1
|
Fouladgar F, Powell R, Irukuvarjula V, Joy A, Li X, Habibi N. Osteogenic differentiation of mesenchymal stem cells in cell-laden culture of self-assembling peptide hydrogels. OPENNANO 2025; 22:100235. [PMID: 40124089 PMCID: PMC11928154 DOI: 10.1016/j.onano.2025.100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Mesenchymal stem cell (MSC) osteogenic differentiation requires scaffolds to support multiple stages of growth and differentiation signals. Fluorenyl-9-methoxycarbonyl diphenylalanine (Fmoc-FF) peptides self-assemble to create 3D nanofibers. Here, we cultured MSC in 2D and 3D Fmoc-FF layers to support their osteogenic differentiation. The stiffness of the hydrogels was tunable between 100 and 10,000 Pa which allows precise modulation of the cellular microenvironment. Scaffold stiffness impacted cell viability which softer scaffolds (100 Pa) favored higher viability. MSC formed spheroids in 3D hydrogel and showed spread morphology in 2D overlayers. Our results demonstrate that the Fmoc-FF 3D cultures significantly enhanced osteogenic differentiation, as evidenced by increased calcium deposition, elevated phosphatase activity, and the secretion of osteocalcin. We propose that the peptides provide integrin-binding sites that activate a cytoplasmic feedback loop essential for differentiation. These findings suggest that self-assembled Fmoc-FF peptide hydrogels, is a promising platform for bone tissue engineering applications.
Collapse
Affiliation(s)
- Faye Fouladgar
- Department of Biomedical Engineering, University of North Texas, TX, USA
| | - Robert Powell
- Department of Biomedical Engineering, University of North Texas, TX, USA
| | | | - Akhila Joy
- Department of Material Science & Engineering, University of North Texas, TX, USA
| | - Xiao Li
- Department of Material Science & Engineering, University of North Texas, TX, USA
| | - Neda Habibi
- Department of Biomedical Engineering, University of North Texas, TX, USA
| |
Collapse
|
2
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
3
|
Shlykov MA, Smirnova PV, Senotov AS, Teterina AY, Minaychev VV, Smirnov IV, Novikov RA, Marchenko EI, Salynkin PS, Komlev VS, Fadeev RS, Fadeeva IS. Comparative Evaluation of Mathematical Model and In Vivo Study of Calcium Phosphate Bone Grafts. J Funct Biomater 2024; 15:368. [PMID: 39728168 DOI: 10.3390/jfb15120368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
One of the key factors of the interaction 'osteoplastic material-organism' is the state of the implant surface. Taking into account the fact that the equilibrium in regeneration conditions is reached only after the reparative histogenesis process is completed, the implant surface is constantly modified. This work is devoted to the numerical description of the dynamic bilateral material-medium interaction under close to physiological conditions, as well as to the assessment of the comparability of the model with in vitro and in vivo experimental results. The semi-empirical model obtained on the basis of chemical kinetics allows us to describe numerically the processes occurring in the in vitro systems and extrapolates well to assess the behavior of dicalcium phosphate dihydrate (DCPD) material under conditions of ectopic (subcutaneous) implantation in Wistar rats. It is shown that an experiment conducted using a perfusion-diffusion bioreactor in a cell culture medium with the addition of fetal bovine serum (FBS) allows for achieving morphologically and chemically identical changes in the surface of the material in comparison with the real organism. This fact opens up wide possibilities for the creation of an analog of a 'laboratory-on-a-chip' and the transition from classical in vivo models to more controlled and mathematically based in vitro systems.
Collapse
Affiliation(s)
- Mikhail A Shlykov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Polina V Smirnova
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Anatoliy S Senotov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia
| | - Anastasia Yu Teterina
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Vladislav V Minaychev
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia
| | - Igor V Smirnov
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Roman A Novikov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninskiy Prospect 47, 119991 Moscow, Russia
| | - Ekaterina I Marchenko
- Department of Crystallography and Crystal Chemistry, Faculty of Geology, Moscow State University, 119234 Moscow, Russia
| | - Pavel S Salynkin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia
| | - Vladimir S Komlev
- Baikov Institute of Metallurgy and Materials Science, Russian Academy of Sciences, Leninskiy Prospect 49, 119334 Moscow, Russia
| | - Roman S Fadeev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia
| | - Irina S Fadeeva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia
| |
Collapse
|
4
|
Jin B, Liao Y, Ding Z, Zou R, Xu F, Li Y, Cheng B, Niu L. The role of biophysical cues and their modulated exosomes in dental diseases: from mechanism to therapy. Stem Cell Res Ther 2024; 15:373. [PMID: 39427216 PMCID: PMC11491033 DOI: 10.1186/s13287-024-03990-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Dental diseases such as caries and periodontitis have been common public health problems. Dental disease treatment can be achieved through stem cell-based dental regeneration. Biophysical cues determine the fate of stem cells and govern the success of dental regeneration. Some studies have manifested exosomes derived from stem cells could not only inherit biophysical signals in microenvironment but also evade some issues in the treatment with stem cells. Nowadays, biophysical cue-regulated exosomes become another promising therapy in dental regenerative medicine. However, methods to improve the efficacy of exosome therapy and the underlying mechanisms are still unresolved. In this review, the association between biophysical cues and dental diseases was summarized. We retrospected the role of exosomes regulated by biophysical cues in curing dental diseases and promoting dental regeneration. Our research also delved into the mechanisms by which biophysical cues control the biogenesis, release, and uptake of exosomes, as well as potential methods to enhance the effectiveness of exosomes. The aim of this review was to underscore the important place biophysical cue-regulated exosomes occupy in the realm of dentistry, and to explore novel targets for dental diseases.
Collapse
Affiliation(s)
- Bilun Jin
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China
| | - Yuxin Liao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Zhaojing Ding
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China
| | - Ye Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
- College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China.
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China.
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
- College of Stomatology, Xi'an Jiaotong University, Xi'an, China.
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
5
|
Inchingolo AM, Inchingolo AD, Nardelli P, Latini G, Trilli I, Ferrante L, Malcangi G, Palermo A, Inchingolo F, Dipalma G. Stem Cells: Present Understanding and Prospects for Regenerative Dentistry. J Funct Biomater 2024; 15:308. [PMID: 39452606 PMCID: PMC11508604 DOI: 10.3390/jfb15100308] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Regenerative medicine in dentistry focuses on repairing damaged oral tissues using advanced tools like stem cells, biomaterials, and tissue engineering (TE). Mesenchymal stem cells (MSCs) from dental sources, such as dental pulp and periodontal ligament, show significant potential for tissue regeneration due to their proliferative and differentiative abilities. This systematic review, following PRISMA guidelines, evaluated fifteen studies and identified effective strategies for improving dental, periodontal, and bone tissue regeneration through scaffolds, secretomes, and bioengineering methods. Key advancements include the use of dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) to boost cell viability and manage inflammation. Additionally, pharmacological agents like matrine and surface modifications on biomaterials improve stem cell adhesion and promote osteogenic differentiation. By integrating these approaches, regenerative medicine and TE can optimize dental therapies and enhance patient outcomes. This review highlights the potential and challenges in this field, providing a critical assessment of current research and future directions.
Collapse
Affiliation(s)
- Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Paola Nardelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giulia Latini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Irma Trilli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| |
Collapse
|
6
|
Wan W, Zhang H, Niu L, Zhang M, Xu F, Li A, Pei D, Lin M, Cheng B. TGF-β1 promotes osteogenesis of mesenchymal stem cells via integrin mediated mechanical positive autoregulation. iScience 2024; 27:110262. [PMID: 39021801 PMCID: PMC11253692 DOI: 10.1016/j.isci.2024.110262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/18/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Positive autoregulation (PAR), one type of network motifs, provides a high phenotypic heterogeneity for cells to better adapt to their microenvironments. Typical mechanosensitive proteins can also form PAR, e.g., integrin mediated PAR, but the role of such mechanical PAR in physiological development and pathological process remains elusive. In this study, we found that transforming growth factor β1 (TGF-β1) and integrin levels decrease with tissue softening after the development of paradentium in vivo in rat model of periodontitis (an inflammatory disease with bone defect). Interestingly, TGF-β1 could induce the formation of mechanical PAR involving the integrin-FAK-YAP axis in mesenchymal stem cells (MSCs) by both in vitro experiments and in silico computational model. The computational model predicted a mechanical PAR involving the bimodal distribution of focus adhesions, which enables cells to accurately perceive extracellular mechanical cues. Thus, our analysis of TGF-β1 mediated mechanosensing mechanism on MSCs may help to better understand the molecular process underlying bone regeneration.
Collapse
Affiliation(s)
- Wanting Wan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004 P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049 P.R. China
| | - Hui Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004 P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049 P.R. China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004 P.R. China
| | - Min Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi’an 710032 P.R. China
| | - Feng Xu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049 P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049 P.R. China
| | - Ang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004 P.R. China
| | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004 P.R. China
| | - Min Lin
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049 P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049 P.R. China
| | - Bo Cheng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049 P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an 710049 P.R. China
| |
Collapse
|
7
|
Wang H, Zhou T, Ma W, Zheng J, Cao Z, He C, Lemos PA, Luo J. Transcriptome analysis revealed the new mechanism of the intra-myocardial injectable alginate-hydrogel in the treatment of ventricular function degradation. J Thorac Dis 2024; 16:2443-2459. [PMID: 38738236 PMCID: PMC11087630 DOI: 10.21037/jtd-24-358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/10/2024] [Indexed: 05/14/2024]
Abstract
Background Myocardial infarction (MI) is one of the most lethal cardiovascular diseases. The loss of cardiomyocytes and the degradation of the extracellular matrix leads to high ventricular wall stress, which further drives the pathological thinning of the ventricular wall during MI. Injecting biomaterials to thicken the infarct ventricular wall provides mechanical support, thereby inhibiting the continued expansion of the heart. As an injectable biomaterial, alginate hydrogel has achieved exciting results in clinical trials, but further research needs to be conducted to determine whether it can improve cardiac function in addition to providing mechanical support. This study sought to explore these mechanisms in an animal model of MI. Methods A MI model was established in male C57BL/6J mice by ligation of the proximal left anterior descending (LAD) coronary artery. Intramyocardial injections (hydrogel or saline group) were performed in the proximal wall regions bordering the infarct area (with one 20-µL injection). Four weeks after MI, RNA sequencing revealed that 342 messenger RNAs (mRNAs) from the infarcted hearts were differentially expressed between the saline group and hydrogel group. We subsequently conducted a Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis to analyze the RNA sequencing data. In addition, we employed both western blotting and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) techniques to verify a number of genes that were differentially expressed and could potentially affect cardiac function after MI. Subsequently, we confirmed these findings through in vitro experiments. Results We found that compared with hydrogel treatment group, 250 mRNAs were upregulated and 92 mRNAs were downregulated in saline group (P<0.05). And by exploring the GO and KEGG signaling pathways as well as the protein-protein interaction (PPI) network, we found that administration of alginate hydrogel modulated cardiomyocyte inflammation-associated proteins as well as chemokine-related proteins during the inflammatory response phase after MI. In addition, our analysis at both the protein and RNA level revealed that B2M was effective in improving cardiac function after MI in the hydrogel treatment group, which was consistent in the myocardium oxygen and glucose deprivation (OGD) injury model. Conclusions We explored the transcriptome changes of infarcted hearts after alginate-hydrogel injection during the inflammatory response period. Our findings suggest that the injectable hydrogel directly alters the inflammatory response and the chemokine-mediated signaling pathway of cardiomyocytes, ultimately improving cardiac function.
Collapse
Affiliation(s)
- Huiyong Wang
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Zhou
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wentao Ma
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | | | | | - Caiyun He
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pedro A. Lemos
- Department of Cardiology, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
- Department of Cardiology, Heart Institute-InCor, University of Sao Paulo Medical School, Sao Paulo, SP, Brazil
| | - Jianfang Luo
- Department of Cardiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
8
|
Doron G, Wood LB, Guldberg RE, Temenoff JS. Poly(ethylene glycol)-Based Hydrogel Microcarriers Alter Secretory Activity of Genetically Modified Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2023; 9:6282-6292. [PMID: 37906515 PMCID: PMC10646834 DOI: 10.1021/acsbiomaterials.3c00954] [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: 07/13/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023]
Abstract
In order to scale up culture therapeutic cells, such as mesenchymal stromal cells (MSCs), culture in suspension bioreactors using microcarriers (μCs) is preferred. However, the impact of microcarrier type on the resulting MSC secretory activity has not been investigated. In this study, two poly(ethylene glycol) hydrogel formulations with different swelling ratios (named "stiffer" and "softer") were fabricated as μC substrates to culture MSCs and MSCs genetically modified to express the interleukin-1 receptor antagonist (IL-1Ra-MSCs). Changes in cell number, secretory and angiogenic activity, and changes in MAPK signaling were evaluated when cultured on hydrogel μCs, as well as on tissue culture plastic-based Synthemax μCs. We demonstrated that culture on stiffer μCs increased secretion of IL-1Ra compared to culture on Synthemax μCs by IL-1Ra-MSCs by 1.2- to 1.6-fold, as well as their in vitro angiogenic activity, compared to culture on Synthemax μCs, while culture on both stiffer and softer μCs altered the secretion of several other factors compared to culture on Synthemax μCs. Changes in angiogenic activity corresponded with increased gene expression and secretion of hepatocyte growth factor by MSCs cultured on softer μCs by 2.5- to 6-fold compared to MSCs cultured on Synthemax μCs. Quantification of phosphoprotein signaling with the MAPK pathway revealed broad reduction of pathway activation by IL-1Ra-MSCs cultured on both stiffer and softer μCs compared to Synthemax, where phosphorylated c-Jun, ATF2, and MEK1 were reduced specifically on softer μCs. Overall, this study showed that μC surfaces can influence the secretory activity of genetically modified MSCs and identified associated changes in MAPK pathway signaling, which is a known central regulator of cytokine secretion.
Collapse
Affiliation(s)
- Gilad Doron
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| | - Levi B. Wood
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Dr. NW, Atlanta, Georgia 30318, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| | - Robert E. Guldberg
- Knight
Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, Oregon 97403, United States
| | - Johnna S. Temenoff
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, 313 Ferst Dr. NW, Atlanta, Georgia 30332, United States
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, Georgia 30332, United States
| |
Collapse
|
9
|
李 驰, 樊 瑜, 郑 丽. [Differentiation of stem cells regulated by biophysical cues]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2023; 40:609-616. [PMID: 37666749 PMCID: PMC10477397 DOI: 10.7507/1001-5515.202208002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/03/2022] [Indexed: 09/06/2023]
Abstract
Stem cells have been regarded with promising application potential in tissue engineering and regenerative medicine due to their self-renewal and multidirectional differentiation abilities. However, their fate is relied on their local microenvironment, or niche. Recent studied have demonstrated that biophysical factors, defined as physical microenvironment in which stem cells located play a vital role in regulating stem cell committed differentiation. In vitro, synthetic physical microenvironments can be used to precisely control a variety of biophysical properties. On this basis, the effect of biophysical properties such as matrix stiffness, matrix topography and mechanical force on the committed differentiation of stem cells was further investigated. This paper summarizes the approach of mechanical models of artificial physical microenvironment and reviews the effects of different biophysical characteristics on stem cell differentiation, in order to provide reference for future research and development in related fields.
Collapse
Affiliation(s)
- 驰宇 李
- 北京航空航天大学 生物与医学工程学院 北京市生物医学工程高精尖创新中心 生物力学与力生物学教育部重点实验室(北京 100083)Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P. R. China
| | - 瑜波 樊
- 北京航空航天大学 生物与医学工程学院 北京市生物医学工程高精尖创新中心 生物力学与力生物学教育部重点实验室(北京 100083)Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P. R. China
| | - 丽沙 郑
- 北京航空航天大学 生物与医学工程学院 北京市生物医学工程高精尖创新中心 生物力学与力生物学教育部重点实验室(北京 100083)Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P. R. China
| |
Collapse
|
10
|
Sheng R, Liu J, Zhang W, Luo Y, Chen Z, Chi J, Mo Q, Wang M, Sun Y, Liu C, Zhang Y, Zhu Y, Kuang B, Yan C, Liu H, Backman LJ, Chen J. Material Stiffness in Cooperation with Macrophage Paracrine Signals Determines the Tenogenic Differentiation of Mesenchymal Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206814. [PMID: 37097733 DOI: 10.1002/advs.202206814] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/18/2023] [Indexed: 06/15/2023]
Abstract
Stiffness is an important physical property of biomaterials that determines stem cell fate. Guiding stem cell differentiation via stiffness modulation has been considered in tissue engineering. However, the mechanism by which material stiffness regulates stem cell differentiation into the tendon lineage remains controversial. Increasing evidence demonstrates that immune cells interact with implanted biomaterials and regulate stem cell behaviors via paracrine signaling; however, the role of this mechanism in tendon differentiation is not clear. In this study, polydimethylsiloxane (PDMS) substrates with different stiffnesses are developed, and the tenogenic differentiation of mesenchymal stem cells (MSCs) exposed to different stiffnesses and macrophage paracrine signals is investigated. The results reveal that lower stiffnesses facilitates tenogenic differentiation of MSCs, while macrophage paracrine signals at these stiffnesses suppress the differentiation. When exposed to these two stimuli, MSCs still exhibit enhanced tendon differentiation, which is further elucidated by global proteomic analysis. Following subcutaneous implantation in rats for 2 weeks, soft biomaterial induces only low inflammation and promotes tendon-like tissue formation. In conclusion, the study demonstrates that soft, rather than stiff, material has a greater potential to guide tenogenic differentiation of stem cells, which provides comprehensive evidence for optimized bioactive scaffold design in tendon tissue engineering.
Collapse
Affiliation(s)
- Renwang Sheng
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Jia Liu
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, P. R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, P. R. China
| | - Yifan Luo
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Jiayu Chi
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Qingyun Mo
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Yuzhi Sun
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, P. R. China
| | - Chuanquan Liu
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Yanan Zhang
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Yue Zhu
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Baian Kuang
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Chunguang Yan
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Haoyang Liu
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, Umeå, SE-901 87, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, Umeå, SE-901 87, Sweden
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing, 210009, P. R. China
- Center for Stem Cell and Regenerative Medicine, Southeast University, Nanjing, 210009, P. R. China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, P. R. China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, P. R. China
| |
Collapse
|
11
|
Qu JY, Xie HT, Xiao YT, Zhang YY, Hu ZX, Wang JS, Zhang MC, Xi H. The inhibition of p38 MAPK blocked inflammation to restore the functions of rat meibomian gland epithelial cells. Exp Eye Res 2023; 231:109470. [PMID: 37059216 DOI: 10.1016/j.exer.2023.109470] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/03/2023] [Accepted: 04/08/2023] [Indexed: 04/16/2023]
Abstract
Meibomian glands (MGs) are vital for ocular surface health. However, the roles of inflammation in the progression of meibomian gland dysfunction (MGD) are largely unknown. In this study, the roles of the inflammation factor interleukin-1β (IL-1β) via the p38 mitogen-activated protein kinases (MAPK) signaling pathway on rat meibomian gland epithelial cells (RMGECs) were explored. Eyelids from adult rat mice at 2 months and 2 years of age were stained with specific antibodies against IL-1β to identify inflammation levels. RMGECs were exposed to IL-1β and/or SB203580, a specific inhibitor of p38 MAPK signaling pathway, for 3 days. Cell proliferation, keratinization, lipid accumulation, and matrix metalloproteinases 9 (MMP9) expression were evaluated by MTT assay, polymerase chain reaction (PCR), immunofluorescence staining, apoptosis assay, lipid staining, and Western blot analyses. We found that IL-1β was significantly higher in the terminal ducts of MGs in rats with age-related MGD than in young rats. IL-1β inhibited cell proliferation, suppressed lipid accumulation and peroxisome proliferator activator receptor γ (PPARγ) expression, and promoted apoptosis while activating the p38 MAPK signaling pathway. Cytokeratin 1 (CK1), a marker for complete keratinization, and MMP9 in RMGECs were also up-regulated by IL-1β. SB203580 effectively diminished the effects of IL-1β on differentiation, keratinization, and MMP9 expression by blocking IL-1β-induced p38 MAPK activation, although it also inhibited cell proliferation. The inhibition of the p38 MAPK signaling pathway blocked IL-1β-induced differentiation reduction, hyperkeratinization, and MMP9 overexpression of RMGECs, which provides a potential therapy for MGD.
Collapse
Affiliation(s)
- Jing-Yu Qu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China; Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266000, China; State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Qingdao, 266000, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu-Ting Xiao
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ying-Ying Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhi-Xin Hu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jia-Song Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Han Xi
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Gross T, Dieterle MP, Vach K, Altenburger MJ, Hellwig E, Proksch S. Biomechanical Modulation of Dental Pulp Stem Cell (DPSC) Properties for Soft Tissue Engineering. Bioengineering (Basel) 2023; 10:bioengineering10030323. [PMID: 36978714 PMCID: PMC10045720 DOI: 10.3390/bioengineering10030323] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Dental pulp regeneration strategies frequently result in hard tissue formation and pulp obliteration. The aim of this study was to investigate whether dental pulp stem cells (DPSCs) can be directed toward soft tissue differentiation by extracellular elasticity. STRO-1-positive human dental pulp cells were magnetically enriched and cultured on substrates with elasticities of 1.5, 15, and 28 kPa. The morphology of DPSCs was assessed visually. Proteins relevant in mechanobiology ACTB, ITGB1, FAK, p-FAK, TALIN, VINCULIN, PAXILLIN, ERK 1/2, and p-ERK 1/2 were detected by immunofluorescence imaging. Transcription of the pulp marker genes BMP2, BMP4, MMP2, MMP3, MMP13, FN1, and IGF2 as well as the cytokines ANGPT1, VEGF, CCL2, TGFB1, IL2, ANG, and CSF1 was determined using qPCR. A low stiffness, i.e., 1.5 kPa, resulted in a soft tissue-like phenotype and gene expression, whereas DPSCs on 28 kPa substrates exhibited a differentiation signature resembling hard tissues with a low cytokine expression. Conversely, the highest cytokine expression was observed in cells cultured on intermediate elasticity, i.e., 15 kPa, substrates possibly allowing the cells to act as “trophic mediators”. Our observations highlight the impact of biophysical cues for DPSC fate and enable the design of scaffold materials for clinical pulp regeneration that prevent hard tissue formation.
Collapse
Affiliation(s)
- Tara Gross
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
- G.E.R.N. Research Center for Tissue Replacement, Regeneration and Neogenesis, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstr. 4, 79108 Freiburg, Germany
- Correspondence: ; Tel.: +49-(0)761-270-48850; Fax: +49-(0)761-270-47620
| | - Martin Philipp Dieterle
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Kirstin Vach
- Institute of Medical Biometry and Statistics, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs—University of Freiburg, Stefan-Meier-Str. 26, 79104 Freiburg, Germany
| | - Markus Joerg Altenburger
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
- G.E.R.N. Research Center for Tissue Replacement, Regeneration and Neogenesis, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstr. 4, 79108 Freiburg, Germany
| | - Elmar Hellwig
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Susanne Proksch
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
- G.E.R.N. Research Center for Tissue Replacement, Regeneration and Neogenesis, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Engesserstr. 4, 79108 Freiburg, Germany
- Dental Clinic 1–Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glückstr. 11, 91054 Erlangen, Germany
| |
Collapse
|
13
|
Ma Y, Zhang X, Tang S, Xue L, Wang J, Zhang X. Extended preconditioning on soft matrices directs human mesenchymal stem cell fate via YAP transcriptional activity and chromatin organization. APL Bioeng 2023; 7:016110. [PMID: 36845904 PMCID: PMC9949900 DOI: 10.1063/5.0124424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Dynamic extracellular matrix (ECM) mechanics plays a crucial role in tissue development and disease progression through regulation of stem cell behavior, differentiation, and fate determination. Periodontitis is a typical case characterized by decreased ECM stiffness within diseased periodontal tissues as well as with irreversible loss of osteogenesis capacity of periodontal tissue-derived human periodontal tissue-derived MSCs (hMSCs) even returning back to a physiological mechanical microenvironment. We hypothesized that the hMSCs extendedly residing in the soft ECM of diseased periodontal tissues may memorize the mechanical information and have further effect on ultimate cell fate besides the current mechanical microenvironment. Using a soft priming and subsequent stiff culture system based on collagen-modified polydimethylsiloxane substrates, we were able to discover that extended preconditioning on soft matrices (e.g., 7 days of exposure) led to approximately one-third decrease in cell spreading, two-third decrease in osteogenic markers (e.g., RUNX2 and OPN) of hMSCs, and one-thirteenth decrease in the production of mineralized nodules. The significant loss of osteogenic ability may attribute to the long-term residing of hMSCs in diseased periodontal tissue featured with reduced stiffness. This is associated with the regulation of transcriptional activity through alterations of subcellular localization of yes-associated protein and nuclear feature-mediated chromatin organization. Collectively, we reconstructed phenomena of irreversible loss of hMSC osteogenesis capacity in diseased periodontal tissues in our system and revealed the critical effect of preconditioning duration on soft matrices as well as the potential mechanisms in determining ultimate hMSC fate.
Collapse
Affiliation(s)
- Yufei Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Xu Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Shaoxin Tang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Li Xue
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-End Equipment, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Jing Wang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Xiaohui Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Center for Mitochondrial Biology and Medicine, School of Life Science and Technology, International Joint Laboratory for Micro/Nano Manufacturing and Measurement Technology, Xi'an Key Laboratory for Biomedical Testing and High-End Equipment, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| |
Collapse
|
14
|
Guan P, Ji Y, Kang X, Liu W, Yang Q, Liu S, Lin Y, Zhang Z, Li J, Zhang Y, Liu C, Fan L, Sun Y. Biodegradable Dual-Cross-Linked Hydrogels with Stem Cell Differentiation Regulatory Properties Promote Growth Plate Injury Repair via Controllable Three-Dimensional Mechanics and a Cartilage-like Extracellular Matrix. ACS APPLIED MATERIALS & INTERFACES 2023; 15:8986-8998. [PMID: 36752284 DOI: 10.1021/acsami.2c20722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Recent breakthroughs in cell transplantation therapy have revealed the promising potential of bone marrow mesenchymal stem cells (BMSCs) for promoting the regeneration of growth plate cartilage injury. However, the high apoptosis rate and the uncertainty of the differentiation direction of cells often lead to poor therapeutic effects. Cells are often grown under three-dimensional (3D) conditions in vivo, and the stiffness and components of the extracellular matrix (ECM) are important regulators of stem cell differentiation. To this end, a 3D cartilage-like ECM hydrogel with tunable mechanical properties was designed and synthesized mainly from gelatin methacrylate (GM) and oxidized chondroitin sulfate (OCS) via dynamic Schiff base bonding under UV. The effects of scaffold stiffness and composition on the survival and differentiation of BMSCs in vitro were investigated. A rat model of growth plate injury was developed to validate the effect of the GMOCS hydrogels encapsulated with BMSCs on the repair of growth plate injury. The results showed that 3D GMOCS hydrogels with an appropriate modulus significantly promoted chondrogenic differentiation of BMSCs, and GMOCS/BMSC transplantation could effectively inhibit bone bridge formation and promote the repair of damaged growth plates. Accordingly, GMOCS/BMSC therapy can be engineered as a promising therapeutic candidate for growth plate injury.
Collapse
Affiliation(s)
- Pengfei Guan
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Yuelun Ji
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Xinchang Kang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Weilu Liu
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qinfeng Yang
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shencai Liu
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yeying Lin
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Zuyu Zhang
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Junji Li
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Can Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lei Fan
- Department of Orthopedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yongjian Sun
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Zanut A, Li R, Deng R, Liu X, Rejhon M, Chen W, Weck M, de Peppo GM, Riedo E. A Polymer Canvas with the Stiffness of the Bone Matrix to Study and Control Mesenchymal Stem Cell Response. Adv Healthc Mater 2022; 12:e2201503. [PMID: 36565136 DOI: 10.1002/adhm.202201503] [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: 06/22/2022] [Revised: 11/30/2022] [Indexed: 12/25/2022]
Abstract
Reproducing in vitro the complex multiscale physical features of human tissues creates novel biomedical opportunities and fundamental understanding of cell-environment interfaces and interactions. While stiffness has been recognized as a key driver of cell behavior, systematic studies on the role of stiffness have been limited to values in the KPa-MPa range, significantly below the stiffness of bone. Here, a platform enabling the tuning of the stiffness of a biocompatible polymeric interface up to values characteristic of human bone is reported, which are in the GPa range, by using extremely thin polymer films on glass and cross-linking the films using ultraviolet (UV) light irradiation. It is shown that a higher stiffness is related to better adhesion, proliferation, and osteogenic differentiation, and that it is possible to switch on/off cell attachment and growth by solely tuning the stiffness of the interface, without any surface chemistry or topography modification. Since the stiffness is tuned directly by UV irradiation, this platform is ideal for rapid and simple fabrication of stiffness patterns and gradients, thus representing an innovative tool for combinatorial studies of the synergistic effect of tissue environmental cues on cell behavior, and creates new opportunities for next-generation biosensors, single-cell patterning, and lab-on-a-chip devices.
Collapse
Affiliation(s)
- Alessandra Zanut
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA
| | - Rui Li
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA
| | - Ru Deng
- Department of Chemistry, New York University, Silver Center Block, 100 Washington Square E, New York, NY, 10003, USA
| | - Xiangyu Liu
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA
| | - Martin Rejhon
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA
| | - Weiqiang Chen
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA
| | - Marcus Weck
- Department of Chemistry, New York University, Silver Center Block, 100 Washington Square E, New York, NY, 10003, USA
| | - Giuseppe Maria de Peppo
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA.,The New York Stem Cell Foundation Research Institute, 619 West 54th Street, New York, NY, 10019, USA.,Mirimus, Inc, 760 Parkside Ave, Brooklyn, NY, 11226, USA
| | - Elisa Riedo
- Tandon School of Engineering, New York University, 6 Metrotech, Brooklyn, NY, 11201, USA
| |
Collapse
|
16
|
Wang J, Xie SA, Li N, Zhang T, Yao W, Zhao H, Pang W, Han L, Liu J, Zhou J. Matrix stiffness exacerbates the proinflammatory responses of vascular smooth muscle cell through the DDR1-DNMT1 mechanotransduction axis. Bioact Mater 2022; 17:406-424. [PMID: 35386458 PMCID: PMC8964982 DOI: 10.1016/j.bioactmat.2022.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/16/2021] [Accepted: 01/06/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular smooth muscle cell (vSMC) is highly plastic as its phenotype can change in response to mechanical cues inherent to the extracellular matrix (ECM). VSMC may be activated from its quiescent contractile phenotype to a proinflammatory phenotype, whereby the cell secretes chemotactic and inflammatory cytokines, e.g. MCP1 and IL6, to functionally regulate monocyte and macrophage infiltration during the development of various vascular diseases including arteriosclerosis. Here, by culturing vSMCs on polyacrylamide (PA) substrates with variable elastic moduli, we discovered a role of discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase that binds collagens, in mediating the mechanical regulation of vSMC gene expression, phenotype, and proinflammatory responses. We found that ECM stiffness induced DDR1 phosphorylation, oligomerization, and endocytosis to repress the expression of DNA methyltransferase 1 (DNMT1), very likely in a collagen-independent manner. The DDR1-to-DNMT1 signaling was sequentially mediated by the extracellular signal-regulated kinases (ERKs) and p53 pathways. ECM stiffness primed vSMC to a proinflammatory phenotype and this regulation was diminished by DDR1 inhibition. In agreement with the in vitro findings, increased DDR1 phosphorylation was observed in human arterial stiffening. DDR1 inhibition in mouse attenuated the acute injury or adenine diet-induced vascular stiffening and inflammation. Furthermore, mouse vasculature with SMC-specific deletion of Dnmt1 exhibited proinflammatory and stiffening phenotypes. Our study demonstrates a role of SMC DDR1 in perceiving the mechanical microenvironments and down-regulating expression of DNMT1 to result in vascular pathologies and has potential implications for optimization of engineering artificial vascular grafts and vascular networks. DDR1 is a mechanosensor in vSMC to perceive ECM stiffness in a collagen binding-independent way. Activation of DDR1 leads to repression of DNMT1 expression via the ERK-p53 pathway. The DDR1-DNMT1 axis mediates ECM stiffening-induced vascular inflammation.
Collapse
Affiliation(s)
- Jin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
| | - Si-an Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, PR China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), And Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, PR China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, PR China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, PR China
| | - Weijuan Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Hucheng Zhao
- Institute of Biomechanics and Medical Engineering, School of Aerospace Engineering, Tsinghua University, Beijing, PR China
| | - Wei Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Lili Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
| | - Jiayu Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides; Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, PR China
- Corresponding author. Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, PR China.
| |
Collapse
|
17
|
Li W, Wang C, Wang Z, Gou L, Zhou Y, Peng G, Zhu M, Zhang J, Li R, Ni H, Wu L, Zhang W, Liu J, Tian Y, Chen Z, Han YP, Tong N, Fu X, Zheng X, Berggren PO. Physically Cross-Linked DNA Hydrogel-Based Sustained Cytokine Delivery for In Situ Diabetic Alveolar Bone Rebuilding. ACS APPLIED MATERIALS & INTERFACES 2022; 14:25173-25182. [PMID: 35638566 DOI: 10.1021/acsami.2c04769] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The development of a biodegradable and shape-adaptable bioscaffold that can enhance local cytokine retention and bioactivity is essential for the application of immunotherapy in periodontal diseases. Here, we report a biodegradable, anti-inflammatory, and osteogenic ILGel that uses a physically cross-linked DNA hydrogel as a soft bioscaffold for the long-term sustained release of cytokine interleukin-10 (IL-10) to accelerate diabetic alveolar bone rebuilding. Porous microstructures of ILGel favored the encapsulation of IL-10 and maintained IL-10 bioactivity for at least 7 days. ILGel can be gradually degraded or hydrolyzed under physiological conditions, avoiding the potential undesired side effects on dental tissues. Long-term sustained release of bioactive IL-10 from ILGel not only promoted M2 macrophage polarization and attenuated periodontal inflammation but also triggered osteogenesis of mesenchymal stem cells (MSCs), leading to accelerated alveolar bone formation and healing of alveolar bone defects under diabetic conditions in vivo. ILGel treatment significantly accelerated the defect healing rate of diabetic alveolar injury up to 93.42 ± 4.6% on day 21 post treatment compared to that of free IL-10 treatment (63.30 ± 7.39%), with improved trabecular architectures. Our findings imply the potential application of the DNA hydrogel as the bioscaffold for cytokine-based immunotherapy in diabetic alveolar bone injury and other periodontal diseases.
Collapse
Affiliation(s)
- Wei Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengshi Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhenghao Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Liping Gou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ge Peng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zhu
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiayi Zhang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruoqing Li
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of General Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, China
| | - Hengfan Ni
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Wu
- Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanli Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yali Tian
- West China School of Nursing, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhong Chen
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yuan-Ping Han
- The Center for Growth, Metabolism and Aging, The College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xianghui Fu
- Division of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Per-Olof Berggren
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden
| |
Collapse
|
18
|
Pan Y, Gu Z, Lyu Y, Yang Y, Chung M, Pan X, Cai S. Link between senescence and cell fate: Senescence-associated secretory phenotype (SASP) and its effects on stem cell fate transition. Rejuvenation Res 2022; 25:160-172. [PMID: 35658548 DOI: 10.1089/rej.2022.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Senescence is a form of durable cell cycle arrest elicited in response to a wide range of stimuli. Senescent cells remain metabolically active and secrete a variety of factors collectively termed senescence-associated secretory phenotype (SASP). SASP is highly pleiotropic and can impact numerous biological processes in which it has both beneficial and deleterious roles. The underlying mechanisms by which SASP exerts its pleiotropic influence remain largely unknown. SASP serves as an environmental factor, which regulates stem cell differentiation and alters its routine. The latter can potentially be accomplished through dedifferentiation, transdifferentiation, or reprogramming. Behavioral changes that cells undergo when exposed to SASP are involved in several senescence-associated physiological and pathological phenomena. These findings provide clues for identifying possible interventions to reduce the deleterious effects without interfering in the beneficial outcomes. Here, we discuss the multifaced effects of SASP and the changes occurring in cellular states upon exposure to SASP factors.
Collapse
Affiliation(s)
- Yu Pan
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Zhenzhen Gu
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Yansi Lyu
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Yi Yang
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Manhon Chung
- Shanghai Jiao Tong University School of Medicine, 56694, Shanghai, China;
| | - Xiaohua Pan
- Shenzhen University, 47890, Shenzhen, Guangdong, China;
| | - Sa Cai
- Shenzhen University, 47890, 3688 Nanhai Avenue, Nanshan District, Shenzhen, Shenzhen, China, 518060;
| |
Collapse
|
19
|
Mo Q, Zhang W, Zhu A, Backman LJ, Chen J. Regulation of osteogenic differentiation by the pro-inflammatory cytokines IL-1β and TNF-α: current conclusions and controversies. Hum Cell 2022; 35:957-971. [PMID: 35522425 DOI: 10.1007/s13577-022-00711-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/23/2022] [Indexed: 12/09/2022]
Abstract
Treatment of complex bone fracture diseases is still a complicated problem that is urged to be solved in orthopedics. In bone tissue engineering, the use of mesenchymal stromal/stem cells (MSCs) for tissue repair brings hope to the medical field of bone diseases. MSCs can differentiate into osteoblasts and promote bone regeneration. An increasing number of studies show that the inflammatory microenvironment affects the osteogenic differentiation of MSCs. It is shown that TNF-α and IL-1β play different roles in the osteogenic differentiation of MSCs via different signal pathways. The main factors that affect the role of TNF-α and IL-1β in osteogenic differentiation of MSCs include concentration and the source of stem cells (different species and different tissues). This review in-depth analyzes the roles of pro-inflammatory cytokines in the osteogenic differentiation of MSCs and reveals some current controversies to provide a reference of comprehensively understanding.
Collapse
Affiliation(s)
- Qingyun Mo
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China
| | - Aijing Zhu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ludvig J Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87, Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, SE-901 87, Umeå, Sweden
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing, 210009, China.
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, China.
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
20
|
Tan L, Liu X, Dou H, Hou Y. Characteristics and regulation of mesenchymal stem cell plasticity by the microenvironment — specific factors involved in the regulation of MSC plasticity. Genes Dis 2022; 9:296-309. [PMID: 35224147 PMCID: PMC8843883 DOI: 10.1016/j.gendis.2020.10.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/05/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), multipotent stromal cells, have attracted extensive attention in the field of regenerative medicine and cell therapy due to the capacity of self-renewal, multilineage differentiation, and immune regulation. MSCs have different cellular effects in different diseases, and even have markedly different curative effects with different tissue sources, indicating the plasticity of MSCs. The phenotypes, secreted factors, and proliferative, migratory, differentiating, and immunomodulatory effects of MSCs depend on certain mediators present in their microenvironment. Understanding microenvironmental factors and their internal mechanisms in MSC responses may help in subsequent prediction and improvement of clinical benefits. This review highlighted the recent advances in MSC plasticity in the physiological and pathological microenvironment and multiple microenvironmental factors regulating MSC plasticity. It also highlighted some progress in the underlying molecular mechanisms of MSC remodeling in the microenvironment. It might provide references for the improvement in vitro culture of MSCs, clinical application, and in vivo induction.
Collapse
|
21
|
Roncada T, Bonithon R, Blunn G, Roldo M. Soft substrates direct stem cell differentiation into the chondrogenic lineage without the use of growth factors. J Tissue Eng 2022; 13:20417314221122121. [PMID: 36199979 PMCID: PMC9528007 DOI: 10.1177/20417314221122121] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 12/03/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold great promise for the treatment of cartilage related injuries. However, selectively promoting stem cell differentiation in vivo is still challenging. Chondrogenic differentiation of MSCs usually requires the use of growth factors that lead to the overexpression of hypertrophic markers. In this study, for the first time the effect of stiffness on MSC differentiation has been tested without the use of growth factors. Three-dimensional collagen and alginate scaffolds were developed and characterised. Stiffness significantly affected gene expression and ECM deposition. While, all hydrogels supported chondrogenic differentiation and allowed deposition of collagen type II and aggrecan, the 5.75 kPa hydrogel showed limited production of collagen type I compared to the other two formulations. These findings demonstrated for the first time that stiffness can guide MSCs differentiation without the use of growth factors within a tissue engineering scaffold suitable for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Tosca Roncada
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Roxane Bonithon
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
- Marta Roldo, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
22
|
Wang L, Zheng F, Song R, Zhuang L, Yang M, Suo J, Li L. Integrins in the Regulation of Mesenchymal Stem Cell Differentiation by Mechanical Signals. Stem Cell Rev Rep 2021; 18:126-141. [PMID: 34536203 DOI: 10.1007/s12015-021-10260-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Mesenchymal stem cells (MSCs) can sense and convert mechanical stimuli signals into a chemical response. Integrins are involved in the mechanotransduction from inside to outside and from outside to inside, and ultimately affect the fate of MSCs responding to different mechanical signals. Different integrins participate in different signaling pathways to regulate MSCs multi-differentiation. In this review, we summarize the latest advances in the effects of mechanical signals on the differentiation of MSCs, the importance of integrins in mechanotransduction, the relationship between integrin heterodimers and different mechanical signals, and the interaction among mechanical signals. We put forward our views on the prospect and challenges of developing mechanical biology in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastrointestinal Surgery, Jilin University First Hospital, Jilin University, 130021, Changchun, People's Republic of China
| | - Fuwen Zheng
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Ruixue Song
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Lequan Zhuang
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Ming Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, 130021, Changchun, People's Republic of China.
| | - Jian Suo
- Department of Gastrointestinal Surgery, Jilin University First Hospital, Jilin University, 130021, Changchun, People's Republic of China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 130021, Changchun, People's Republic of China.
| |
Collapse
|
23
|
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
|
24
|
Wang Z, Zhu X, Cong X. Spatial micro-variation of 3D hydrogel stiffness regulates the biomechanical properties of hMSCs. Biofabrication 2021; 13. [PMID: 34107453 DOI: 10.1088/1758-5090/ac0982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are one of the most promising candidates for cell-based therapeutic products. Nonetheless, their biomechanical phenotype afterin vitroexpansion is still unsatisfactory, for example, restricting the efficiency of microcirculation of delivered hMSCs for further cell therapies. Here, we propose a scheme using maleimide-dextran hydrogel with locally varied stiffness in microscale to modify the biomechanical properties of hMSCs in three-dimensional (3D) niches. We show that spatial micro-variation of stiffness can be controllably generated in the hydrogel with heterogeneously cross-linking via atomic force microscopy measurements. The result of 3D cell culture experiment demonstrates the hydrogels trigger the formation of multicellular spheroids, and the derived hMSCs could be rationally softened via adjustment of the stiffness variation (SV) degree. Importantly,in vitro, the hMSCs modified with the higher SV degree can pass easier through capillary-shaped micro-channels. Further, we discuss the underlying mechanics of the increased cellular elasticity by focusing on the effect of rearranged actin networks, via the proposed microscopic model of biomechanically modified cells. Overall, this work highlights the effectiveness of SV-hydrogels in reprogramming and manufacturing hMSCs with designed biomechanical properties for improved therapeutic potential.
Collapse
Affiliation(s)
- Zheng Wang
- College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China
| | - Xiaolu Zhu
- College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China.,Changzhou Key Laboratory of Digital Manufacture Technology, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China.,Jiangsu Key Laboratory of Special Robot Technology, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China
| | - Xiuli Cong
- Department of Orthopaedics, Zhejiang Hospital, No. 12 Lingyin Road, Hangzhou, Zhejiang 310013, People's Republic of China
| |
Collapse
|
25
|
Guo X, Wu Z. GABARAP ameliorates IL-1β-induced inflammatory responses and osteogenic differentiation in bone marrow-derived stromal cells by activating autophagy. Sci Rep 2021; 11:11561. [PMID: 34078931 PMCID: PMC8172545 DOI: 10.1038/s41598-021-90586-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/13/2021] [Indexed: 12/13/2022] Open
Abstract
Bone mesenchymal stem cells (BMSCs) are the most commonly investigated progenitor cells in bone defect repair and osteoarthritis subchondral bone regeneration; however, these studies are limited by complex inflammatory conditions. In this study, we investigated whether pro-autophagic γ-aminobutyric acid receptor-associated protein (GABARAP) promotes BMSCs proliferation and osteogenic differentiation by modulating autophagy in the presence or absence of interleukin-1 beta (IL-1β) in vitro. The expression levels of all relevant factors were evaluated by qRT-PCR or western blotting where appropriate. BMSCs differentiation were assessed by Alizarin Red, alkaline phosphatase, safranin O, and Oil Red O staining. Furthermore, the interactions between autophagy and osteogenic differentiation were investigated by co-treatment with the autophagy inhibitor 3-methyladenine (3-MA). As the results, we found that treatment with recombinant human His6-GABARAP protein promoted cell proliferation, inhibited apoptosis, and reduced ROS generation by increasing autophagic activity, particularly when co-cultured with IL-1β. Moreover, His6-GABARAP could effectively increase the osteogenic differentiation of BMSCs. The expression levels of inflammatory factors were significantly decreased by His6-GABARAP treatment, whereas its protective effects were attenuated by 3-MA. This study demonstrates that GABARAP maintains BMSCs survival and strengthens their osteogenic differentiation in an inflammatory environment by upregulating mediators of the autophagy pathway.
Collapse
Affiliation(s)
- Xiaobo Guo
- Department of Orthopedics, Jincheng General Hospital, Jincheng, 048000, China.
| | - Zhenyuan Wu
- Department of Orthopedics, Jincheng General Hospital, Jincheng, 048000, China
| |
Collapse
|
26
|
Šimoliūnas E, Ivanauskienė I, Bagdzevičiūtė L, Rinkūnaitė I, Alksnė M, Baltriukienė D. Surface stiffness depended gingival mesenchymal stem cell sensitivity to oxidative stress. Free Radic Biol Med 2021; 169:62-73. [PMID: 33862162 DOI: 10.1016/j.freeradbiomed.2021.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in the fields of cell therapy and tissue engineering, due to their wide spectrum of differentiation potential, immunomodulation function and ongoing oxidative stress (OS) reduction. Nevertheless, OS impact is often overlooked in these research fields. It is not only responsible for the induction and development of many ailments, e.g., diabetes, lung fibrosis, and cancer, moreover, OS causes stem cell death and senescence during cell therapy and tissue engineering practices. As MSCs are used to treat various tissues, they interact with different tissue-specific mechanical environments, thus it is important to understand how the mechanical environment impacts MSC sensitivity to OS. In this work, for the first time, as known to the authors, it was shown that gingival MSCs (GMSCs) sensitivity to OS depends on the stiffness of the surface, on which the cells are grown. Furthermore, the activity and expression of mitogen activated protein kinases ERK, JNK, and p38 were surface stiffness dependent. GMSCs isolated from intermediate/stiff gingiva tissue (~20 kPa) have shown the best proliferative and survival properties, then grown on the stiffest tissues mimicking polyacrylamide hydrogels (40 kPa). Therefore, MSC source might determine their sensitivity to OS in different stiffness environments and should be accounted when developing a treatment strategy.
Collapse
Affiliation(s)
- Egidijus Šimoliūnas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Indrė Ivanauskienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Lina Bagdzevičiūtė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Rinkūnaitė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Milda Alksnė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daiva Baltriukienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
27
|
Involvement of the long noncoding RNA H19 in osteogenic differentiation and bone regeneration. Stem Cell Res Ther 2021; 12:74. [PMID: 33478579 PMCID: PMC7819155 DOI: 10.1186/s13287-021-02149-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Osteogenic differentiation and bone regeneration are complex processes involving multiple genes and multiple steps. In this review, we summarize the effects of the long noncoding RNA (lncRNA) H19 on osteogenic differentiation. Osteogenic differentiation includes matrix secretion and calcium mineralization as hallmarks of osteoblast differentiation and the absorption of calcium and phosphorus as hallmarks of osteoclast differentiation. Mesenchymal stem cells (MSCs) form osteoprogenitor cells, pre-osteoblasts, mature osteoblasts, and osteocytes through induction and differentiation. lncRNAs regulate the expression of coding genes and play essential roles in osteogenic differentiation and bone regeneration. The lncRNA H19 is known to have vital roles in osteogenic induction. This review highlights the role of H19 as a novel target for osteogenic differentiation and the promotion of bone regeneration.
Collapse
|
28
|
Zhang W, Huang G, Xu F. Engineering Biomaterials and Approaches for Mechanical Stretching of Cells in Three Dimensions. Front Bioeng Biotechnol 2020; 8:589590. [PMID: 33154967 PMCID: PMC7591716 DOI: 10.3389/fbioe.2020.589590] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Mechanical stretch is widely experienced by cells of different tissues in the human body and plays critical roles in regulating their behaviors. Numerous studies have been devoted to investigating the responses of cells to mechanical stretch, providing us with fruitful findings. However, these findings have been mostly observed from two-dimensional studies and increasing evidence suggests that cells in three dimensions may behave more closely to their in vivo behaviors. While significant efforts and progresses have been made in the engineering of biomaterials and approaches for mechanical stretching of cells in three dimensions, much work remains to be done. Here, we briefly review the state-of-the-art researches in this area, with focus on discussing biomaterial considerations and stretching approaches. We envision that with the development of advanced biomaterials, actuators and microengineering technologies, more versatile and predictive three-dimensional cell stretching models would be available soon for extensive applications in such fields as mechanobiology, tissue engineering, and drug screening.
Collapse
Affiliation(s)
- Weiwei Zhang
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Guoyou Huang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing University, Chongqing, China
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center, Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
29
|
Tseng SJ, Wu CC, Cheng CH, Lin JC. Studies of surface grafted collagen and transforming growth factor β1 combined with cyclic stretching as a dual chemical and physical stimuli approach for rat adipose-derived stem cells (rADSCs) chondrogenesis differentiation. J Mech Behav Biomed Mater 2020; 112:104062. [PMID: 32891975 DOI: 10.1016/j.jmbbm.2020.104062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
The adipose-derived stem cell has been used in various regenerative medicine research due to its multiple differentiation capabilities. Developing a stable and quick approach for the differentiation of stem cells is a critical issue in tissue regenerative field. In this investigation, rat adipose-derived stem cells (rADSCs) were seeded onto the type I collagen/transforming growth factor β1 (TGF-β1) immobilized polydimethylsiloxane (PDMS) substrate and then combined with short term dynamic stretching stimulation (intermittent or continuous stretching for 6 h) to induce the rADSCs chondrogenesis differentiation using the induction medium without growth factors added in vitro. Via regulating the extracellular chemical- and mechano-receptors of the cultured rADSCs, the chondrogenic differentiation was examined. After 72 h of static culture, proteoglycan secretion was noted on the surfaces modified by collagen with or without TGF-β1. After different stretching stimulations, significant proteoglycan secretion was noted on the surface modified by both collagen and collagen/TGF-β1, especially after the intermittent stretching culturing. Nonetheless, genetic expression of the chondrogenic markers: SOX-9, Col2a1, and aggrecan, instead, were dependent upon the surface grafted layer and the stretching mode utilized. These findings suggested that the surface chemical characteristics and external mechanical stimulation could synergistically affect the efficacy of chondrogenic differentiation of rADSCs.
Collapse
Affiliation(s)
- Shen-Jui Tseng
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chi-Hui Cheng
- Department of Pediatrics, College of Medicine, Chang Gung University, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Jui-Che Lin
- Department of Chemical Engineering, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
30
|
Zhu H, Yang H, Ma Y, Lu TJ, Xu F, Genin GM, Lin M. Spatiotemporally Controlled Photoresponsive Hydrogels: Design and Predictive Modeling from Processing through Application. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2000639. [PMID: 32802013 PMCID: PMC7418561 DOI: 10.1002/adfm.202000639] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/16/2020] [Indexed: 05/16/2023]
Abstract
Photoresponsive hydrogels (PRHs) are soft materials whose mechanical and chemical properties can be tuned spatially and temporally with relative ease. Both photo-crosslinkable and photodegradable hydrogels find utility in a range of biomedical applications that require tissue-like properties or programmable responses. Progress in engineering with PRHs is facilitated by the development of theoretical tools that enable optimization of their photochemistry, polymer matrices, nanofillers, and architecture. This review brings together models and design principles that enable key applications of PRHs in tissue engineering, drug delivery, and soft robotics, and highlights ongoing challenges in both modeling and application.
Collapse
Affiliation(s)
- Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Haiqian Yang
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical StructuresNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
- MOE Key Laboratory for Multifunctional Materials and StructuresXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Guy M. Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
- Department of Mechanical Engineering & Materials ScienceWashington University in St. LouisSt. LouisMO63130USA
- NSF Science and Technology Center for Engineering MechanobiologyWashington University in St. LouisSt. LouisMO63130USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| |
Collapse
|