1
|
Pérez-Araluce M, Cianciosi A, Iglesias-García O, Jüngst T, Sanmartín C, Navarro-Blasco Í, Prósper F, Plano D, Mazo MM. Pristine Photopolymerizable Gelatin Hydrogels: A Low-Cost and Easily Modifiable Platform for Biomedical Applications. Antioxidants (Basel) 2024; 13:1238. [PMID: 39456491 PMCID: PMC11505247 DOI: 10.3390/antiox13101238] [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: 09/05/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The study addresses the challenge of temperature sensitivity in pristine gelatin hydrogels, widely used in biomedical applications due to their biocompatibility, low cost, and cell adhesion properties. Traditional gelatin hydrogels dissolve at physiological temperatures, limiting their utility. Here, we introduce a novel method for creating stable hydrogels at 37 °C using pristine gelatin through photopolymerization without requiring chemical modifications. This approach enhances consistency and simplifies production and functionalization of the gelatin with bioactive molecules. The stabilization mechanism involves the partial retention of the triple-helix structure of gelatin below 25 °C, which provides specific crosslinking sites. Upon activation by visible light, ruthenium (Ru) acts as a photosensitizer that generates sulphate radicals from sodium persulphate (SPS), inducing covalent bonding between tyrosine residues and "locking" the triple-helix conformation. The primary focus of this work is the characterization of the mechanical properties, swelling ratio, and biocompatibility of the photopolymerized gelatin hydrogels. Notably, these hydrogels supported better cell viability and elongation in normal human dermal fibroblasts (NHDFs) compared to GelMA, and similar performance was observed for human pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). As a proof of concept for functionalization, gelatin was modified with selenous acid (GelSe), which demonstrated antioxidant and antimicrobial capacities, particularly against E. coli and S. aureus. These results suggest that pristine gelatin hydrogels, enhanced through this new photopolymerization method and functionalized with bioactive molecules, hold potential for advancing regenerative medicine and tissue engineering by providing robust, biocompatible scaffolds for cell culture and therapeutic applications.
Collapse
Affiliation(s)
- Maria Pérez-Araluce
- Biomedical Engineering Program, Enabling Technologies Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain; (M.P.-A.); (O.I.-G.)
| | - Alessandro Cianciosi
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication, University of Würzburg, 97070 Würzburg, Germany; (A.C.); (T.J.)
- Bavarian Polymer Institute, University of Bayreuth, 95447 Bayreuth, Germany
| | - Olalla Iglesias-García
- Biomedical Engineering Program, Enabling Technologies Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain; (M.P.-A.); (O.I.-G.)
| | - Tomasz Jüngst
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication, University of Würzburg, 97070 Würzburg, Germany; (A.C.); (T.J.)
- Bavarian Polymer Institute, University of Bayreuth, 95447 Bayreuth, Germany
| | - Carmen Sanmartín
- Department of Pharmaceutical Sciences, Universidad de Navarra, 31008 Pamplona, Spain;
| | | | - Felipe Prósper
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC) CB16/12/00489, 28029 Madrid, Spain
- Hemato-Oncology Program, Cancer Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Sciences, Universidad de Navarra, 31008 Pamplona, Spain;
| | - Manuel M. Mazo
- Biomedical Engineering Program, Enabling Technologies Division, CIMA Universidad de Navarra, 31008 Pamplona, Spain; (M.P.-A.); (O.I.-G.)
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| |
Collapse
|
2
|
Mierke CT. Bioprinting of Cells, Organoids and Organs-on-a-Chip Together with Hydrogels Improves Structural and Mechanical Cues. Cells 2024; 13:1638. [PMID: 39404401 PMCID: PMC11476109 DOI: 10.3390/cells13191638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
The 3D bioprinting technique has made enormous progress in tissue engineering, regenerative medicine and research into diseases such as cancer. Apart from individual cells, a collection of cells, such as organoids, can be printed in combination with various hydrogels. It can be hypothesized that 3D bioprinting will even become a promising tool for mechanobiological analyses of cells, organoids and their matrix environments in highly defined and precisely structured 3D environments, in which the mechanical properties of the cell environment can be individually adjusted. Mechanical obstacles or bead markers can be integrated into bioprinted samples to analyze mechanical deformations and forces within these bioprinted constructs, such as 3D organoids, and to perform biophysical analysis in complex 3D systems, which are still not standard techniques. The review highlights the advances of 3D and 4D printing technologies in integrating mechanobiological cues so that the next step will be a detailed analysis of key future biophysical research directions in organoid generation for the development of disease model systems, tissue regeneration and drug testing from a biophysical perspective. Finally, the review highlights the combination of bioprinted hydrogels, such as pure natural or synthetic hydrogels and mixtures, with organoids, organoid-cell co-cultures, organ-on-a-chip systems and organoid-organ-on-a chip combinations and introduces the use of assembloids to determine the mutual interactions of different cell types and cell-matrix interferences in specific biological and mechanical environments.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
3
|
Pucha SA, Hasson M, Solomon H, McColgan GE, Robinson JL, Vega SL, Patel JM. Revealing Early Spatial Patterns of Cellular Responsivity in Fiber-Reinforced Microenvironments. Tissue Eng Part A 2024; 30:614-626. [PMID: 38517095 DOI: 10.1089/ten.tea.2024.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Fiber-reinforcement approaches have been used to replace aligned tissues with engineered constructs after injury or surgical resection, strengthening soft biomaterial scaffolds and replicating anisotropic, load-bearing properties. However, most studies focus on the macroscale aspects of these scaffolds, rarely considering the cell-biomaterial interactions that govern remodeling and extracellular matrix organization toward aligned neo-tissues. As initial cell-biomaterial responses within fiber-reinforced microenvironments likely influence the long-term efficacy of repair and regeneration strategies, here we elucidate the roles of spatial orientation, substrate stiffness, and matrix remodeling on early cell-fiber interactions. Bovine mesenchymal stromal cells (MSCs) were cultured in soft fibrin gels reinforced with a stiff 100 µm polyglycolide-co-caprolactone fiber. Gel stiffness and remodeling capacity were modulated by fibrinogen concentration and aprotinin treatment, respectively. MSCs were imaged at 3 days and evaluated for morphology, mechanoresponsiveness (nuclear Yes-associated protein [YAP] localization), and spatial features including distance and angle deviation from fiber. Within these constructs, morphological conformity decreased as a function of distance from fiber. However, these correlations were weak (R2 = 0.01043 for conformity and R2 = 0.05542 for nuclear YAP localization), illustrating cellular heterogeneity within fiber-enforced microenvironments. To better assess cell-fiber interactions, we applied machine-learning strategies to our heterogeneous dataset of cell-shape and mechanoresponsive parameters. Principal component analysis (PCA) was used to project 23 input parameters (not including distance) onto 5 principal components (PCs), followed by agglomerative hierarchical clustering to classify cells into 3 groups. These clusters exhibited distinct levels of morpho-mechanoresponse (combination of morphological conformity and YAP signaling) and were classified as high response (HR), medium response (MR), and low response (LR) clusters. Cluster distribution varied spatially, with most cells (61%) closest to the fiber (0-75 µm) belonging to the HR cluster, and most cells (55%) furthest from the fiber (225-300 µm) belonging to the LR cluster. Modulation of gel stiffness and fibrin remodeling showed differential effects for HR cells, with stiffness influencing the level of mechanoresponse and remodeling capacity influencing the location of responding cells. Together, these novel findings demonstrate early trends in cellular patterning of the fiber-reinforced microenvironment, showing how spatial orientation, substrate biophysical properties, and matrix remodeling may guide the amplitude and localization of cellular mechanoresponses. These trends may guide approaches to optimize the design of microscale scaffold architecture and substrate properties for enhancing organized tissue assembly at the macroscale.
Collapse
Affiliation(s)
- Saitheja A Pucha
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Maddie Hasson
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Hanna Solomon
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Gail E McColgan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Jennifer L Robinson
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, USA
- Department of Mechanical Engineering, University of Washington, Seattle, Washington, USA
| | - Sebastián L Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
- Department of Orthopaedic Surgery, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Jay M Patel
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Veterans Affairs, Atlanta VA Medical Center, Decatur, Georgia, USA
| |
Collapse
|
4
|
Liu W, Hsieh HT, He Z, Xiao X, Song C, Lee EX, Dong J, Lei CL, Wang J, Chen G. Medium acidosis drives cardiac differentiation during mesendoderm cell fate specification from human pluripotent stem cells. Stem Cell Reports 2024; 19:1304-1319. [PMID: 39178847 PMCID: PMC11411300 DOI: 10.1016/j.stemcr.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/26/2024] Open
Abstract
Effective lineage-specific differentiation is essential to fulfilling the great potentials of human pluripotent stem cells (hPSCs). In this report, we investigate how modulation of medium pH and associated metabolic changes influence mesendoderm differentiation from hPSCs. We show that daily medium pH fluctuations are critical for the heterogeneity of cell fates in the absence of exogenous inducers. Acidic environment alone leads to cardiomyocyte generation without other signaling modulators. In contrast, medium alkalinization is inhibitory to cardiac fate even in the presence of classic cardiac inducers. We then demonstrate that acidic environment suppresses glycolysis to facilitate cardiac differentiation, while alkaline condition promotes glycolysis and diverts the differentiation toward other cell types. We further show that glycolysis inhibition or AMPK activation can rescue cardiac differentiation under alkalinization, and glycolysis inhibition alone can drive cardiac cell fate. This study highlights that pH changes remodel metabolic patterns and modulate signaling pathways to control cell fate.
Collapse
Affiliation(s)
- Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China; Biological Imaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Hsun-Ting Hsieh
- Biological Imaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ziqing He
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China; GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Xia Xiao
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chengcheng Song
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - En Xin Lee
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Ji Dong
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Chon Lok Lei
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Jiaxian Wang
- HELP Stem Cell Innovations Ltd. Co., Nanjing, Jiangsu, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau SAR, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau SAR, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, China.
| |
Collapse
|
5
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [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: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
6
|
Moheimani H, Stealey S, Neal S, Ferchichi E, Zhang J, Foston M, Setton LA, Genin GM, Huebsch N, Zustiak SP. Tunable Viscoelasticity of Alginate Hydrogels via Serial Autoclaving. Adv Healthc Mater 2024:e2401550. [PMID: 39075933 DOI: 10.1002/adhm.202401550] [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: 07/01/2024] [Revised: 07/02/2024] [Indexed: 07/31/2024]
Abstract
Alginate hydrogels are widely used as biomaterials for cell culture and tissue engineering due to their biocompatibility and tunable mechanical properties. Reducing alginate molecular weight is an effective strategy for modulating hydrogel viscoelasticity and stress relaxation behavior, which can significantly impact cell spreading and fate. However, current methods like gamma irradiation to produce low molecular weight alginates suffer from high cost and limited accessibility. Here, a facile and cost-effective approach to reduce alginate molecular weight in a highly controlled manner using serial autoclaving is presented. Increasing the number of autoclave cycles results in proportional reductions in intrinsic viscosity, hydrodynamic radius, and molecular weight of the polymer while maintaining its chemical composition. Hydrogels fabricated from mixtures of the autoclaved alginates exhibit tunable mechanical properties, with inclusion of lower molecular weight alginate leading to softer gels with faster stress relaxation behaviors. The method is demonstrated by establishing how viscoelastic relaxation affects the spreading of encapsulated fibroblasts and glioblastoma cells. Results establish repetitive autoclaving as an easily accessible technique to generate alginates with a range of molecular weights and to control the viscoelastic properties of alginate hydrogels, and demonstrate utility across applications in mechanobiology, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Hamidreza Moheimani
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Samuel Stealey
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO, 63103, USA
| | - Sydney Neal
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Eya Ferchichi
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Jialiang Zhang
- Department of Energy, Environmental & Chemical Engineering, Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Marcus Foston
- Department of Energy, Environmental & Chemical Engineering, Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Lori A Setton
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Guy M Genin
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, Saint Louis, MO, 63130, USA
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Nathaniel Huebsch
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, Saint Louis, MO, 63130, USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Silviya P Zustiak
- NSF Science and Technology Center for Engineering MechanoBiology (CEMB), Washington University in Saint Louis, Saint Louis, MO, 63130, USA
- Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, Saint Louis, MO, 63103, USA
| |
Collapse
|
7
|
Kandasamy A, Yeh YT, Serrano R, Mercola M, Del Álamo JC. Uncertainty-Aware Traction Force Microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602172. [PMID: 39026786 PMCID: PMC11257441 DOI: 10.1101/2024.07.05.602172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Traction Force Microscopy (TFM) is a versatile tool to quantify cell-exerted forces by imaging and tracking fiduciary markers embedded in elastic substrates. The computations involved in TFM are ill-conditioned, and data smoothing or regularization is required to avoid overfitting the noise in the tracked substrate displacements. Most TFM calculations depend critically on the heuristic selection of regularization (hyper)parameters affecting the balance between overfitting and smoothing. However, TFM methods rarely estimate or account for measurement errors in substrate deformation to adjust the regularization level accordingly. Moreover, there is a lack of tools to quantify how these errors propagate to the recovered traction stresses. These limitations make it difficult to interpret TFM readouts and hinder comparing different experiments. This manuscript presents an uncertainty-aware TFM technique that estimates the variability in the magnitude and direction of the traction stress vector recovered at each point in space and time of each experiment. In this technique, substrate deformation and its uncertainty are quantified using a non-parametric bootstrap PIV method by resampling the microscopy image pixels (PIV-UQ). This information is passed to a hierarchical Bayesian framework that automatically selects its hyperparameters to perform spatially adaptive regularization conditioned on image quality and propagates the uncertainty to the traction stress readouts (TFM-UQ). We validate the performance of PIV-UQ and TFM-UQ using synthetic datasets with prescribed image quality variations and demonstrate the application of PIV-UQ and TFM-UQ to experimental datasets. These studies show that TFM-UQ locally adapts the level of smoothing, outperforming traditional regularization methods. They also illustrate how uncertainty-aware TFM tools can be used to objectively choose key image analysis parameters like PIV-UQ interrogation window size. We anticipate that these tools will allow for decoupling biological heterogeneity from measurement variability and facilitate automating the analysis of large datasets by parameter-free, input data-based regularization.
Collapse
|
8
|
Alasaadi DN, Mayor R. Mechanically guided cell fate determination in early development. Cell Mol Life Sci 2024; 81:242. [PMID: 38811420 PMCID: PMC11136904 DOI: 10.1007/s00018-024-05272-6] [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: 02/01/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024]
Abstract
Cell fate determination, a vital process in early development and adulthood, has been the focal point of intensive investigation over the past decades. Its importance lies in its critical role in shaping various and diverse cell types during embryonic development and beyond. Exploration of cell fate determination started with molecular and genetic investigations unveiling central signaling pathways and molecular regulatory networks. The molecular studies into cell fate determination yielded an overwhelming amount of information invoking the notion of the complexity of cell fate determination. However, recent advances in the framework of biomechanics have introduced a paradigm shift in our understanding of this intricate process. The physical forces and biochemical interplay, known as mechanotransduction, have been identified as a pivotal drive influencing cell fate decisions. Certainly, the integration of biomechanics into the process of cell fate pushed our understanding of the developmental process and potentially holds promise for therapeutic applications. This integration was achieved by identifying physical forces like hydrostatic pressure, fluid dynamics, tissue stiffness, and topography, among others, and examining their interplay with biochemical signals. This review focuses on recent advances investigating the relationship between physical cues and biochemical signals that control cell fate determination during early embryonic development.
Collapse
Affiliation(s)
- Delan N Alasaadi
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
9
|
Mendoza-Cerezo L, Rodríguez-Rego JM, Macías-García A, Callejas-Marín A, Sánchez-Guardado L, Marcos-Romero AC. Three-Dimensional Bioprinting of GelMA Hydrogels with Culture Medium: Balancing Printability, Rheology and Cell Viability for Tissue Regeneration. Polymers (Basel) 2024; 16:1437. [PMID: 38794630 PMCID: PMC11124935 DOI: 10.3390/polym16101437] [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: 03/27/2024] [Revised: 04/14/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Three-dimensional extrusion bioprinting technology aims to become a fundamental tool for tissue regeneration using cell-loaded hydrogels. These biomaterials must have highly specific mechanical and biological properties that allow them to generate biosimilar structures by successive layering of material while maintaining cell viability. The rheological properties of hydrogels used as bioinks are critical to their printability. Correct printability of hydrogels allows the replication of biomimetic structures, which are of great use in medicine, tissue engineering and other fields of study that require the three-dimensional replication of different tissues. When bioprinting cell-loaded hydrogels, a small amount of culture medium can be added to ensure adequate survival, which can modify the rheological properties of the hydrogels. GelMA is a hydrogel used in bioprinting, with very interesting properties and rheological parameters that have been studied and defined for its basic formulation. However, the changes that occur in its rheological parameters and therefore in its printability, when it is mixed with the culture medium necessary to house the cells inside, are unknown. Therefore, in this work, a comparative study of GelMA 100% and GelMA in the proportions 3:1 (GelMA 75%) and 1:1 (GelMA 50%) with culture medium was carried out to determine the printability of the gel (using a device of our own invention), its main rheological parameters and its toxicity after the addition of the medium and to observe whether significant differences in cell viability occur. This raises the possibility of its use in regenerative medicine using a 3D extrusion bioprinter.
Collapse
Affiliation(s)
- Laura Mendoza-Cerezo
- Department of Graphic Expression, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (L.M.-C.); (A.C.M.-R.)
| | - Jesús M. Rodríguez-Rego
- Department of Graphic Expression, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (L.M.-C.); (A.C.M.-R.)
| | - Antonio Macías-García
- Department of Mechanical, Energy and Materials Engineering, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain;
| | - Antuca Callejas-Marín
- Department of Anatomy, Cell Biology and Zoology, Faculty of Science, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (A.C.-M.); (L.S.-G.)
| | - Luís Sánchez-Guardado
- Department of Anatomy, Cell Biology and Zoology, Faculty of Science, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (A.C.-M.); (L.S.-G.)
| | - Alfonso C. Marcos-Romero
- Department of Graphic Expression, School of Industrial Engineering, University of Extremadura, Avenida de Elvas, s/n, 06006 Badajoz, Spain; (L.M.-C.); (A.C.M.-R.)
| |
Collapse
|
10
|
Ledwon JK, Applebaum SA, Progri B, Han T, Vignesh O, Gutowski KS, Chang AB, Reddy NK, Tepole AB, Gosain AK. Acellular Dermal Matrix Cover Improves Skin Growth during Tissue Expansion by Affecting Distribution of Mechanical Forces. Plast Reconstr Surg 2024; 153:663e-672e. [PMID: 37220332 DOI: 10.1097/prs.0000000000010709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
BACKGROUND Biological cover over tissue expander prostheses has been introduced to provide soft-tissue support for tissue expanders during breast reconstruction. However, its impact on mechanically induced skin growth remains unknown. This study investigates the hypothesis that covering the tissue expander with acellular dermal matrix (ADM) affects mechanotransduction without compromising the efficacy of tissue expansion. METHODS Tissue expansion, with and without use of ADM, was performed on a porcine model. The tissue expanders were inflated twice with 45 mL of saline, and the full-thickness skin biopsy specimens were harvested from expanded and control unexpanded skin 1 week and 8 weeks after the final inflation. Histologic evaluation, immunohistochemistry staining, and gene expression analysis were performed. Skin growth and total deformation were evaluated using isogeometric analysis. RESULTS The authors' results demonstrate that use of ADM as a biological cover during tissue expansion does not impede mechanotransduction that leads to skin growth and blood vessel formation. Isogeometric analysis revealed similar total deformation and growth of expanded skin with and without a biological cover, confirming that its use does not inhibit mechanically induced skin growth. In addition, the authors found that use of an ADM cover results in more uniform distribution of mechanical forces applied by the tissue expander. CONCLUSIONS These results suggest that ADM improves mechanically induced skin growth during tissue expansion by facilitating a more uniform distribution of mechanical forces applied by the tissue expander. Therefore, the use of a biological cover has potential to improve outcomes in tissue expansion-based reconstruction.
Collapse
Affiliation(s)
- Joanna K Ledwon
- Northwestern University Feinberg School of Medicine, Division of Plastic Surgery, Chicago, IL, USA
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Sarah A Applebaum
- Northwestern University Feinberg School of Medicine, Division of Plastic Surgery, Chicago, IL, USA
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Bianka Progri
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Tianhong Han
- Purdue University, Department of Mechanical Engineering, West Lafayette, IN, USA
| | - Oveyaa Vignesh
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Kristof S Gutowski
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Alec B Chang
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Narainsai K Reddy
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Adrian B Tepole
- Purdue University, Department of Mechanical Engineering, West Lafayette, IN, USA
| | - Arun K Gosain
- Northwestern University Feinberg School of Medicine, Division of Plastic Surgery, Chicago, IL, USA
- Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| |
Collapse
|
11
|
Kaku M, Thant L, Dobashi A, Ono Y, Kitami M, Mizukoshi M, Arai M, Iwama H, Kitami K, Kakihara Y, Matsumoto M, Saito I, Uoshima K. Multiomics analysis of cultured mouse periodontal ligament cell-derived extracellular matrix. Sci Rep 2024; 14:354. [PMID: 38172274 PMCID: PMC10764881 DOI: 10.1038/s41598-023-51054-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024] Open
Abstract
A comprehensive understanding of the extracellular matrix (ECM) is essential for developing biomimetic ECM scaffolds for tissue regeneration. As the periodontal ligament cell (PDLC)-derived ECM has shown potential for periodontal tissue regeneration, it is vital to gain a deeper understanding of its comprehensive profile. Although the PDLC-derived ECM exhibits extracellular environment similar to that of periodontal ligament (PDL) tissue, details of its molecular composition are lacking. Thus, using a multiomics approach, we systematically analyzed cultured mouse PDLC-derived ECM and compared it to mouse PDL tissue as a reference. Proteomic analysis revealed that, compared to PDL tissue, the cultured PDLC-derived ECM had a lower proportion of fibrillar collagens with increased levels of glycoprotein, corresponding to an immature ECM status. The gene expression signature was maintained in cultured PDLCs and was similar to that in cells from PDL tissues, with additional characteristics representative of naturally occurring progenitor cells. A combination of proteomic and transcriptomic analyses revealed that the cultured mouse PDLC-derived ECM has multiple advantages in tissue regeneration, providing an extracellular environment that closely mimics the environment in the native PDL tissue. These findings provide valuable insights for understanding PDLC-derived ECM and should contribute to the development of biomimetic ECM scaffolds for reliable periodontal tissue regeneration.
Collapse
Affiliation(s)
- Masaru Kaku
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, 2-5274, Gakkocho-dori, Chuo-ku, Niigata, Niigata, 951-8514, Japan.
| | - Lay Thant
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Dental Pharmacology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Azusa Dobashi
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoshiki Ono
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Megumi Kitami
- Division of Dental Pharmacology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaru Mizukoshi
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Moe Arai
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hajime Iwama
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kohei Kitami
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yoshito Kakihara
- Division of Dental Pharmacology, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Isao Saito
- Division of Orthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsumi Uoshima
- Division of Bio-Prosthodontics, Faculty of Dentistry and Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
12
|
Arcuri S, Pennarossa G, Ledda S, Gandolfi F, Brevini TAL. Use of Epigenetic Cues and Mechanical Stimuli to Generate Blastocyst-Like Structures from Mammalian Skin Dermal Fibroblasts. Methods Mol Biol 2024; 2767:161-173. [PMID: 37199907 DOI: 10.1007/7651_2023_486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Mammalian embryogenesis is characterized by complex interactions between embryonic and extra-embryonic tissues that coordinate morphogenesis, coupling bio-mechanical and bio-chemical cues, to regulate gene expression and influence cell fate. Deciphering such mechanisms is essential to understand early embryogenesis, as well as to harness differentiation disorders. Currently, several early developmental events remain unclear, mainly due to ethical and technical limitations related to the use of natural embryos.Here, we describe a three-step approach to generate 3D spherical structures, arbitrarily defined "epiBlastoids," whose phenotype is remarkably similar to natural embryos. In the first step, adult dermal fibroblasts are converted into trophoblast-like cells, combining the use of 5-azacytidine, to erase the original cell phenotype, with an ad hoc induction protocol, to drive erased cells into the trophoblast lineage. In the second step, once again epigenetic erasing is applied, in combination with mechanosensing-related cues, to generate inner cell mass (ICM)-like spheroids. More specifically, erased cells are encapsulated in micro-bioreactors to promote 3D cell rearrangement and boost pluripotency. In the third step, chemically induced trophoblast-like cells and ICM-like spheroids are co-cultured in the same micro-bioreactors. The newly generated embryoids are then transferred to microwells, to encourage further differentiation and favor epiBlastoid formation. The procedure here described is a novel strategy for in vitro generation of 3D spherical structures, phenotypically similar to natural embryos. The use of easily accessible dermal fibroblasts and the lack of retroviral gene transfection make this protocol a promising strategy to study early embryogenesis as well as embryo disorders.
Collapse
Affiliation(s)
- Sharon Arcuri
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Centre for Stem Cell Research, Università degli Studi di Milano, Lodi, Italy
| | - Georgia Pennarossa
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Centre for Stem Cell Research, Università degli Studi di Milano, Lodi, Italy
| | - Sergio Ledda
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy, Centre for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| | - Tiziana A L Brevini
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Centre for Stem Cell Research, Università degli Studi di Milano, Lodi, Italy
| |
Collapse
|
13
|
Wang S, Wang H, Lu P, Gong L, Gu X, Li M. Mechanisms underlying the cell-matrixed nerve grafts repairing peripheral nerve defects. Bioact Mater 2024; 31:563-577. [PMID: 37753326 PMCID: PMC10518682 DOI: 10.1016/j.bioactmat.2023.09.002] [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: 04/23/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
Decellularized extracellular matrix (dECM), with its distinct biological properties, has gained significant attention as a natural biomaterial. Leveraging its potentials, we successfully developed a three-dimensional matrix-based oriented nerve graft by encapsulating a fibrous scaffold with multilayered conformationally intact and biologically active human bone marrow mesenchymal stem cell-derived decellularized extracellular matrix (hBMSC-dECM). Convincingly, the hBMSC-dECM group exhibited comparable functional recoveries to the autograft group by postoperative week 12. In the comprehensive analysis, the molecular regulations in the hBMSC-dECM group were more intricate and nuanced compared to the autograft group. Nevertheless, both groups displayed similar molecular regulatory processes in terms of vascularization and extracellular matrix. Notably, the hBMSC-dECM group demonstrated sustained high levels of regulation in axon and myelin regeneration at week 12, while the immunomodulation returned to the normal levels after peaking at week 2. Collectively, our findings illustrated the satisfactory construction of a cell-matrixed nerve graft that established a microenvironment conducive to nerve regeneration, and elucidated the distinct molecular regulation patterns and characteristics associated with different repair modes.
Collapse
Affiliation(s)
- Shanshan Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, 226001, PR China
- Department of Obstetrics and Gynecology , Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, PR China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, 226001, PR China
| | - Panjian Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, 226001, PR China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, 226001, PR China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, 226001, PR China
| | - Meiyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, 226001, PR China
| |
Collapse
|
14
|
Huang Y, Lee S, Liu W, Takayama S, Jia S. OctoShaker: A versatile robotic biomechanical agitator for cellular and organoid research. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2023; 94:124104. [PMID: 38126811 PMCID: PMC10746356 DOI: 10.1063/5.0174526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023]
Abstract
Mechanical forces have increasingly been recognized as a key regulator in the fate of cellular development and functionality. Different mechanical transduction methods, such as substrate stiffness and magnetic bead vibration, have been experimented with to understand the interaction between the biophysical cues and cellular outcome. In the exploration and utilization of the intrinsic cellular mechanism, bio-shakers, traditionally invented for stirring liquid, have garnered more interest as a tool to provide precise mechanical stimuli to aid in this study. Nonetheless, despite the usefulness of current bio-shaking technology, each type of shaker often offers a single mode of motion, insufficient for generating complex force dynamics needed to resemble the actual physical condition that occurs inside living organisms. In this study, we present OctoShaker, a robotic instrument capable of creating a multitude of motions that could be sequenced or programmed to mimic sophisticated hemodynamics in vivo. We demonstrated the programmed motion of circular convection and investigated its influence on micro-particle distribution in 96-well culture microplates. Biological samples, including HeLa cells and organoids, were tested, and unique resultant patterns were observed. We anticipate the open-source dissemination of OctoShaker in diverse biological applications, encompassing biomechanical studies for cellular and organoid research, as well as other disciplines that demand dynamic mechanical force generation.
Collapse
Affiliation(s)
- Yan Huang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Soojung Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | | | - Shu Jia
- Author to whom correspondence should be addressed:
| |
Collapse
|
15
|
Cyr JA, Colzani M, Bayraktar S, Köhne M, Bax DV, Graup V, Farndale R, Sinha S, Best SM, Cameron RE. Extracellular macrostructure anisotropy improves cardiac tissue-like construct function and phenotypic cellular maturation. BIOMATERIALS ADVANCES 2023; 155:213680. [PMID: 37944449 DOI: 10.1016/j.bioadv.2023.213680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/02/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Regenerative cardiac tissue is a promising field of study with translational potential as a therapeutic option for myocardial repair after injury, however, poor electrical and contractile function has limited translational utility. Emerging research suggests scaffolds that recapitulate the structure of the native myocardium improve physiological function. Engineered cardiac constructs with anisotropic extracellular architecture demonstrate improved tissue contractility, signaling synchronicity, and cellular organization when compared to constructs with reduced architectural order. The complexity of scaffold fabrication, however, limits isolated variation of individual structural and mechanical characteristics. Thus, the isolated impact of scaffold macroarchitecture on tissue function is poorly understood. Here, we produce isotropic and aligned collagen scaffolds seeded with embryonic stem cell derived cardiomyocytes (hESC-CM) while conserving all confounding physio-mechanical features to independently assess the effects of macroarchitecture on tissue function. We quantified spatiotemporal tissue function through calcium signaling and contractile strain. We further examined intercellular organization and intracellular development. Aligned tissue constructs facilitated improved signaling synchronicity and directional contractility as well as dictated uniform cellular alignment. Cells on aligned constructs also displayed phenotypic and genetic markers of increased maturity. Our results isolate the influence of scaffold macrostructure on tissue function and inform the design of optimized cardiac tissue for regenerative and model medical systems.
Collapse
Affiliation(s)
- Jamie A Cyr
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Maria Colzani
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Semih Bayraktar
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Maria Köhne
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Daniel V Bax
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Vera Graup
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Richard Farndale
- Department of Biochemistry, Cambridge University, Hopkins Building Tennis Court Road, Cambridge CB2 1QW, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK.
| | - Serena M Best
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Ruth E Cameron
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| |
Collapse
|
16
|
Islam MS, Molley TG, Hung TT, Sathish CI, Putra VDL, Jalandhra GK, Ireland J, Li Y, Yi J, Kruzic JJ, Kilian KA. Magnetic Nanofibrous Hydrogels for Dynamic Control of Stem Cell Differentiation. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37643902 DOI: 10.1021/acsami.3c07021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The extracellular matrix in tissue consists of complex heterogeneous soft materials with hierarchical structure and dynamic mechanical properties dictating cell and tissue level function. In many natural matrices, there are nanofibrous structures that serve to guide cell activity and dictate the form and function of tissue. Synthetic hydrogels with integrated nanofibers can mimic the structural properties of native tissue; however, model systems with dynamic mechanical properties remain elusive. Here we demonstrate modular nanofibrous hydrogels that can be reversibly stiffened in response to applied magnetic fields. Iron oxide nanoparticles were incorporated into gelatin nanofibers through electrospinning, followed by chemical stabilization and fragmentation. These magnetoactive nanofibers can be mixed with virtually any hydrogel material and reversibly stiffen the matrix at a low fiber content (≤3%). In contrast to previous work, where a large quantity of magnetic material disallowed cell encapsulation, the low nanofiber content allows matrix stiffening with cells in 3D. Using adipose derived stem cells, we show how nanofibrous matrices are beneficial for both osteogenesis and adipogenesis, where stiffening the hydrogel with applied magnetic fields enhances osteogenesis while discouraging adipogenesis. Skeletal myoblast progenitors were used as a model of tissue morphogenesis with matrix stiffening augmenting myogenesis and multinucleated myotube formation. The ability to reversibly stiffen fibrous hydrogels through magnetic stimulation provides a useful tool for studying nanotopography and dynamic mechanics in cell culture, with a scope for stimuli responsive materials for tissue engineering.
Collapse
Affiliation(s)
- Md Shariful Islam
- School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Thomas G Molley
- School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Tzong-Tyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - C I Sathish
- School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Vina D L Putra
- School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Gagan K Jalandhra
- School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Jake Ireland
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Yancheng Li
- School of Civil and Environmental Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Jiabao Yi
- School of Engineering, College of Engineering, Science and Environment, The University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - Jamie J Kruzic
- School of Mechanical and Manufacturing Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| | - Kristopher A Kilian
- School of Materials Science and Engineering, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
- School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales (UNSW Sydney), Sydney, New South Wales 2052, Australia
| |
Collapse
|
17
|
Coscarella IL, Landim-Vieira M, Rastegarpouyani H, Chase PB, Irianto J, Pinto JR. Nucleus Mechanosensing in Cardiomyocytes. Int J Mol Sci 2023; 24:13341. [PMID: 37686151 PMCID: PMC10487505 DOI: 10.3390/ijms241713341] [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: 07/14/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Cardiac muscle contraction is distinct from the contraction of other muscle types. The heart continuously undergoes contraction-relaxation cycles throughout an animal's lifespan. It must respond to constantly varying physical and energetic burdens over the short term on a beat-to-beat basis and relies on different mechanisms over the long term. Muscle contractility is based on actin and myosin interactions that are regulated by cytoplasmic calcium ions. Genetic variants of sarcomeric proteins can lead to the pathophysiological development of cardiac dysfunction. The sarcomere is physically connected to other cytoskeletal components. Actin filaments, microtubules and desmin proteins are responsible for these interactions. Therefore, mechanical as well as biochemical signals from sarcomeric contractions are transmitted to and sensed by other parts of the cardiomyocyte, particularly the nucleus which can respond to these stimuli. Proteins anchored to the nuclear envelope display a broad response which remodels the structure of the nucleus. In this review, we examine the central aspects of mechanotransduction in the cardiomyocyte where the transmission of mechanical signals to the nucleus can result in changes in gene expression and nucleus morphology. The correlation of nucleus sensing and dysfunction of sarcomeric proteins may assist the understanding of a wide range of functional responses in the progress of cardiomyopathic diseases.
Collapse
Affiliation(s)
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Hosna Rastegarpouyani
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
- Institute for Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Prescott Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
18
|
Shang Y, Liu R, Gan J, Yang Y, Sun L. Construction of cardiac fibrosis for biomedical research. SMART MEDICINE 2023; 2:e20230020. [PMID: 39188350 PMCID: PMC11235890 DOI: 10.1002/smmd.20230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/22/2023] [Indexed: 08/28/2024]
Abstract
Cardiac remodeling is critical for effective tissue recuperation, nevertheless, excessive formation and deposition of extracellular matrix components can result in the onset of cardiac fibrosis. Despite the emergence of novel therapies, there are still no lifelong therapeutic solutions for this issue. Understanding the detrimental cardiac remodeling may aid in the development of innovative treatment strategies to prevent or reverse fibrotic alterations in the heart. Further combining the latest understanding of disease pathogenesis with cardiac tissue engineering has provided the conversion of basic laboratory studies into the therapy of cardiac fibrosis patients as an increasingly viable prospect. This review presents the current main mechanisms and the potential tissue engineering of cardiac fibrosis. Approaches using biomedical materials-based cardiac constructions are reviewed to consider key issues for simulating in vitro cardiac fibrosis, outlining a future perspective for preclinical applications.
Collapse
Affiliation(s)
- Yixuan Shang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rui Liu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jingjing Gan
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yuzhi Yang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
19
|
Solowiej-Wedderburn J, Dunlop CM. Cell-strain-energy costs of active control of contractility. Phys Rev E 2023; 107:L062401. [PMID: 37464714 DOI: 10.1103/physreve.107.l062401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
Cell mechanosensing is implicated in the control of a broad range of cell behaviors, with cytoskeletal contractility a key component. Experimentally, it is observed that the contractility of the cell responds to increasing substrate stiffness, showing increased contractile force and changing the distribution of cytoskeletal elements. Here, we show using a theoretical model of active cell contractility that upregulation of contractility need not be energetically expensive, especially when combined with changes in adhesion and contractile distribution. Indeed, we show that a feedback mechanism based on the maintenance of strain energy would require an upregulation in contractile pressure on all but the softest substrates. We consider both the commonly reported substrate strain energy and active work done. We demonstrate substrate strain energy would preferentially select for the experimentally observed clustering of cell adhesions on stiffer substrates which effectively soften the substrate and enable an upregulation of total contractile pressure, while the localization of contractility has the greatest impact on the internal work.
Collapse
Affiliation(s)
| | - Carina M Dunlop
- School of Mathematics and Physics, University of Surrey, Guildford GU2 7XH, United Kingdom
| |
Collapse
|
20
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
21
|
De Vincentiis S, Baggiani M, Merighi F, Cappello V, Lopane J, Di Caprio M, Costa M, Mainardi M, Onorati M, Raffa V. Low Forces Push the Maturation of Neural Precursors into Neurons. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2205871. [PMID: 37058009 DOI: 10.1002/smll.202205871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 03/15/2023] [Indexed: 06/19/2023]
Abstract
Mechanical stimulation modulates neural development and neuronal activity. In a previous study, magnetic "nano-pulling" is proposed as a tool to generate active forces. By loading neural cells with magnetic nanoparticles (MNPs), a precise force vector is remotely generated through static magnetic fields. In the present study, human neural stem cells (NSCs) are subjected to a standard differentiation protocol, in the presence or absence of nano-pulling. Under mechanical stimulation, an increase in the length of the neural processes which showed an enrichment in microtubules, endoplasmic reticulum, and mitochondria is found. A stimulation lasting up to 82 days induces a strong remodeling at the level of synapse density and a re-organization of the neuronal network, halving the time required for the maturation of neural precursors into neurons. The MNP-loaded NSCs are then transplanted into mouse spinal cord organotypic slices, demonstrating that nano-pulling stimulates the elongation of the NSC processes and modulates their orientation even in an ex vivo model. Thus, it is shown that active mechanical stimuli can guide the outgrowth of NSCs transplanted into the spinal cord tissue. The findings suggest that mechanical forces play an important role in neuronal maturation which could be applied in regenerative medicine.
Collapse
Affiliation(s)
| | - Matteo Baggiani
- Department of Biology, Università di Pisa, Pisa, 56127, Italy
| | | | - Valentina Cappello
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, Pontedera, 56025, Italy
| | - Jakub Lopane
- Department of Biology, Università di Pisa, Pisa, 56127, Italy
| | - Mariachiara Di Caprio
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri 7, Pisa, 56126, Italy
| | - Mario Costa
- Neuroscience Institute, National Research Council, via Giuseppe Moruzzi 1, Pisa, 56124, Italy
| | - Marco Mainardi
- Neuroscience Institute, National Research Council, via Giuseppe Moruzzi 1, Pisa, 56124, Italy
| | - Marco Onorati
- Department of Biology, Università di Pisa, Pisa, 56127, Italy
| | - Vittoria Raffa
- Department of Biology, Università di Pisa, Pisa, 56127, Italy
| |
Collapse
|
22
|
Karaz S, Senses E. Liposomes Under Shear: Structure, Dynamics, and Drug Delivery Applications. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Selcan Karaz
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| | - Erkan Senses
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| |
Collapse
|
23
|
Tanaka HY, Nakazawa T, Enomoto A, Masamune A, Kano MR. Therapeutic Strategies to Overcome Fibrotic Barriers to Nanomedicine in the Pancreatic Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15030724. [PMID: 36765684 PMCID: PMC9913712 DOI: 10.3390/cancers15030724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Pancreatic cancer is notorious for its dismal prognosis. The enhanced permeability and retention (EPR) effect theory posits that nanomedicines (therapeutics in the size range of approximately 10-200 nm) selectively accumulate in tumors. Nanomedicine has thus been suggested to be the "magic bullet"-both effective and safe-to treat pancreatic cancer. However, the densely fibrotic tumor microenvironment of pancreatic cancer impedes nanomedicine delivery. The EPR effect is thus insufficient to achieve a significant therapeutic effect. Intratumoral fibrosis is chiefly driven by aberrantly activated fibroblasts and the extracellular matrix (ECM) components secreted. Fibroblast and ECM abnormalities offer various potential targets for therapeutic intervention. In this review, we detail the diverse strategies being tested to overcome the fibrotic barriers to nanomedicine in pancreatic cancer. Strategies that target the fibrotic tissue/process are discussed first, which are followed by strategies to optimize nanomedicine design. We provide an overview of how a deeper understanding, increasingly at single-cell resolution, of fibroblast biology is revealing the complex role of the fibrotic stroma in pancreatic cancer pathogenesis and consider the therapeutic implications. Finally, we discuss critical gaps in our understanding and how we might better formulate strategies to successfully overcome the fibrotic barriers in pancreatic cancer.
Collapse
Affiliation(s)
- Hiroyoshi Y. Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Takuya Nakazawa
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya-shi 466-8550, Aichi, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai-shi 980-8574, Miyagi, Japan
| | - Mitsunobu R. Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
- Correspondence:
| |
Collapse
|
24
|
Tassinari R, Olivi E, Cavallini C, Taglioli V, Zannini C, Marcuzzi M, Fedchenko O, Ventura C. Mechanobiology: A landscape for reinterpreting stem cell heterogeneity and regenerative potential in diseased tissues. iScience 2022; 26:105875. [PMID: 36647385 PMCID: PMC9839966 DOI: 10.1016/j.isci.2022.105875] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mechanical forces play a fundamental role in cellular dynamics from the molecular level to the establishment of complex heterogeneity in somatic and stem cells. Here, we highlight the role of cytoskeletal mechanics and extracellular matrix in generating mechanical forces merging into oscillatory synchronized patterns. We discuss how cellular mechanosensing/-transduction can be modulated by mechanical forces to control tissue metabolism and set the basis for nonpharmacologic tissue rescue. Control of bone anabolic activity and repair, as well as obesity prevention, through a fine-tuning of the stem cell morphodynamics are highlighted. We also discuss the use of mechanical forces in the treatment of cardiovascular diseases and heart failure through the fine modulation of stem cell metabolic activity and regenerative potential. We finally focus on the new landscape of delivering specific mechanical stimuli to reprogram tissue-resident stem cells and enhance our self-healing potential, without the need for stem cell or tissue transplantation.
Collapse
Affiliation(s)
| | - Elena Olivi
- ELDOR LAB, via Corticella 183, 40129 Bologna, Italy
| | | | | | | | - Martina Marcuzzi
- NIBB, National Institute of Biostructures and Biosystems, National Laboratory of Molecular Biology and Stem Cell Engineering, via Corticella 183, 40129 Bologna, Italy
| | - Oleksandra Fedchenko
- NIBB, National Institute of Biostructures and Biosystems, National Laboratory of Molecular Biology and Stem Cell Engineering, via Corticella 183, 40129 Bologna, Italy
| | - Carlo Ventura
- ELDOR LAB, via Corticella 183, 40129 Bologna, Italy,NIBB, National Institute of Biostructures and Biosystems, National Laboratory of Molecular Biology and Stem Cell Engineering, via Corticella 183, 40129 Bologna, Italy,Corresponding author
| |
Collapse
|
25
|
Lammerding J, Engler AJ, Kamm R. Mechanobiology of the cell nucleus. APL Bioeng 2022; 6:040401. [PMID: 36536804 PMCID: PMC9759352 DOI: 10.1063/5.0135299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jan Lammerding
- Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853-6007, USA,Authors to whom correspondence should be addressed:; ; and
| | - Adam J. Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, California 92093-0412, USA,Authors to whom correspondence should be addressed:; ; and
| | - Roger Kamm
- Department of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA,Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
26
|
Spatio-Temporal Changes of Extracellular Matrix (ECM) Stiffness in the Development of the Leech Hirudo verbana. Int J Mol Sci 2022; 23:ijms232415953. [PMID: 36555595 PMCID: PMC9787456 DOI: 10.3390/ijms232415953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The invertebrate leech Hirudo verbana represents a powerful experimental animal model for improving the knowledge about the functional interaction between the extracellular matrix (ECM) and cells within the tissue microenvironment (TME), and the key role played by ECM stiffness during development and growth. Indeed, the medicinal leech is characterized by a simple anatomical organization reproducing many aspects of the basic biological processes of vertebrates and in which a rapid spatiotemporal development is well established and easily assessed. Our results show that ECM structural organization, as well as the amount of fibrillar and non-fibrillar collagen are deeply different from hatching leeches to adult ones. In addition, the changes in ECM remodelling occurring during the different leech developmental stages, leads to a gradient of stiffness regulating both the path of migratory cells and their fates. The ability of cells to perceive and respond to changes in ECM composition and mechanics strictly depend on nuclear or cytoplasmic expression of Yes-Associated Protein 1 (YAP1), a key mediator converting mechanical signals into transcriptional outputs, expression, and activation.
Collapse
|
27
|
Zhao Z, Chen X, Dowbaj AM, Sljukic A, Bratlie K, Lin L, Fong ELS, Balachander GM, Chen Z, Soragni A, Huch M, Zeng YA, Wang Q, Yu H. Organoids. NATURE REVIEWS. METHODS PRIMERS 2022; 2:94. [PMID: 37325195 PMCID: PMC10270325 DOI: 10.1038/s43586-022-00174-y] [Citation(s) in RCA: 243] [Impact Index Per Article: 121.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/28/2022] [Indexed: 06/17/2023]
Abstract
Organoids have attracted increasing attention because they are simple tissue-engineered cell-based in vitro models that recapitulate many aspects of the complex structure and function of the corresponding in vivo tissue. They can be dissected and interrogated for fundamental mechanistic studies on development, regeneration, and repair in human tissues. Organoids can also be used in diagnostics, disease modeling, drug discovery, and personalized medicine. Organoids are derived from either pluripotent or tissue-resident stem (embryonic or adult) or progenitor or differentiated cells from healthy or diseased tissues, such as tumors. To date, numerous organoid engineering strategies that support organoid culture and growth, proliferation, differentiation and maturation have been reported. This Primer serves to highlight the rationale underlying the selection and development of these materials and methods to control the cellular/tissue niche; and therefore, structure and function of the engineered organoid. We also discuss key considerations for generating robust organoids, such as those related to cell isolation and seeding, matrix and soluble factor selection, physical cues and integration. The general standards for data quality, reproducibility and deposition within the organoid community is also outlined. Lastly, we conclude by elaborating on the limitations of organoids in different applications, and key priorities in organoid engineering for the coming years.
Collapse
Affiliation(s)
- Zixuan Zhao
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xinyi Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Anna M. Dowbaj
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Aleksandra Sljukic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Kaitlin Bratlie
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, USA
| | - Luda Lin
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, California, USA
- Molecular Biology Institute, University of California Los Angeles, California, USA
| | - Eliza Li Shan Fong
- Translational Tumor Engineering Laboratory, Department of Biomedical Engineering, National University of Singapore, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore
| | - Gowri Manohari Balachander
- Department of Physiology, Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, Singapore
| | - Zhaowei Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Alice Soragni
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, California, USA
- Molecular Biology Institute, University of California Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, California, USA
- California NanoSystems Institute, University of California Los Angeles, California, USA
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, USA
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Physiology, Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, Singapore
- Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, Singapore
| |
Collapse
|
28
|
Hashimoto M, Takeichi K, Murata K, Kozakai A, Yagi A, Ishikawa K, Suzuki-Nakagawa C, Kasuya Y, Fukamizu A, Nakagawa T. Regulation of neural stem cell proliferation and survival by protein arginine methyltransferase 1. Front Neurosci 2022; 16:948517. [PMID: 36440275 PMCID: PMC9685794 DOI: 10.3389/fnins.2022.948517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/17/2022] [Indexed: 12/22/2024] Open
Abstract
Protein arginine methyltransferase 1 (PRMT1), a major type I arginine methyltransferase in mammals, methylates histone and non-histone proteins to regulate various cellular functions, such as transcription, DNA damage response, and signal transduction. PRMT1 is highly expressed in neural stem cells (NSCs) and embryonic brains, suggesting that PRMT1 is essential for early brain development. Although our previous reports have shown that PRMT1 positively regulates oligodendrocyte development, it has not been studied whether PRMT1 regulates NSC proliferation and its survival during development. To examine the role of PRMT1 in NSC activity, we cultured NSCs prepared from embryonic mouse forebrains deficient in PRMT1 specific for NSCs and performed neurosphere assays. We found that the primary neurospheres of PRMT1-deficient NSCs were small and the number of spheres was decreased, compared to those of control NSCs. Primary neurospheres deficient in PRMT1 expressed an increased level of cleaved caspase-3, suggesting that PRMT1 deficiency-induced apoptosis. Furthermore, p53 protein was significantly accumulated in PRMT1-deficient NSCs. In parallel, p53-responsive pro-apoptotic genes including Pmaip1 and Perp were upregulated in PRMT1-deficient NSCs. p53-target p21 mRNA and its protein levels were shown to be upregulated in PRMT1-deficient NSCs. Moreover, the 5-bromo-2'-deoxyuridine (BrdU) incorporation assay showed that the loss of PRMT1 led to cell cycle defects in the embryonic NSCs. In contrast to the above in vitro observations, NSCs normally proliferated and survived in the fetal brains of NSC-specific PRMT1-deficient mice. We also found that Lama1, which encodes the laminin subunit α1, was significantly upregulated in the embryonic brains of PRMT1-deficient mice. These data implicate that extracellular factors provided by neighboring cells in the microenvironment gave a trophic support to NSCs in the PRMT1-deficient brain and recovered NSC activity to maintain brain homeostasis. Our study implies that PRMT1 plays a cell-autonomous role in the survival and proliferation of embryonic NSCs.
Collapse
Affiliation(s)
- Misuzu Hashimoto
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kaho Takeichi
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kazuya Murata
- Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Aoi Kozakai
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Atsushi Yagi
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Kohei Ishikawa
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Chiharu Suzuki-Nakagawa
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Yoshitoshi Kasuya
- Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
- World Premier International Research Center Initiative, International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Tsutomu Nakagawa
- Laboratory of Biological Chemistry, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
29
|
Bohere J, Eldridge-Thomas BL, Kolahgar G. Vinculin recruitment to α-catenin halts the differentiation and maturation of enterocyte progenitors to maintain homeostasis of the Drosophila intestine. eLife 2022; 11:e72836. [PMID: 36269226 PMCID: PMC9586559 DOI: 10.7554/elife.72836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/11/2022] [Indexed: 11/23/2022] Open
Abstract
Mechanisms communicating changes in tissue stiffness and size are particularly relevant in the intestine because it is subject to constant mechanical stresses caused by peristalsis of its variable content. Using the Drosophila intestinal epithelium, we investigate the role of vinculin, one of the best characterised mechanoeffectors, which functions in both cadherin and integrin adhesion complexes. We discovered that vinculin regulates cell fate decisions, by preventing precocious activation and differentiation of intestinal progenitors into absorptive cells. It achieves this in concert with α-catenin at sites of cadherin adhesion, rather than as part of integrin function. Following asymmetric division of the stem cell into a stem cell and an enteroblast (EB), the two cells initially remain connected by adherens junctions, where vinculin is required, only on the EB side, to maintain the EB in a quiescent state and inhibit further divisions of the stem cell. By manipulating cell tension, we show that vinculin recruitment to adherens junction regulates EB activation and numbers. Consequently, removing vinculin results in an enlarged gut with improved resistance to starvation. Thus, mechanical regulation at the contact between stem cells and their progeny is used to control tissue cell number.
Collapse
Affiliation(s)
- Jerome Bohere
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| | - Buffy L Eldridge-Thomas
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| | - Golnar Kolahgar
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
30
|
Picchio V, Floris E, Derevyanchuk Y, Cozzolino C, Messina E, Pagano F, Chimenti I, Gaetani R. Multicellular 3D Models for the Study of Cardiac Fibrosis. Int J Mol Sci 2022; 23:ijms231911642. [PMID: 36232943 PMCID: PMC9569892 DOI: 10.3390/ijms231911642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Ex vivo modelling systems for cardiovascular research are becoming increasingly important in reducing lab animal use and boosting personalized medicine approaches. Integrating multiple cell types in complex setups adds a higher level of significance to the models, simulating the intricate intercellular communication of the microenvironment in vivo. Cardiac fibrosis represents a key pathogenetic step in multiple cardiovascular diseases, such as ischemic and diabetic cardiomyopathies. Indeed, allowing inter-cellular interactions between cardiac stromal cells, endothelial cells, cardiomyocytes, and/or immune cells in dedicated systems could make ex vivo models of cardiac fibrosis even more relevant. Moreover, culture systems with 3D architectures further enrich the physiological significance of such in vitro models. In this review, we provide a summary of the multicellular 3D models for the study of cardiac fibrosis described in the literature, such as spontaneous microtissues, bioprinted constructs, engineered tissues, and organs-on-chip, discussing their advantages and limitations. Important discoveries on the physiopathology of cardiac fibrosis, as well as the screening of novel potential therapeutic molecules, have been reported thanks to these systems. Future developments will certainly increase their translational impact for understanding and modulating mechanisms of cardiac fibrosis even further.
Collapse
Affiliation(s)
- Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Erica Floris
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Yuriy Derevyanchuk
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - Claudia Cozzolino
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Elisa Messina
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), 00015 Monterotondo, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
- Correspondence: ; Tel.: +39-077-3175-7234
| | - Roberto Gaetani
- Department of Molecular Medicine, Sapienza University, 00161 Rome, Italy
| |
Collapse
|
31
|
Ribeiro S, Pugliese E, Korntner SH, Fernandes EM, Gomes ME, Reis RL, O'Riordan A, Bayon Y, Zeugolis DI. Assessing the combined effect of surface topography and substrate rigidity in human bone marrow stem cell cultures. Eng Life Sci 2022; 22:619-633. [PMID: 36247829 PMCID: PMC9550738 DOI: 10.1002/elsc.202200029] [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: 05/31/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/11/2022] Open
Abstract
The combined effect of surface topography and substrate rigidity in stem cell cultures is still under-investigated, especially when biodegradable polymers are used. Herein, we assessed human bone marrow stem cell response on aliphatic polyester substrates as a function of anisotropic grooved topography and rigidity (7 and 12 kPa). Planar tissue culture plastic (TCP, 3 GPa) and aliphatic polyester substrates were used as controls. Cell morphology analysis revealed that grooved substrates caused nuclei orientation/alignment in the direction of the grooves. After 21 days in osteogenic and chondrogenic media, the 3 GPa TCP and the grooved 12 kPa substrate induced significantly higher calcium deposition and alkaline phosphatase (ALP) activity and glycosaminoglycan (GAG) deposition, respectively, than the other groups. After 14 days in tenogenic media, the 3 GPa TCP upregulated four and downregulated four genes; the planar 7 kPa substrate upregulated seven genes and downregulated one gene; and the grooved 12 kPa substrate upregulated seven genes and downregulated one gene. After 21 days in adipogenic media, the softest (7 kPa) substrates induced significantly higher oil droplet deposition than the other substrates and the grooved substrate induced significantly higher droplet deposition than the planar. Our data pave the way for more rational design of bioinspired constructs.
Collapse
Affiliation(s)
- Sofia Ribeiro
- MedtronicSofradim ProductionTrevouxFrance
- RegenerativeModular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM)National University of Ireland Galway (NUI Galway)GalwayIreland
| | - Eugenia Pugliese
- RegenerativeModular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM)National University of Ireland Galway (NUI Galway)GalwayIreland
| | - Stefanie H. Korntner
- RegenerativeModular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM)National University of Ireland Galway (NUI Galway)GalwayIreland
| | - Emanuel M. Fernandes
- 3B's Research GroupI3Bs – Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAveParkParque de Ciência e TecnologiaZona Industrial da GandraBarcoGuimarãesPortugal
- ICVS/3B's – PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Manuela E. Gomes
- 3B's Research GroupI3Bs – Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAveParkParque de Ciência e TecnologiaZona Industrial da GandraBarcoGuimarãesPortugal
- ICVS/3B's – PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on BiomaterialsBiodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineAveParkParque de Ciência e TecnologiaZona Industrial da GandraBarcoGuimarãesPortugal
- ICVS/3B's – PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | | | - Yves Bayon
- MedtronicSofradim ProductionTrevouxFrance
| | - Dimitrios I. Zeugolis
- RegenerativeModular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM)National University of Ireland Galway (NUI Galway)GalwayIreland
- RegenerativeModular & Developmental Engineering Laboratory (REMODEL)Charles Institute of DermatologyConway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials EngineeringUniversity College Dublin (UCD)DublinIreland
| |
Collapse
|
32
|
Shin W, Jeong S, Lee JU, Jeong SY, Shin J, Kim HH, Cheon J, Lee JH. Magnetogenetics with Piezo1 Mechanosensitive Ion Channel for CRISPR Gene Editing. NANO LETTERS 2022; 22:7415-7422. [PMID: 36069378 DOI: 10.1021/acs.nanolett.2c02314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Regulation of genetic activity in single cells and tissues is pivotal to determine key cellular functions in current biomedicine, yet the conventional biochemical activators lack spatiotemporal precision due to the diffusion-mediated slow kinetics and nonselectivity. Here, we describe a magnetogenetic method for target-specific activation of a clustered regularly interspaced short palindromic repeats (CRISPR) system for the regulation of intracellular proteins. We used magnetomechanical force generated by the magnetic nanostructure to activate pre-encoded Piezo1, the mechanosensitive ion channel, on the target cell. The activated Piezo1 further triggers the intracellular Ca2+ signaling pathway, inducing the pre-encoded genes to express genes of interest (GOIs), which is Cas9 protein for the CRISPR regulation of the target proteins. We demonstrated that this magnetogenetic CRISPR system successfully edits the target genome for both in vitro and pseudo-in vivo environments, providing a versatile magnetic platform for remote gene editing of animals with various size scales.
Collapse
Affiliation(s)
- Wookjin Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Sumin Jeong
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung-Uk Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Soo Yeun Jeong
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Jeonghong Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyongbum Henry Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
33
|
Harnessing conserved signaling and metabolic pathways to enhance the maturation of functional engineered tissues. NPJ Regen Med 2022; 7:44. [PMID: 36057642 PMCID: PMC9440900 DOI: 10.1038/s41536-022-00246-3] [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: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The development of induced-pluripotent stem cell (iPSC)-derived cell types offers promise for basic science, drug testing, disease modeling, personalized medicine, and translatable cell therapies across many tissue types. However, in practice many iPSC-derived cells have presented as immature in physiological function, and despite efforts to recapitulate adult maturity, most have yet to meet the necessary benchmarks for the intended tissues. Here, we summarize the available state of knowledge surrounding the physiological mechanisms underlying cell maturation in several key tissues. Common signaling consolidators, as well as potential synergies between critical signaling pathways are explored. Finally, current practices in physiologically relevant tissue engineering and experimental design are critically examined, with the goal of integrating greater decision paradigms and frameworks towards achieving efficient maturation strategies, which in turn may produce higher-valued iPSC-derived tissues.
Collapse
|
34
|
Jain N, Lord JM, Vogel V. Mechanoimmunology: Are inflammatory epigenetic states of macrophages tuned by biophysical factors? APL Bioeng 2022; 6:031502. [PMID: 36051106 PMCID: PMC9427154 DOI: 10.1063/5.0087699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Many inflammatory diseases that are responsible for a majority of deaths are still uncurable, in part as the underpinning pathomechanisms and how to combat them is still poorly understood. Tissue-resident macrophages play pivotal roles in the maintenance of tissue homeostasis, but if they gradually convert to proinflammatory phenotypes, or if blood-born proinflammatory macrophages persist long-term after activation, they contribute to chronic inflammation and fibrosis. While biochemical factors and how they regulate the inflammatory transcriptional response of macrophages have been at the forefront of research to identify targets for therapeutic interventions, evidence is increasing that physical factors also tune the macrophage phenotype. Recently, several mechanisms have emerged as to how physical factors impact the mechanobiology of macrophages, from the nuclear translocation of transcription factors to epigenetic modifications, perhaps even DNA methylation. Insight into the mechanobiology of macrophages and associated epigenetic modifications will deliver novel therapeutic options going forward, particularly in the context of increased inflammation with advancing age and age-related diseases. We review here how biophysical factors can co-regulate pro-inflammatory gene expression and epigenetic modifications and identify knowledge gaps that require urgent attention if this therapeutic potential is to be realized.
Collapse
Affiliation(s)
- Nikhil Jain
- Authors to whom correspondence should be addressed: and
| | | | - Viola Vogel
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Guglielmo M, Marta B. Stem Cells and the Microenvironment: Reciprocity with Asymmetry in Regenerative Medicine. Acta Biotheor 2022; 70:24. [PMID: 35962861 DOI: 10.1007/s10441-022-09448-0] [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: 11/17/2021] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022]
Abstract
Much of the current research in regenerative medicine concentrates on stem-cell therapy that exploits the regenerative capacities of stem cells when injected into different types of human tissues. Although new therapeutic paths have been opened up by induced pluripotent cells and human mesenchymal cells, the rate of success is still low and mainly due to the difficulties of managing cell proliferation and differentiation, giving rise to non-controlled stem cell differentiation that ultimately leads to cancer. Despite being still far from becoming a reality, these studies highlight the role of physical and biological constraints (e.g., cues and morphogenetic fields) placed by tissue microenvironment on stem cell fate. This asks for a clarification of the coupling of stem cells and microenvironmental factors in regenerative medicine. We argue that extracellular matrix and stem cells have a causal reciprocal and asymmetric relationship in that the 3D organization and composition of the extracellular matrix establish a spatial, temporal, and mechanical control over the fate of stem cells, which enable them to interact and control (as well as be controlled by) the cellular components and soluble factors of microenvironment. Such an account clarifies the notions of stemness and stem cell regeneration consistently with that of microenvironment.
Collapse
Affiliation(s)
- Militello Guglielmo
- IAS-Research Centre, University of the Basque Country, San Sebastián, Spain.
| | - Bertolaso Marta
- University Campus Bio-Medico of Rome, Institute of Scientific and Technological Practice, Rome, Italy
| |
Collapse
|
36
|
De Pieri A, Korntner SH, Capella-Monsonis H, Tsiapalis D, Kostjuk SV, Churbanov S, Timashev P, Gorelov A, Rochev Y, Zeugolis DI. Macromolecular crowding transforms regenerative medicine by enabling the accelerated development of functional and truly three-dimensional cell assembled micro tissues. Biomaterials 2022; 287:121674. [PMID: 35835003 DOI: 10.1016/j.biomaterials.2022.121674] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/03/2022] [Accepted: 07/06/2022] [Indexed: 11/22/2022]
Abstract
Scaffold-free in vitro organogenesis exploits the innate ability of cells to synthesise and deposit their own extracellular matrix to fabricate tissue-like assemblies. Unfortunately, cell-assembled tissue engineered concepts require prolonged ex vivo culture periods of very high cell numbers for the development of a borderline three-dimensional implantable device, which are associated with phenotypic drift and high manufacturing costs, thus, hindering their clinical translation and commercialisation. Herein, we report the accelerated (10 days) development of a truly three-dimensional (338.1 ± 42.9 μm) scaffold-free tissue equivalent that promotes fast wound healing and induces formation of neotissue composed of mature collagen fibres, using human adipose derived stem cells seeded at only 50,000 cells/cm2 on an poly (N-isopropylacrylamide-co-N-tert-butylacrylamide (PNIPAM86-NTBA14) temperature-responsive electrospun scaffold and grown under macromolecular crowding conditions (50 μg/ml carrageenan). Our data pave the path for a new era in scaffold-free regenerative medicine.
Collapse
Affiliation(s)
- Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Spiddal, Galway, Ireland
| | - Stefanie H Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Hector Capella-Monsonis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios Tsiapalis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Sergei V Kostjuk
- Department of Chemistry, Belarusian State University and Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus; Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Semyon Churbanov
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alexander Gorelov
- School of Chemistry & Chemical Biology, University College Dublin, Dublin, Ireland
| | - Yuri Rochev
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
37
|
Protick F, Amit SK, Amar K, Nath SD, Akand R, Davis VA, Nilufar S, Chowdhury F. Additive Manufacturing of Viscoelastic Polyacrylamide Substrates for Mechanosensing Studies. ACS OMEGA 2022; 7:24384-24395. [PMID: 35874232 PMCID: PMC9301700 DOI: 10.1021/acsomega.2c01817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Polymerized polyacrylamide (PAA) substrates are linearly elastic hydrogels that are widely used in mechanosensing studies due to their biocompatibility, wide range of functionalization capability, and tunable mechanical properties. However, such cellular response on purely elastic substrates, which do not mimic the viscoelastic living tissues, may not be physiologically relevant. Because the cellular response on 2D viscoelastic PAA substrates remains largely unknown, we used stereolithography (SLA)-based additive manufacturing technique to create viscoelastic PAA substrates with tunable mechanical properties that allow us to identify physiologically relevant cellular behaviors. Three PAA substrates of different complex moduli were fabricated by SLA. By embedding fluorescent markers during the additive manufacturing of the substrates, we show a homogeneous and uniform composition throughout, which conventional manufacturing techniques cannot produce. Rheological investigation of the additively manufactured PAA substrates shows a viscoelastic behavior with a 5-10% loss moduli compared to their elastic moduli, mimicking the living tissues. To understand the cell mechanosensing on the dissipative PAA substrates, single live cells were seeded on PAA substrates to establish the basic relationships between cell traction, cytoskeletal prestress, and cell spreading. With the increasing substrate moduli, we observed a concomitant increase in cellular traction and prestress, but not cell spreading, suggesting that cell spreading can be decoupled from traction and intracellular prestress in physiologically relevant environments. Together, additively manufactured PAA substrates fill the void of lacking real tissue like viscoelastic materials that can be used in a variety of mechanosensing studies with superior reproducibility.
Collapse
Affiliation(s)
- Fardeen
Kabir Protick
- School
of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, Illinois 62901, United States
| | - Sadat Kamal Amit
- Samuel
Ginn Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Kshitij Amar
- School
of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, Illinois 62901, United States
| | - Shukantu Dev Nath
- School
of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, Illinois 62901, United States
| | - Rafee Akand
- School
of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, Illinois 62901, United States
| | - Virginia A. Davis
- Samuel
Ginn Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Sabrina Nilufar
- School
of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, Illinois 62901, United States
| | - Farhan Chowdhury
- School
of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, Illinois 62901, United States
- Biomedical
Engineering Program, School of Electrical, Computer, and Biomedical
Engineering, Southern Illinois University
Carbondale, Carbondale, Illinois 62901, United
States
- Materials
Technology Center, Southern Illinois University
Carbondale, Carbondale, Illinois 62901, United
States
| |
Collapse
|
38
|
Guo T, He C, Venado A, Zhou Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr Physiol 2022; 12:3523-3558. [PMID: 35766837 PMCID: PMC10088466 DOI: 10.1002/cphy.c210032] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The extracellular matrix (ECM) provides structural support and imparts a wide variety of environmental cues to cells. In the past decade, a growing body of work revealed that the mechanical properties of the ECM, commonly known as matrix stiffness, regulate the fundamental cellular processes of the lung. There is growing appreciation that mechanical interplays between cells and associated ECM are essential to maintain lung homeostasis. Dysregulation of ECM-derived mechanical signaling via altered mechanosensing and mechanotransduction pathways is associated with many common lung diseases. Matrix stiffening is a hallmark of lung fibrosis. The stiffened ECM is not merely a sequelae of lung fibrosis but can actively drive the progression of fibrotic lung disease. In this article, we provide a comprehensive view on the role of matrix stiffness in lung health and disease. We begin by summarizing the effects of matrix stiffness on the function and behavior of various lung cell types and on regulation of biomolecule activity and key physiological processes, including host immune response and cellular metabolism. We discuss the potential mechanisms by which cells probe matrix stiffness and convert mechanical signals to regulate gene expression. We highlight the factors that govern matrix stiffness and outline the role of matrix stiffness in lung development and the pathogenesis of pulmonary fibrosis, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. We envision targeting of deleterious matrix mechanical cues for treatment of fibrotic lung disease. Advances in technologies for matrix stiffness measurements and design of stiffness-tunable matrix substrates are also explored. © 2022 American Physiological Society. Compr Physiol 12:3523-3558, 2022.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA.,Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Aida Venado
- Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
39
|
Thant L, Kaku M, Kakihara Y, Mizukoshi M, Kitami M, Arai M, Kitami K, Kobayashi D, Yoshida Y, Maeda T, Saito I, Uoshima K, Saeki M. Extracellular Matrix-Oriented Proteomic Analysis of Periodontal Ligament Under Mechanical Stress. Front Physiol 2022; 13:899699. [PMID: 35669581 PMCID: PMC9163570 DOI: 10.3389/fphys.2022.899699] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022] Open
Abstract
The periodontal ligament (PDL) is a specialized connective tissue that provides structural support to the tooth and is crucial for oral functions. The mechanical properties of the PDL are mainly derived from the tissue-specific composition and structural characteristics of the extracellular matrix (ECM). The ECM also plays key roles in determining cell fate in the cellular microenvironment thus crucial in the PDL tissue homeostasis. In the present study, we determined the comprehensive ECM profile of mouse molar PDL using laser microdissection and mass spectrometry-based proteomic analysis with ECM-oriented data curation. Additionally, we evaluated changes in the ECM proteome under mechanical loading using a mouse orthodontic tooth movement (OTM) model and analyzed potential regulatory networks using a bioinformatics approach. Proteomic changes were evaluated in reference to the novel second harmonic generation (SHG)-based fiber characterization. Our ECM-oriented proteomics approach succeeded in illustrating the comprehensive ECM profile of the mouse molar PDL. We revealed the presence of type II collagen in PDL, possibly associated with the load-bearing function upon occlusal force. Mechanical loading induced unique architectural changes in collagen fibers along with dynamic compositional changes in the matrisome profile, particularly involving ECM glycoproteins and matrisome-associated proteins. We identified several unique matrisome proteins which responded to the different modes of mechanical loading in PDL. Notably, the proportion of type VI collagen significantly increased at the mesial side, contributing to collagen fibrogenesis. On the other hand, type XII collagen increased at the PDL-cementum boundary of the distal side. Furthermore, a multifaceted bioinformatics approach illustrated the potential molecular cues, including PDGF signaling, that maintain ECM homeostasis under mechanical loading. Our findings provide fundamental insights into the molecular network underlying ECM homeostasis in PDL, which is vital for clinical diagnosis and development of biomimetic tissue-regeneration strategies.
Collapse
Affiliation(s)
- Lay Thant
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaru Kaku
- Division of Bio-prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- *Correspondence: Masaru Kaku,
| | - Yoshito Kakihara
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaru Mizukoshi
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Megumi Kitami
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Moe Arai
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kohei Kitami
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Daiki Kobayashi
- Omics Unit, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yutaka Yoshida
- Department of Structural Pathology, Kidney Research Center, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Isao Saito
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsumi Uoshima
- Division of Bio-prosthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Makio Saeki
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
40
|
Jun I, Li N, Shin J, Park J, Kim YJ, Jeon H, Choi H, Cho JG, Chan Choi B, Han HS, Song JJ. Synergistic stimulation of surface topography and biphasic electric current promotes muscle regeneration. Bioact Mater 2022; 11:118-129. [PMID: 34938917 PMCID: PMC8665271 DOI: 10.1016/j.bioactmat.2021.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/27/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Developing a universal culture platform that manipulates cell fate is one of the most important tasks in the investigation of the role of the cellular microenvironment. This study focuses on the application of topographical and electrical field stimuli to human myogenic precursor cell (hMPC) cultures to assess the influences of the adherent direction, proliferation, and differentiation, and induce preconditioning-induced therapeutic benefits. First, a topographical surface of commercially available culture dishes was achieved by femtosecond laser texturing. The detachable biphasic electrical current system was then applied to the hMPCs cultured on laser-textured culture dishes. Laser-textured topographies were remarkably effective in inducing the assembly of hMPC myotubes by enhancing the orientation of adherent hMPCs compared with flat surfaces. Furthermore, electrical field stimulation through laser-textured topographies was found to promote the expression of myogenic regulatory factors compared with nonstimulated cells. As such, we successfully demonstrated that the combined stimulation of topographical and electrical cues could effectively enhance the myogenic maturation of hMPCs in a surface spatial and electrical field-dependent manner, thus providing the basis for therapeutic strategies.
Collapse
Affiliation(s)
- Indong Jun
- Environmental Safety Group, Korea Institute of Science & Technology Europe (KIST-EUROPE), Saarbrücken, 66123, Germany
| | - Na Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jaehee Shin
- Department of Medical Sciences, Graduate School of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jaeho Park
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Republic of Korea
| | - Young Jun Kim
- Environmental Safety Group, Korea Institute of Science & Technology Europe (KIST-EUROPE), Saarbrücken, 66123, Germany
| | - Hojeong Jeon
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyuk Choi
- Department of Medical Sciences, Graduate School of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jae-Gu Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Byoung Chan Choi
- Laser Surface Texturing Group, AYECLUS, Gyeonggi-do, 14255, Republic of Korea
| | - Hyung-Seop Han
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| |
Collapse
|
41
|
Banerjee S, Nara R, Chakraborty S, Chowdhury D, Haldar S. Integrin Regulated Autoimmune Disorders: Understanding the Role of Mechanical Force in Autoimmunity. Front Cell Dev Biol 2022; 10:852878. [PMID: 35372360 PMCID: PMC8971850 DOI: 10.3389/fcell.2022.852878] [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: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
The pathophysiology of autoimmune disorders is multifactorial, where immune cell migration, adhesion, and lymphocyte activation play crucial roles in its progression. These immune processes are majorly regulated by adhesion molecules at cell–extracellular matrix (ECM) and cell–cell junctions. Integrin, a transmembrane focal adhesion protein, plays an indispensable role in these immune cell mechanisms. Notably, integrin is regulated by mechanical force and exhibit bidirectional force transmission from both the ECM and cytosol, regulating the immune processes. Recently, integrin mechanosensitivity has been reported in different immune cell processes; however, the underlying mechanics of these integrin-mediated mechanical processes in autoimmunity still remains elusive. In this review, we have discussed how integrin-mediated mechanotransduction could be a linchpin factor in the causation and progression of autoimmune disorders. We have provided an insight into how tissue stiffness exhibits a positive correlation with the autoimmune diseases’ prevalence. This provides a plausible connection between mechanical load and autoimmunity. Overall, gaining insight into the role of mechanical force in diverse immune cell processes and their dysregulation during autoimmune disorders will open a new horizon to understand this physiological anomaly.
Collapse
|
42
|
Aprile P, Whelan IT, Sathy BN, Carroll SF, Kelly DJ. Soft Hydrogel Environments that Facilitate Cell Spreading and Aggregation Preferentially Support Chondrogenesis of Adult Stem Cells. Macromol Biosci 2022; 22:e2100365. [PMID: 35171524 DOI: 10.1002/mabi.202100365] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Mesenchymal stem/stromal cells (MSCs) represent a promising cell type for treating damaged and diseased synovial joints. The therapeutic potential of MSCs will be facilitated by the engineering of biomaterial environments capable of directing their fate. Here we explored the interplay between matrix elasticity and cell morphology in regulating the chondrogenic differentiation of MSCs when seeded onto or encapsulated within hydrogels made of interpenetrating networks (IPN) of alginate and collagen type I. This IPN system enabled the independent control of substrate stiffness (in 2D and in 3D) and cell morphology (3D only). In a 2D culture environment, the expression of chondrogenic markers SOX9, ACAN and COL2 increased on a soft substrate, which correlated with increased SMAD2/3 nuclear localization, enhanced MSCs condensation and the formation of larger cellular aggregates. The encapsulation of spread MSCs within a soft IPN dramatically increased the expression of cartilage-specific genes, which was linked to higher levels of cellular condensation and nuclear SMAD2/3 localization. Surprisingly, cells forced to adopt a more rounded morphology within the same soft IPNs expressed higher levels of the osteogenic markers RUNX2 and COL1. The insight provided by this study suggests that a mechanobiology informed approach to biomaterial development will be integral to the development of successful cartilage tissue engineering strategies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Paola Aprile
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Ian T Whelan
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,CÚRAM Center for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Binulal N Sathy
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Centre for Nanoscience and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Simon F Carroll
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,CÚRAM Center for Research in Medical Devices, National University of Ireland, Galway, Ireland.,The Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Trinity College Dublin, Ireland
| |
Collapse
|
43
|
Veenvliet JV, Lenne PF, Turner DA, Nachman I, Trivedi V. Sculpting with stem cells: how models of embryo development take shape. Development 2021; 148:dev192914. [PMID: 34908102 PMCID: PMC8722391 DOI: 10.1242/dev.192914] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During embryogenesis, organisms acquire their shape given boundary conditions that impose geometrical, mechanical and biochemical constraints. A detailed integrative understanding how these morphogenetic information modules pattern and shape the mammalian embryo is still lacking, mostly owing to the inaccessibility of the embryo in vivo for direct observation and manipulation. These impediments are circumvented by the developmental engineering of embryo-like structures (stembryos) from pluripotent stem cells that are easy to access, track, manipulate and scale. Here, we explain how unlocking distinct levels of embryo-like architecture through controlled modulations of the cellular environment enables the identification of minimal sets of mechanical and biochemical inputs necessary to pattern and shape the mammalian embryo. We detail how this can be complemented with precise measurements and manipulations of tissue biochemistry, mechanics and geometry across spatial and temporal scales to provide insights into the mechanochemical feedback loops governing embryo morphogenesis. Finally, we discuss how, even in the absence of active manipulations, stembryos display intrinsic phenotypic variability that can be leveraged to define the constraints that ensure reproducible morphogenesis in vivo.
Collapse
Affiliation(s)
- Jesse V. Veenvliet
- Stembryogenesis Lab, Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, Ihnestrasse 63-73, 14195 Berlin, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany
| | - Pierre-François Lenne
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, 13288, Marseille, France
| | - David A. Turner
- Institute of Life Course and Medical Sciences, William Henry Duncan Building, University of Liverpool, Liverpool, L7 8TX, UK
| | - Iftach Nachman
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Vikas Trivedi
- European Molecular Biology Laboratories (EMBL), Barcelona, 08003, Spain
- EMBL Heidelberg, Developmental Biology Unit, 69117, Heidelberg, Germany
| |
Collapse
|
44
|
The Response of Corneal Endothelial Cells to Shear Stress in an In Vitro Flow Model. J Ophthalmol 2021; 2021:9217866. [PMID: 34873452 PMCID: PMC8643247 DOI: 10.1155/2021/9217866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose Corneal endothelial cells are usually exposed to shear stress caused by the aqueous humour, which is similar to the exposure of vascular endothelial cells to shear stress caused by blood flow. However, the effect of fluid shear stress on corneal endothelial cells is still poorly understood. The purpose of this study was to explore whether the shear stress that results from the aqueous humour influences corneal endothelial cells. Methods An in vitro model was established to generate fluid flow on cells, and the effect of fluid flow on corneal endothelial cells after exposure to two levels of shear stress for different durations was investigated. The mRNA and protein expression of corneal endothelium-related markers in rabbit corneal endothelial cells was evaluated by real-time PCR and western blotting. Results The expression of the corneal endothelium-related markers ZO-1, N-cadherin, and Na+-K+-ATPase in rabbit corneal endothelial cells (RCECs) was upregulated at both the mRNA and protein levels after exposure to shear stress. Conclusion This study demonstrates that RCECs respond favourably to fluid shear stress, which may contribute to the maintenance of corneal endothelial cell function. Furthermore, this study also provides a theoretical foundation for further investigating the response of human corneal endothelial cells to the shear stress caused by the aqueous humour.
Collapse
|
45
|
Valencia FR, Sandoval E, Du J, Iu E, Liu J, Plotnikov SV. Force-dependent activation of actin elongation factor mDia1 protects the cytoskeleton from mechanical damage and promotes stress fiber repair. Dev Cell 2021; 56:3288-3302.e5. [PMID: 34822787 DOI: 10.1016/j.devcel.2021.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/02/2021] [Accepted: 11/02/2021] [Indexed: 01/16/2023]
Abstract
Plasticity of cell mechanics underlies a wide range of cell and tissue behaviors allowing cells to migrate through narrow spaces, resist shear forces, and safeguard against mechanical damage. Such plasticity depends on spatiotemporal regulation of the actomyosin cytoskeleton, but mechanisms of adaptive change in cell mechanics remain elusive. Here, we report a mechanism of mechanically activated actin polymerization at focal adhesions (FAs), specifically requiring the actin elongation factor mDia1. By combining live-cell imaging with mathematical modeling, we show that actin polymerization at FAs exhibits pulsatile dynamics where spikes of mDia1 activity are triggered by contractile forces. The suppression of mDia1-mediated actin polymerization increases tension on stress fibers (SFs) leading to an increased frequency of spontaneous SF damage and decreased efficiency of zyxin-mediated SF repair. We conclude that tension-controlled actin polymerization acts as a safety valve dampening excessive tension on the actin cytoskeleton and safeguarding SFs against mechanical damage.
Collapse
Affiliation(s)
- Fernando R Valencia
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Eduardo Sandoval
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joy Du
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Ernest Iu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jian Liu
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
46
|
Zhang J, Wong SHD, Wu X, Lei H, Qin M, Shi P, Wang W, Bian L, Cao Y. Engineering Photoresponsive Ligand Tethers for Mechanical Regulation of Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2105765. [PMID: 34561928 DOI: 10.1002/adma.202105765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/26/2021] [Indexed: 06/13/2023]
Abstract
Regulating stem cell functions by precisely controlling the nanoscale presentation of bioactive ligands has a substantial impact on tissue engineering and regenerative medicine but remains a major challenge. Here it is shown that bioactive ligands can become mechanically "invisible" by increasing their tether lengths to the substrate beyond a critical length, providing a way to regulate mechanotransduction without changing the biochemical conditions. Building on this finding, light switchable tethers are rationally designed, whose lengths can be modulated reversibly by switching a light-responsive protein, pdDronpa, in between monomer and dimer states. This allows the regulation of the adhesion, spreading, and differentiation of stem cells by light on substrates of well-defined biochemical and physical properties. Spatiotemporal regulation of differential cell fates on the same substrate is further demonstrated, which may represent an important step toward constructing complex organoids or mini tissues by spatially defining the mechanical cues of the cellular microenvironment with light.
Collapse
Affiliation(s)
- Junsheng Zhang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Xin Wu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Hai Lei
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Meng Qin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Peng Shi
- School of Biomedical Sciences and EngineeringSouth China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Liming Bian
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
- School of Biomedical Sciences and EngineeringSouth China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
47
|
Yan H, Ramirez-Guerrero D, Lowengrub J, Wu M. Stress generation, relaxation and size control in confined tumor growth. PLoS Comput Biol 2021; 17:e1009701. [PMID: 34932555 PMCID: PMC8726498 DOI: 10.1371/journal.pcbi.1009701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 01/04/2022] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Experiments on tumor spheroids have shown that compressive stress from their environment can reversibly decrease tumor expansion rates and final sizes. Stress release experiments show that nonuniform anisotropic elastic stresses can be distributed throughout. The elastic stresses are maintained by structural proteins and adhesive molecules, and can be actively relaxed by a variety of biophysical processes. In this paper, we present a new continuum model to investigate how the growth-induced elastic stresses and active stress relaxation, in conjunction with cell size control feedback machinery, regulate the cell density and stress distributions within growing tumors as well as the tumor sizes in the presence of external physical confinement and gradients of growth-promoting chemical fields. We introduce an adaptive reference map that relates the current position with the reference position but adapts to the current position in the Eulerian frame (lab coordinates) via relaxation. This type of stress relaxation is similar to but simpler than the classical Maxwell model of viscoelasticity in its formulation. By fitting the model to experimental data from two independent studies of tumor spheroid growth and their cell density distributions, treating the tumors as incompressible, neo-Hookean elastic materials, we find that the rates of stress relaxation of tumor tissues can be comparable to volumetric growth rates. Our study provides insight on how the biophysical properties of the tumor and host microenvironment, mechanical feedback control and diffusion-limited differential growth act in concert to regulate spatial patterns of stress and growth. When the tumor is stiffer than the host, our model predicts tumors are more able to change their size and mechanical state autonomously, which may help to explain why increased tumor stiffness is an established hallmark of malignant tumors.
Collapse
Affiliation(s)
- Huaming Yan
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
| | - Daniel Ramirez-Guerrero
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
- Center for Multiscale Cell Fate Studies, University of California, Irvine, Irvine, California, United States of America
| | - John Lowengrub
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
- Center for Multiscale Cell Fate Studies, University of California, Irvine, Irvine, California, United States of America
- Department Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
| | - Min Wu
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
| |
Collapse
|
48
|
Petzold J, Gentleman E. Intrinsic Mechanical Cues and Their Impact on Stem Cells and Embryogenesis. Front Cell Dev Biol 2021; 9:761871. [PMID: 34820380 PMCID: PMC8606660 DOI: 10.3389/fcell.2021.761871] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Collapse
Affiliation(s)
- Jonna Petzold
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
49
|
Wang S, Zhu C, Zhang B, Hu J, Xu J, Xue C, Bao S, Gu X, Ding F, Yang Y, Gu X, Gu Y. BMSC-derived extracellular matrix better optimizes the microenvironment to support nerve regeneration. Biomaterials 2021; 280:121251. [PMID: 34810037 DOI: 10.1016/j.biomaterials.2021.121251] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022]
Abstract
A favorable microenvironment plays an important role in nerve regeneration. Extracellular matrix (ECM) derived from cultured cells or natural tissues can facilitate nerve regeneration in the presence of various microenvironmental cues, including biochemical, spatial, and biomechanical factors. This study, through proteomics and three-dimensional image analysis, determines that the components and spatial organization of the ECM secreted by bone marrow mesenchymal cells (BMSCs) are more similar to acellular nerves than those of the ECMs derived from Schwann cells (SCs), skin-derived precursor Schwann cells (SKP-SCs), or fibroblasts (FBs). ECM-modified nerve grafts (ECM-NGs) are engineered by co-cultivating BMSCs, SCs, FBs, SKP-SCs with well-designed nerve grafts used to bridge nerve defects. BMSC-ECM-NGs exhibit the most promising nerve repair properties based on the histology, neurophysiology, and behavioral analyses. The regeneration microenvironment formed by the ECM-NGs is also characterized by proteomics, and the advantages of BMSC-ECM-NGs are evidenced by the enhanced expression of factors related to neural regeneration and reduced immune response. Together, these findings indicate that BMSC-derived ECMs create a more superior microenvironment for nerve regeneration than that by the other ECMs and may, therefore, represent a potential alternative for the clinical repair of peripheral nerve defects.
Collapse
Affiliation(s)
- Shengran Wang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Changlai Zhu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Bin Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Junxia Hu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Jinghui Xu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Chengbin Xue
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Shuangxi Bao
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Xiaokun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Yumin Yang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Xiaosong Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China.
| | - Yun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China.
| |
Collapse
|
50
|
Shi X, Tang D, Xing Y, Zhao S, Fan C, Zhong J, Cui Y, Shi K, Jiu Y. Actin nucleator formins regulate the tension-buffering function of caveolin-1. J Mol Cell Biol 2021; 13:876-888. [PMID: 34718633 PMCID: PMC8800513 DOI: 10.1093/jmcb/mjab070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Both the mechanosensitive actin cytoskeleton and caveolae contribute to active processes such as cell migration, morphogenesis, and vesicular trafficking. Although distinct actin components are well studied, how they contribute to cytoplasmic caveolae, especially in the context of mechano-stress, has remained elusive. Here, we identify two actin-associated mobility stereotypes of caveolin-1 (CAV-1)-marked intracellular vesicles, which are characterized as ‘dwelling’ and ‘go and dwelling’. In order to exploit the reason for their distinct dynamics, elongated actin-associated formin functions are perturbed. We find drastically decreased density, increased clustering, and compromised motility of cytoplasmic CAV-1 vesicles resulting from lacking actin nucleator formins by both chemical treatment and RNA silencing of formin genes. Furthermore, hypo-osmosis-stimulated diminishing of CAV-1 is dramatically intensified upon blocking formins. The clustering of CAV-1 vesicles when cells are cultured on soft substrate is also aggravated under formin inhibition condition. Together, we reveal that actin-associated formins are essential for maintaining the dynamic organization of cytoplasmic CAV-1 and importantly its sensitivity upon mechanical challenge. We conclude that tension-controlled actin formins act as a safety valve dampening excessive tension on CAV-1 and safeguarding CAV-1 against mechanical damage.
Collapse
Affiliation(s)
- Xuemeng Shi
- The Joint Program in Infection and Immunity, a. Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 and b. Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Daijiao Tang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yifan Xing
- University of Chinese Academy of Sciences, Beijing, 100049 China.,Unit of Viral Hepatitis, CAS Key Laboratory of Molecular Virology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Shuangshuang Zhao
- The Joint Program in Infection and Immunity, a. Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 and b. Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Changyuan Fan
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jin Zhong
- University of Chinese Academy of Sciences, Beijing, 100049 China.,Unit of Viral Hepatitis, CAS Key Laboratory of Molecular Virology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yanqin Cui
- The Joint Program in Infection and Immunity, a. Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 and b. Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Kun Shi
- The Joint Program in Infection and Immunity, a. Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 and b. Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yaming Jiu
- The Joint Program in Infection and Immunity, a. Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 and b. Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China.,Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|