductive media, there was an upregulation of Collagen Type I (COL1) and osteonectin (ON) gene expression, especially on day 14. Modifications in the expression of genes engaged in stemness were also detected. In conclusion, the proposed decellularized bone matrix can induce the in vitro hUC-SCs differentiation and has the potential to be tested for in in vivo tissue regeneration.
Collapse
Affiliation(s)
- Monica Mattioli-Belmonte
- Dipartimento di Scienze Cliniche e Molecolari-DISCLIMO, Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy.
| | - Francesca Montemurro
- Centro di Ricerca "E. Piaggio", Università di Pisa, Via Diotisalvi 1, 56122 Pisa, Italy.
| | - Caterina Licini
- Dipartimento di Scienza Applicata e Tecnologia-DISAT, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy.
| | - Iolanda Iezzi
- Dipartimento di Scienze Cliniche e Molecolari-DISCLIMO, Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy.
| | - Manuela Dicarlo
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, 70013 Castellana Grotte (BA), Italy.
| | - Giorgia Cerqueni
- Dipartimento di Scienze Cliniche e Molecolari-DISCLIMO, Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy.
| | - Florinda Coro
- Centro di Ricerca "E. Piaggio", Università di Pisa, Via Diotisalvi 1, 56122 Pisa, Italy.
| | - Giovanni Vozzi
- Centro di Ricerca "E. Piaggio", Università di Pisa, Via Diotisalvi 1, 56122 Pisa, Italy.
- Dipartimento di Ingegneria dell'Informazione-DII, Università di Pisa, Via Caruso 16, 56122 Pisa, Italy.
| |
Collapse
|
48
|
Tendon-Derived Stem Cell Differentiation in the Degenerative Tendon Microenvironment. Stem Cells Int 2018; 2018:2613821. [PMID: 30510582 PMCID: PMC6230403 DOI: 10.1155/2018/2613821] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/26/2018] [Indexed: 01/03/2023] Open
Abstract
Tendinopathy is prevalent in athletic and many occupational populations; nevertheless, the pathogenesis of tendinopathy remains unclear. Tendon-derived stem cells (TDSCs) were regarded as the key culprit for the development of tendinopathy. However, it is uncertain how TDSCs differentiate into adipocytes, chondrocytes, or osteocytes in the degenerative microenvironment of tendinopathy. So in this study, the regulating effects of the degenerative tendon microenvironment on differentiation of TDSCs were investigated. TDSCs were isolated from rat Achilles tendons and were grown on normal and degenerative (prepared by stress-deprived culture) decellularized tendon slices (DTSs). Immunofluorescence staining, H&E staining, real-time PCR, and Western blot were used to delineate the morphology, proliferation, and differentiation of TDSCs in the degenerative microenvironment. It was found that TDSCs were much more spread on the degenerative DTSs than those on normal DTSs. The tenocyte-related markers, COL1 and TNMD, were highly expressed on normal DTSs than the degenerative DTSs. The expression of chondrogenic and osteogenic markers, COL2, SOX9, Runx2, and ALP, was higher on the degenerative DTSs compared with TDSCs on normal DTSs. Furthermore, phosphorylated FAK and ERK1/2 were reduced on degenerative DTSs. In conclusion, this study found that the degenerative tendon microenvironment induced TDSCs to differentiate into chondrogenic and osteogenic lineages. It could be attributed to the cell morphology changes and reduced FAK and ERK1/2 activation in the degenerative microenvironment of tendinopathy.
Collapse
|
49
|
Liu C, Luo JW, Liang T, Lin LX, Luo ZP, Zhuang YQ, Sun YL. Matrix stiffness regulates the differentiation of tendon-derived stem cells through FAK-ERK1/2 activation. Exp Cell Res 2018; 373:62-70. [PMID: 30138615 DOI: 10.1016/j.yexcr.2018.08.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 12/12/2022]
Abstract
Tendon derived stem cells (TDSCs) were vital in tendon homeostasis. Nevertheless, the regulation of TDSCs differentiation in tendinopathy is unclear. Matrix stiffness modulated stem cells differentiation, and matrix stiffness of tendinopathic tissues decreased significantly. In order to clarify the role of matrix stiffness in TDSCs differentiation, they were cultured on the gelatin hydrogels with the stiffness from 2.34 ± 1.48 kPa to 24.09 ± 14.03 kPa. The effect of matrix stiffness on TDSCs proliferation and differentiation were investigated with CCK8 assay, immunofluorescences, real time PCR and western blot. It was found the proliferation of TDSCs increased and more stress fibers formed with increasing matrix stiffness. The differentiation of TDSCs into tenogenic, chondrogenic, and osteogenic lineages were inhibited on stiff hydrogel evidenced by reduced expression of tenocyte markers THBS4, TNMD, SCX, chondrocyte marker COL2, and osteocyte markers Runx2, Osterix, and ALP. Furthermore, the phosphorylation of FAK and ERK1/2 were enhanced when TDSCs grew on stiff hydrogel. After FAK or ERK1/2 was inhibited, the effect of matrix stiffness on differentiation of TDSCs was inhibited as well. The above results indicated matrix stiffness modulated the proliferation and differentiation of TDSCs, and the regulation effect could correlate to the activation of FAK or ERK1/2.
Collapse
Affiliation(s)
- Chang Liu
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518000, China; Central Laboratory, Dalian Municipal Central Hospital, Dalian 116033, China.
| | - Jing-Wan Luo
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518000, China.
| | - Ting Liang
- Institute of Orthopaedics, Soochow University, Suzhou 215007, China.
| | - Long-Xiang Lin
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518000, China.
| | - Zong-Ping Luo
- Institute of Orthopaedics, Soochow University, Suzhou 215007, China
| | | | - Yu-Long Sun
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518000, China.
| |
Collapse
|
50
|
He XT, Wu RX, Xu XY, Wang J, Yin Y, Chen FM. Macrophage involvement affects matrix stiffness-related influences on cell osteogenesis under three-dimensional culture conditions. Acta Biomater 2018; 71:132-147. [PMID: 29462712 DOI: 10.1016/j.actbio.2018.02.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/29/2018] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
Accumulating evidence indicates that the physicochemical properties of biomaterials exert profound influences on stem cell fate decisions. However, matrix-based regulation selected through in vitro analyses based on a given cell population do not genuinely reflect the in vivo conditions, in which multiple cell types are involved and interact dynamically. This study constitutes the first investigation of how macrophages (Mφs) in stiffness-tunable transglutaminase cross-linked gelatin (TG-gel) affect the osteogenesis of bone marrow-derived mesenchymal stem cells (BMMSCs). When a single cell type was cultured, low-stiffness TG-gels promoted BMMSC proliferation, whereas high-stiffness TG-gels supported cell osteogenic differentiation. However, Mφs in high-stiffness TG-gels were more likely to polarize toward the pro-inflammatory M1 phenotype. Using either conditioned medium (CM)-based incubation or Transwell-based co-culture, we found that Mφs encapsulated in the low-stiffness matrix exerted a positive effect on the osteogenesis of co-cultured BMMSCs. Conversely, Mφs in high-stiffness TG-gels negatively affected cell osteogenic differentiation. When both cell types were cultured in the same TG-gel type and placed into the Transwell system, the stiffness-related influences of Mφs on BMMSCs were significantly altered; both the low- and high-stiffness matrix induced similar levels of BMMSC osteogenesis. Although the best material parameter for synergistically affecting Mφs and BMMSCs remains unknown, our data suggest that Mφ involvement in the co-culture system alters previously identified material-related influences on BMMSCs, such as matrix stiffness-related effects, which were identified based on a culture system involving a single cell type. Such Mφ-stem cell interactions should be considered when establishing proper matrix parameter-associated cell regulation in the development of biomimetic biomaterials for regenerative applications. STATEMENT OF SIGNIFICANCE The substrate stiffness of a scaffold plays critical roles in modulating both reparative cells, such as mesenchymal stem cells (MSCs), and immune cells, such as macrophages (Mφs). Although the influences of material stiffness on either Mφs or MSCs, have been extensively described, how the two cell types respond to matrix cues to dynamically affect each other in a three-dimensional (3D) biosystem remains largely unknown. Here, we report our findings that, in a platform wherein Mφs and bone marrow-derived MSCs coexist, matrix stiffness can influence stem cell fate through both direct matrix-associated regulation and indirect Mφ-based modulation. Our data support future studies of the MSC-Mφ-matrix interplay in the 3D context to optimize matrix parameters for the development of the next biomaterial.
Collapse
|