201
|
Wubshet NH, Arreguin-Martinez E, Nail M, Annamalai H, Koerner R, Rousseva M, Tom T, Gillespie RB, Liu AP. Simulating microgravity using a random positioning machine for inducing cellular responses to mechanotransduction in human osteoblasts. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2021; 92:114101. [PMID: 34852501 PMCID: PMC9643046 DOI: 10.1063/5.0056366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
The mechanotransduction pathways that mediate cellular responses to contact forces are better understood than those that mediate response to distance forces, especially the force of gravity. Removing or reducing gravity for significant periods of time involves either sending samples to space, inducing diamagnetic levitation with high magnetic fields, or continually reorienting samples for a period, all in a manner that supports cell culturing. Undesired secondary effects due to high magnetic fields or shear forces associated with fluid flow while reorienting must be considered in the design of ground-based devices. We have developed a lab-friendly and compact random positioning machine (RPM) that fits in a standard tissue culture incubator. Using a two-axis gimbal, it continually reorients samples in a manner that produces an equal likelihood that all possible orientations are visited. We contribute a new control algorithm by which the distribution of probabilities over all possible orientations is completely uniform. Rather than randomly varying gimbal axis speed and/or direction as in previous algorithms (which produces non-uniform probability distributions of orientation), we use inverse kinematics to follow a trajectory with a probability distribution of orientations that is uniform by construction. Over a time period of 6 h of operation using our RPM, the average gravity is within 0.001 23% of the gravity of Earth. Shear forces are minimized by limiting the angular speed of both gimbal motors to under 42 °/s. We demonstrate the utility of our RPM by investigating the effects of simulated microgravity on adherent human osteoblasts immediately after retrieving samples from our RPM. Cytoskeletal disruption and cell shape changes were observed relative to samples cultured in a 1 g environment. We also found that subjecting human osteoblasts in suspension to simulated microgravity resulted in less filamentous actin and lower cell stiffness.
Collapse
Affiliation(s)
- Nadab H. Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | - Hariprasad Annamalai
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Robert Koerner
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Maria Rousseva
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Tristan Tom
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | - Allen P. Liu
- Author to whom correspondence should be addressed: . Current address: University of Michigan, 2350 Hayward Street, Ann Arbor, Michigan 48109, USA. Tel.: +1 734-764-7719
| |
Collapse
|
202
|
Eugenis I, Wu D, Rando TA. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021; 278:121173. [PMID: 34619561 PMCID: PMC8556323 DOI: 10.1016/j.biomaterials.2021.121173] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/01/2021] [Accepted: 08/14/2021] [Indexed: 12/20/2022]
Abstract
Severe traumatic skeletal muscle injuries, such as volumetric muscle loss (VML), result in the obliteration of large amounts of skeletal muscle and lead to permanent functional impairment. Current clinical treatments are limited in their capacity to regenerate damaged muscle and restore tissue function, promoting the need for novel muscle regeneration strategies. Advances in tissue engineering, including cell therapy, scaffold design, and bioactive factor delivery, are promising solutions for VML therapy. Herein, we review tissue engineering strategies for regeneration of skeletal muscle, development of vasculature and nerve within the damaged muscle, and achievements in immunomodulation following VML. In addition, we discuss the limitations of current state of the art technologies and perspectives of tissue-engineered bioconstructs for muscle regeneration and functional recovery following VML.
Collapse
Affiliation(s)
- Ioannis Eugenis
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Di Wu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
203
|
Rahvar M, Ahmadi Lakalayeh G, Nazeri N, Marouf BT, Shirzad M, Najafi T Shabankareh A, Ghanbari H. Assessment of structural, biological and drug release properties of electro-sprayed poly lactic acid-dexamethasone coating for biomedical applications. Biomed Eng Lett 2021; 11:393-406. [PMID: 34616584 DOI: 10.1007/s13534-021-00205-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/28/2021] [Accepted: 08/20/2021] [Indexed: 10/20/2022] Open
Abstract
The efficacy of an implant is highly depends on its coating characteristics mainly determined by polymer properties and coating technique. Electro-spraying is an inexpensive and versatile coating technique with various advantages for biomedical application. In this study, the efficacy of electro-sprayed (ES) poly lactic acid (PLA)-dexamethasone (DEX) coatings for medical implants was evaluated and compared with spin-coated samples as control. Structural properties of coatings were investigated using X-ray diffraction (XRD) and differential scanning calorimetry (DSC). Confocal and scanning electron microscopy (SEM), contact angle measurement and nanoindentation tests were used to study surface properties. Coating degradation rate and drug release profile were studied for 40 days. Cell viability experiments were also performed on human endothelial (HUVEC) and smooth muscle cells (HUASMC) using MTT assay and SEM. XRD and DSC analysis showed electro-spraying significantly reduce PLA and DEX crystallinity. Surface studies showed ES coatings has significantly higher hydrophobicity and roughness with microbead-nanofiber morphology vs. micro-nanoporous structure of spin-coated samples. Initial burst release of DEX was 22% and 10% after 6 h and total release was 71% and 46% after 40 days for ES and spin-coated samples, respectively. HUVEC viability of ES samples was higher than spin-coated ones after 1 and 4 days. However, dexamethasone release profile reduced HUASMC proliferation in ES PLA-DEX samples in comparison to spin-coated after 1 and 3 days. In conclusion, in vitro results showed potential of ES PLA-DEX as a biocompatible and efficient anti-inflammatory coating with suitable drug release profile for future applications such as coronary drug eluting stents.
Collapse
Affiliation(s)
- Mostafa Rahvar
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATM), Tehran University of Medical Sciences (TUMS), Italia Street, Tehran, Iran
| | - Gholamreza Ahmadi Lakalayeh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATM), Tehran University of Medical Sciences (TUMS), Italia Street, Tehran, Iran
| | - Niloofar Nazeri
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATM), Tehran University of Medical Sciences (TUMS), Italia Street, Tehran, Iran.,Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Bahereh T Marouf
- Department of Materials Science and Engineering, Faculty of Engineering, Urmia University, Urmia, Iran
| | - Mahdieh Shirzad
- Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Azar Najafi T Shabankareh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATM), Tehran University of Medical Sciences (TUMS), Italia Street, Tehran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine (SATM), Tehran University of Medical Sciences (TUMS), Italia Street, Tehran, Iran.,Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
204
|
Nguyen CT, Ebrahimi M, Gilbert PM, Stewart BA. Electrophysiological analysis of healthy and dystrophic 3-D bioengineered skeletal muscle tissues. Am J Physiol Cell Physiol 2021; 321:C749-C759. [PMID: 34406904 DOI: 10.1152/ajpcell.00049.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recently, methods for creating three-dimensional (3-D) human skeletal muscle tissues from myogenic cell lines have been reported. Bioengineered muscle tissues are contractile and respond to electrical and chemical stimulation. In this study, we provide an electrophysiological analysis of healthy and dystrophic 3-D bioengineered skeletal muscle tissues, focusing on Duchenne muscular dystrophy (DMD). We enlist the 3-D in vitro model of DMD muscle tissue to evaluate muscle cell electrical properties uncoupled from presynaptic neural inputs, an understudied aspect of DMD. Our data show that previously reported electrophysiological aspects of DMD, including effects on membrane potential and membrane resistance, are replicated in the 3-D muscle tissue model. Furthermore, we test a potential therapeutic compound, poloxamer 188, and demonstrate capacity for improving the membrane potential in DMD muscle. Therefore, this study serves as a baseline for a new in vitro method to examine potential therapies for muscular disorders.
Collapse
Affiliation(s)
- Christine T Nguyen
- Department of Biology, University of Toronto Mississauga, Toronto, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Majid Ebrahimi
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Penney M Gilbert
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Bryan A Stewart
- Department of Biology, University of Toronto Mississauga, Toronto, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
205
|
Niu Y, Stadler FJ, Fang J, Galluzzi M. Hyaluronic acid-functionalized poly-lactic acid (PLA) microfibers regulate vascular endothelial cell proliferation and phenotypic shape expression. Colloids Surf B Biointerfaces 2021; 206:111970. [PMID: 34280683 DOI: 10.1016/j.colsurfb.2021.111970] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 01/06/2023]
Abstract
This work was designed to evaluate the efficacy of hyaluronic acid (HA) functionalized tubular poly-lactic acid (PLA) microfibers in directing the luminal pre-endothelialization of vascular endothelial cells (ECs). Tubular HA/PLA microfibers with hierarchical architecture were prepared by electrospinning and chemical cross-linking process. A layer of HA microfibrous film coating was fixed on the inner wall surface of the tubular HA/PLA microfibers, resulting in higher anisotropy wettability and relatively lower surface energy and roughness. We confirmed that HA coating on PLA microfibers surface have reduced hemolytic activity and coagulation degree. Mouse vascular ECs exhibited surface-dependent differences in cell elongation and proliferation (HA/PLA > PLA). Compared with PLA microfibers, the gene expression levels of platelet EC adhesion molecule-1 (PECAM-1/CD31) and vascular endothelial growth factor (VEGF) in ECs of HA/PLA microfibers surface were up-regulated. Immunostaining analysis revealed that the surface of HA/PLA nanofibers supported the expression of mature vascular EC phenotype CD31 protein. In vitro co-culture analysis showed that the luminal pre-endothelialization induced vascular smooth muscle cells (SMCs) to maintain their phenotypic shape and establish natural behavior patterns in the hierarchical tubular scaffold. These studies indicate that the biophysical cues of scaffolds are potent regulators of vascular EC endothelialization.
Collapse
Affiliation(s)
- Yuqing Niu
- Department of Pediatric Surgery, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, PR China.
| | - Florian J Stadler
- Nanshan District Key Lab for Biopolymers and Safety Evaluation, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518055, PR China
| | - Jiahui Fang
- Materials Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Massimiliano Galluzzi
- Materials Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| |
Collapse
|
206
|
Lagerwaard B, Nieuwenhuizen AG, Bunschoten A, de Boer VC, Keijer J. Matrisome, innervation and oxidative metabolism affected in older compared with younger males with similar physical activity. J Cachexia Sarcopenia Muscle 2021; 12:1214-1231. [PMID: 34219410 PMCID: PMC8517362 DOI: 10.1002/jcsm.12753] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Due to the interaction between skeletal muscle ageing and lifestyle factors, it is often challenging to attribute the decline in muscle mass and quality to either changes in lifestyle or to advancing age itself. Because many of the physiological factors affecting muscle mass and quality are modulated by physical activity and physical activity declines with age, the aim of this study is to better understand the effects of early ageing on muscle function by comparing a population of healthy older and young males with similar physical activity patterns. METHODS Eighteen older (69 ± 2.0 years) and 20 young (22 ± 2.0 years) males were recruited based on similar self-reported physical activity, which was verified using accelerometry measurements. Gene expression profiles of vastus lateralis biopsies obtained by RNA sequencing were compared, and key results were validated using quantitative polymerase chain reaction and western blot. RESULTS Total physical activity energy expenditure was similar between the young and old group (404 ± 215 vs. 411 ± 189 kcal/day, P = 0.11). Three thousand seven hundred ninety-seven differentially expressed coding genes (DEGs) were identified (adjusted P-value cut-off of <0.05), of which 1891 were higher and 1906 were lower expressed in the older muscle. The matrisome, innervation and inflammation were the main upregulated processes, and oxidative metabolism was the main downregulated process in old compared with young muscle. Lower protein levels of mitochondrial transcription factor A (TFAM, P = 0.030) and mitochondrial respiratory Complexes IV and II (P = 0.011 and P = 0.0009, respectively) were observed, whereas a trend was observed for Complex I (P = 0.062), in older compared with young muscle. Protein expression of Complexes I and IV was significantly correlated to mitochondrial capacity in the vastus lateralis as measured in vivo (P = 0.017, R2 = 0.42 and P = 0.030, R2 = 0.36). A trend for higher muscle-specific receptor kinase (MUSK) protein levels in the older group was observed (P = 0.08). CONCLUSIONS There are clear differences in the transcriptome signatures of the vastus lateralis muscle of healthy older and young males with similar physical activity levels, including significant differences at the protein level. By disentangling physical activity and ageing, we appoint early skeletal muscle ageing processes that occur despite similar physical activity. Improved understanding of these processes will be key to design targeted anti-ageing therapies.
Collapse
Affiliation(s)
- Bart Lagerwaard
- Human and Animal PhysiologyWageningen University and ResearchWageningenThe Netherlands
- TI Food and NutritionWageningenThe Netherlands
| | - Arie G. Nieuwenhuizen
- Human and Animal PhysiologyWageningen University and ResearchWageningenThe Netherlands
| | - Annelies Bunschoten
- Human and Animal PhysiologyWageningen University and ResearchWageningenThe Netherlands
| | - Vincent C.J. de Boer
- Human and Animal PhysiologyWageningen University and ResearchWageningenThe Netherlands
| | - Jaap Keijer
- Human and Animal PhysiologyWageningen University and ResearchWageningenThe Netherlands
| |
Collapse
|
207
|
Ting MS, Travas-Sejdic J, Malmström J. Modulation of hydrogel stiffness by external stimuli: soft materials for mechanotransduction studies. J Mater Chem B 2021; 9:7578-7596. [PMID: 34596202 DOI: 10.1039/d1tb01415c] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Mechanotransduction is an important process in determining cell survival, proliferation, migration and differentiation. The extracellular matrix (ECM) is the component of natural tissue that provides structural support and biochemical signals to adhering cells. The ECM is dynamic and undergoes physical and biochemical changes in response to various stimuli and there is an interest in understanding the effect of dynamic changes in stiffness on cell behaviour and fate. Therefore, stimuli-responsive hydrogels have been developed to mimic the cells' microenvironment in a controlled fashion. Herein, we review strategies for dynamic modulation of stiffness using various stimuli, such as light, temperature and pH. Special emphasis is placed on conducting polymer (CP) hydrogels and their fabrication procedures. We believe that the redox properties of CPs and hydrogels' biological properties make CPs hydrogels a promising substrate to investigate the effect of dynamic stiffness changes and mechanical actuation on cell fate in future studies.
Collapse
Affiliation(s)
- Matthew S Ting
- Department of Chemical and Materials Engineering, The University of Auckland, Auckland, New Zealand. .,MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand.,Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Jadranka Travas-Sejdic
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand.,Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Jenny Malmström
- Department of Chemical and Materials Engineering, The University of Auckland, Auckland, New Zealand. .,MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand.,Polymer Biointerface Centre, School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
208
|
Sung TC, Lu MW, Tian Z, Lee HHC, Pan J, Ling QD, Higuchi A. Poly(vinyl alcohol- co-itaconic acid) hydrogels grafted with several designed peptides for human pluripotent stem cell culture and differentiation into cardiomyocytes. J Mater Chem B 2021; 9:7662-7673. [PMID: 34586153 DOI: 10.1039/d1tb01555a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We developed poly(vinyl alcohol-co-itaconic acid) (PV) hydrogels grafted with laminin-derived peptides that had different joint segments and several specific designs, including dual chain motifs. PV hydrogels grafted with a peptide derived from laminin-β4 (PMQKMRGDVFSP) containing a joint segment, dual chain motif and cationic amino acid insertion could attach human pluripotent stem (hPS) cells and promoted high expansion folds in long-term culture (over 10 passages) with low differentiation rates, whereas hPS cells attached poorly on PV hydrogels grafted with laminin-α5 peptides that had joint segments with and without a cationic amino acid or on PV hydrogels grafted with laminin-β4 peptides containing the joint segment only. The inclusion of a cationic amino acid in the laminin-β4 peptide was critical for hPS cell attachment on PV hydrogels, which contributed to the zeta potential shifting to higher values (3-4 mV enhancement). The novel peptide segment-grafted PV hydrogels developed in this study supported hPS cell proliferation, which induced better hPS cell expansion than recombinant vitronectin-coated dishes (gold standard of hPS cell culture dishes) in xeno-free culture conditions. After long-term culture on peptide-grafted hydrogels, hPS cells could be induced to differentiate into specific lineages of cells, such as cardiomyocytes, with high efficiency.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Ming-Wei Lu
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan.
| | - Zeyu Tian
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Henry Hsin-Chung Lee
- Department of Surgery, Hsinchu Cathay General Hospital, No. 678, Sec 2, Zhonghua Rd., Hsinchu, 30060, Taiwan.,Graduate Institute of Translational and Interdisciplinary Medicine, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan
| | - Jiandong Pan
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China.
| | - Qing-Dong Ling
- Cathay Medical Research Institute, Cathay General Hospital, No. 32, Ln 160, Jian-Cheng Road, Hsi-Chi City, Taipei 221, Taiwan
| | - Akon Higuchi
- School of Ophthalmology and Optometry, The Eye Hospital of Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang, 325027, China. .,Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan, 32001, Taiwan. .,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan.,Nano Medical Engineering Laboratory, Riken Cluster for Pioneering Research, Riken, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.,Department of Reproduction, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| |
Collapse
|
209
|
Tonti OR, Larson H, Lipp SN, Luetkemeyer CM, Makam M, Vargas D, Wilcox SM, Calve S. Tissue-specific parameters for the design of ECM-mimetic biomaterials. Acta Biomater 2021; 132:83-102. [PMID: 33878474 PMCID: PMC8434955 DOI: 10.1016/j.actbio.2021.04.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules that mechanically and biochemically directs cell behavior and is crucial for maintaining tissue function and health. The heterogeneous organization and composition of the ECM varies within and between tissue types, directing mechanics, aiding in cell-cell communication, and facilitating tissue assembly and reassembly during development, injury and disease. As technologies like 3D printing rapidly advance, researchers are better able to recapitulate in vivo tissue properties in vitro; however, tissue-specific variations in ECM composition and organization are not given enough consideration. This is in part due to a lack of information regarding how the ECM of many tissues varies in both homeostatic and diseased states. To address this gap, we describe the components and organization of the ECM, and provide examples for different tissues at various states of disease. While many aspects of ECM biology remain unknown, our goal is to highlight the complexity of various tissues and inspire engineers to incorporate unique components of the native ECM into in vitro platform design and fabrication. Ultimately, we anticipate that the use of biomaterials that incorporate key tissue-specific ECM will lead to in vitro models that better emulate human pathologies. STATEMENT OF SIGNIFICANCE: Biomaterial development primarily emphasizes the engineering of new materials and therapies at the expense of identifying key parameters of the tissue that is being emulated. This can be partially attributed to the difficulty in defining the 3D composition, organization, and mechanics of the ECM within different tissues and how these material properties vary as a function of homeostasis and disease. In this review, we highlight a range of tissues throughout the body and describe how ECM content, cell diversity, and mechanical properties change in diseased tissues and influence cellular behavior. Accurately mimicking the tissue of interest in vitro by using ECM specific to the appropriate state of homeostasis or pathology in vivo will yield results more translatable to humans.
Collapse
Affiliation(s)
- Olivia R Tonti
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Hannah Larson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah N Lipp
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Callan M Luetkemeyer
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Megan Makam
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Diego Vargas
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sean M Wilcox
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States.
| |
Collapse
|
210
|
Engineering hydrogels with homogeneous mechanical properties for controlling stem cell lineage specification. Proc Natl Acad Sci U S A 2021; 118:2110961118. [PMID: 34504006 PMCID: PMC8449376 DOI: 10.1073/pnas.2110961118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2021] [Indexed: 11/19/2022] Open
Abstract
Hydrogels are extensively used for cell culture, tissue engineering, and flexible electronics. In all of these applications, mechanical properties of hydrogels play an important role. Although tremendous studies have been devoted to optimizing the stiffness, strain, toughness, and dynamic mechanical response, the mechanical homogeneity of hydrogels has rarely been considered. By developing a general strategy to control the mechanical homogeneity of hydrogels, here we show that nanoscale variation in matrix stiffness can considerably affect the lineage specification of human embryonic stem cells. Inhomogeneous hydrogels suppress mechanotransduction and facilitate stemness maintenance, while homogenous hydrogels promote mechanotransduction and osteogenic differentiation. Therefore, engineering hydrogels with controllable and well-defined nanoscale homogeneity may have considerable implications in stem cell culture and regenerative medicine. The extracellular matrix (ECM) is mechanically inhomogeneous due to the presence of a wide spectrum of biomacromolecules and hierarchically assembled structures at the nanoscale. Mechanical inhomogeneity can be even more pronounced under pathological conditions due to injury, fibrogenesis, or tumorigenesis. Although considerable progress has been devoted to engineering synthetic hydrogels to mimic the ECM, the effect of the mechanical inhomogeneity of hydrogels has been widely overlooked. Here, we develop a method based on host–guest chemistry to control the homogeneity of maleimide–thiol cross-linked poly(ethylene glycol) hydrogels. We show that mechanical homogeneity plays an important role in controlling the differentiation or stemness maintenance of human embryonic stem cells. Inhomogeneous hydrogels disrupt actin assembly and lead to reduced YAP activation levels, while homogeneous hydrogels promote mechanotransduction. Thus, the method we developed to minimize the mechanical inhomogeneity of hydrogels may have broad applications in cell culture and tissue engineering.
Collapse
|
211
|
Stottlemire BJ, Chakravarti AR, Whitlow JW, Berkland CJ, He M. Remote-Controlled 3D Porous Magnetic Interface toward High-Throughput Dynamic 3D Cell Culture. ACS Biomater Sci Eng 2021; 7:4535-4544. [PMID: 34468120 DOI: 10.1021/acsbiomaterials.1c00459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mechanical stimuli have been shown to play a large role in cellular behavior, including cellular growth, differentiation, morphology, homeostasis, and disease. Therefore, developing bioreactor systems that can create complex mechanical environments for both tissue engineering and disease modeling drug screening is appealing. However, many of existing systems are restricted because of their bulky size with external force generators, destructive microenvironment control, and low throughput. These shortcomings have preceded to the utilization of magnetic stimuli responsive materials, given their untethered, fast, and tunable actuation potential at both the microscale and macroscale level, for seamless integration into cell culture wells and microfluidic systems. Nevertheless, magnetic soft materials for cell culture have been limited due to the inability to develop well-defined 3D structures for more complex and physiological relevant mechanical actuation. Herein, we introduce a facile fabrication process to develop magnetic-PDMS (polydimethylsiloxane) porous composite designs with both well-defined and controllable microlevel and macrolevel features to dynamically manipulate 3D cell-laden gel at the scale. The intrinsic stiffness of the magnetic-PDMS porous composites is also modulated to control the deformation potential to mimic physiological relevant strain levels, with 2.89-11% observed in magnetic actuation studies. High cell viability was achieved with the culturing of both human adipose stem cells (hADMSCs) and human umbilical cord mesenchymal stem cells (hUCMSCs) in 3D cell-laden gel interfaced with the magnetic-PDMS porous composite. Also, the highly interconnected porous network of the magnetic-PDMS composites facilitated free diffusion throughout the porous structure showcasing the potential of a multisurface contact 3D porous magnetic structure in both reservoir and 96-well plate insert designs for more complex dynamic mechanical actuation. In conclusion, these studies provide a means for establishing a biocompatible, tunable magnetic-PDMS porous composite with fast and programmable dynamic strain potential making it a suitable platform for high-throughput, dynamic 3D cell culture.
Collapse
Affiliation(s)
- Bryce J Stottlemire
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States
| | - Aparna R Chakravarti
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States
| | - Jonathan W Whitlow
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States
| | - Cory J Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States.,Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Mei He
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, Kansas 66045, United States.,Department of Pharmaceutics, University of Florida, Gainesville, Florida 32608, United States
| |
Collapse
|
212
|
Syndecan-2 expression enriches for hematopoietic stem cells and regulates stem cell repopulating capacity. Blood 2021; 139:188-204. [PMID: 34767029 DOI: 10.1182/blood.2020010447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 08/27/2021] [Indexed: 11/20/2022] Open
Abstract
The discovery of novel hematopoietic stem cell (HSC) surface markers can enhance understanding of HSC identity and function. We have discovered a population of primitive bone marrow (BM) HSCs distinguished by their expression of the heparan sulfate proteoglycan, Syndecan-2, which serves as both a marker and regulator of HSC function. Syndecan-2 expression was increased 10-fold in CD150+CD48-CD34-c-Kit+Sca-1+Lineage- cells (long-term - HSCs, LT-HSCs) compared to differentiated hematopoietic cells. Isolation of BM cells based solely on Syndecan-2 surface expression produced a 24-fold enrichment for LT-HSCs, 6-fold enrichment for alpha-catulin+c-kit+ HSCs, and yielded HSCs with superior in vivo repopulating capacity compared to CD150+ cells. Competitive repopulation assays revealed the HSC frequency to be 17-fold higher in Syndecan-2+CD34-KSL cells compared to Syndecan-2-CD34-KSL cells and indistinguishable from CD150+CD34-KSL cells. Syndecan-2 expression also identified nearly all repopulating HSCs within the CD150+CD34-KSL population. Mechanistically, Syndecan-2 regulates HSC repopulating capacity through control of expression of Cdkn1c (p57) and HSC quiescence. Loss of Syndecan-2 expression caused increased HSC cell cycle entry, downregulation of Cdkn1c and loss of HSC long-term - repopulating capacity. Syndecan-2 is a novel marker of HSCs which regulates HSC repopulating capacity via control of HSC quiescence.
Collapse
|
213
|
Günay KA, Silver JS, Chang TL, Bednarski OJ, Bannister KL, Rogowski CJ, Olwin BB, Anseth KS. Myoblast mechanotransduction and myotube morphology is dependent on BAG3 regulation of YAP and TAZ. Biomaterials 2021; 277:121097. [PMID: 34481290 DOI: 10.1016/j.biomaterials.2021.121097] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022]
Abstract
Skeletal muscle tissue is mechanically dynamic with changes in stiffness influencing function, maintenance, and regeneration. We modeled skeletal muscle mechanical changes in culture with dynamically stiffening hydrogels demonstrating that the chaperone protein BAG3 transduces matrix stiffness by redistributing YAP and TAZ subcellular localization in muscle progenitor cells. BAG3 depletion increases cytoplasmic retention of YAP and TAZ, desensitizing myoblasts to changes in hydrogel elastic moduli. Upon differentiation, muscle progenitors depleted of BAG3 formed enlarged, round myotubes lacking the typical cylindrical morphology. The aberrant morphology is dependent on YAP/TAZ signaling, which was sequestered in the cytoplasm in BAG3-depleted myotubes but predominately nuclear in cylindrical myotubes of control cells. Control progenitor cells induced to differentiate on soft (E' = 4 and 12 kPa) hydrogels formed circular myotubes similar to those observed in BAG3-depleted cells. Inhibition of the Hippo pathway partially restored myotube morphologies, permitting nuclear translocation of YAP and TAZ in BAG3-depleted myogenic progenitors. Thus, BAG3 is a critical mediator of dynamic stiffness changes in muscle tissue, coupling mechanical alterations to intracellular signals and inducing changes in gene expression that influence muscle progenitor cell morphology and differentiation.
Collapse
Affiliation(s)
- K Arda Günay
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Jason S Silver
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA; Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tze-Ling Chang
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Olivia J Bednarski
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Kendra L Bannister
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Cameron J Rogowski
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Bradley B Olwin
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA.
| |
Collapse
|
214
|
Liu C, Li M, Dong ZX, Jiang D, Li X, Lin S, Chen D, Zou X, Zhang XD, Luker GD. Heterogeneous microenvironmental stiffness regulates pro-metastatic functions of breast cancer cells. Acta Biomater 2021; 131:326-340. [PMID: 34246802 PMCID: PMC8784164 DOI: 10.1016/j.actbio.2021.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022]
Abstract
Besides molecular and phenotypic variations observed in cancer cells, intratumoral heterogeneity also occurs in the tumor microenvironment. Correlative stiffness maps of different intratumor locations in breast tumor biopsies show that stiffness increases from core to periphery. However, how different local ECM stiffness regulates key functions of cancer cells in tumor progression remains unclear. Although increased tissue stiffness is an established driver of breast cancer progression, conclusions from 2D cultures do not correspond with newer data from cancer cells in 3D environments. Many past studies of breast cancer in 3D culture fail to recapitulate the stiffness of a real breast tumor or the various local stiffnesses present in a tumor microenvironment. In this study, we developed a series of collagen/alginate hybrid hydrogels with adjustable stiffness to match the core, middle, and peripheral zones of a breast tumor. We used this hydrogel system to investigate effects of different local stiffness on morphology, proliferation, and migration of breast cancer cells. RNA sequencing of cells in hydrogels with different stiffness revealed changes in multiple cellular processes underlying cancer progression, including angiogenesis and metabolism. We discovered that tumor cells in a soft environment enriched YAP1 and AP1 signaling related genes, whereas tumor cells in a stiff environment became more pro-angiogenic by upregulating fibronectin 1 (FN1) and matrix metalloproteinase 9 (MMP9) expression. This systematic study defines how the range of environmental stiffnesses present in a breast tumor regulates cancer cells, providing new insights into tumorigenesis and disease progression at the tumor-stroma interface. STATEMENT OF SIGNIFICANCE: Applied a well-defined hybrid hydrogel system to mimic the tumor microenvironment with heterogeneous local stiffness. Breast cancer cells tended to proliferate in soft core environment while migrate in stiff peripheral environment. Breast cancer cells shift from glycolysis to OXPHOS and fatty acid metabolism responding to stiff matrix microenvironment. The transcriptomic profile of breast cancer cells altered due to microenvironmental stiffness changes.
Collapse
Affiliation(s)
- Chun Liu
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China.
| | - Miao Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhao-Xia Dong
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Dong Jiang
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Xiaojing Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Demeng Chen
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuenong Zou
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Xing-Ding Zhang
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
215
|
Ramani S, Ko D, Kim B, Cho C, Kim W, Jo C, Lee CK, Kang J, Hur S, Park S. Technical requirements for cultured meat production: a review. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:681-692. [PMID: 34447948 PMCID: PMC8367405 DOI: 10.5187/jast.2021.e45] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/28/2022]
Abstract
Environment, food, and disease have a selective force on the present and future
as well as our genome. Adaptation of livestock and the environmental nexus,
including forest encroachment for anthropological needs, has been proven to
cause emerging infectious diseases. Further, these demand changes in meat
production and market systems. Meat is a reliable source of protein, with a
majority of the world population consumes meat. To meet the increasing demands
of meat production as well as address issues, such as current environmental
pollution, animal welfare, and outbreaks, cellular agriculture has emerged as
one of the next industrial revolutions. Lab grown meat or cell cultured meat is
a promising way to pursue this; however, it still needs to resemble traditional
meat and be assured safety for human consumption. Further, to mimic the
palatability of traditional meat, the process of cultured meat production starts
from skeletal muscle progenitor cells isolated from animals that proliferate and
differentiate into skeletal muscle using cell culture techniques. Due to several
lacunae in the current approaches, production of muscle replicas is not possible
yet. Our review shows that constant research in this field will resolve the
existing constraints and enable successful cultured meat production in the near
future. Therefore, production of cultured meat is a better solution that looks
after environmental issues, spread of outbreaks, antibiotic resistance through
the zoonotic spread, food and economic crises.
Collapse
Affiliation(s)
| | - Deunsol Ko
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Bosung Kim
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Changjun Cho
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Woosang Kim
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | | | - Sunjin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Sungkwon Park
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| |
Collapse
|
216
|
Basurto IM, Passipieri JA, Gardner GM, Smith KK, Amacher AR, Hansrisuk AI, Christ GJ, Caliari SR. Photoreactive hydrogel stiffness influences volumetric muscle loss repair. Tissue Eng Part A 2021; 28:312-329. [PMID: 34409861 DOI: 10.1089/ten.tea.2021.0137] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Volumetric muscle loss (VML) injuries are characterized by permanent loss of muscle mass, structure, and function. Hydrogel biomaterials provide an attractive platform for skeletal muscle tissue engineering due to the ability to easily modulate their biophysical and biochemical properties to match a range of tissue characteristics. In this work we successfully developed a mechanically tunable hyaluronic acid (HA) hydrogel system to investigate the influence of hydrogel stiffness on VML repair. HA was functionalized with photoreactive norbornene groups to create hydrogel networks that rapidly crosslink via thiol-ene click chemistry with tailored mechanics. Mechanical properties were controlled by modulating the amount of matrix metalloproteinase (MMP)-degradable peptide crosslinker to produce hydrogels with increasing elastic moduli of 1.1 ± 0.002, 3.0 ± 0.002, and 10.6 ± 0.006 kPa mimicking a relevant range of developing and mature muscle stiffnesses. Functional muscle recovery was assessed following implantation of the HA hydrogels by in situ photopolymerization into rat latissimus dorsi (LD) VML defects at 12 and 24 weeks post-injury. After 12 weeks, muscles treated with medium stiffness (3.0 kPa) hydrogels produced maximum isometric forces most similar to contralateral healthy LD muscles. This trend persisted at 24 weeks post-injury, suggestive of sustained functional recovery. Histological analysis revealed a significantly larger zone of regeneration with more de novo muscle fibers following implantation of medium stiffness hydrogels in VML-injured muscles compared to other experimental groups. Lower (low and medium) stiffness hydrogels also appeared to attenuate the chronic inflammatory response characteristic of VML injuries, displaying similar levels of macrophage infiltration and polarization to healthy muscle. Together these findings illustrate the importance of hydrogel mechanical properties in supporting functional repair of VML injuries.
Collapse
Affiliation(s)
- Ivan M Basurto
- University of Virginia, 2358, Biomedical Engineering, Charlottesville, Virginia, United States;
| | - Juliana A Passipieri
- University of Virginia, 2358, Biomedical Engineering, Orthopaedic Surgery, Charlottesville, Virginia, United States;
| | - Gregg M Gardner
- University of Virginia, 2358, Chemical Engineering, Charlottesville, Virginia, United States;
| | - Kathryn K Smith
- University of Virginia, 2358, Chemical Engineering, Charlottesville, Virginia, United States;
| | - Austin R Amacher
- University of Virginia, 2358, Biomedical Engineering, Charlottesville, Virginia, United States;
| | - Audrey I Hansrisuk
- University of Virginia, 2358, Chemistry, Charlottesville, Virginia, United States;
| | - George J Christ
- University of Virginia, 2358, Biomedical Engineering, Orthopaedic Surgery, Charlottesville, Virginia, United States;
| | - Steven R Caliari
- University of Virginia, 2358, Chemical Engineering, Biomedical Engineering, Charlottesville, Virginia, United States;
| |
Collapse
|
217
|
He C, Wang T, Wang Y, Xu T, Zhao S, Shi H, Zou R. ILK regulates osteogenic differentiation of Human Periodontal Ligament Stem Cells through YAP-mediated Mechanical Memory. Oral Dis 2021; 29:274-284. [PMID: 34370371 DOI: 10.1111/odi.13997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022]
Abstract
Mechanical memory meant the mechanical properties of the matrix could influence the cell fate even after the matrix was changed and has been justified in many kinds of cells. To utilize the phenomenon to improve periodontal tissue engineering, we studied whether mechanical memory existed in human periodontal ligament stem cells and testified if ILK plays a role in this process. The substrate of different stiffness was fabricated by gelatin methacrylate hydrogel. Two groups of hPDLSCs with stiff (St) and soft (So) matrix respectively were cultivated. Then half of the cells exchanged their matrix stiffness in the fourth passage and therefore So, St, So-St and St-So were formed. Morphology of hPDLSCs and intracellular location of YAP was observed via fluorescence staining, osteogenic differentiation of hPDLSCs was assessed by Real-Time PCR, ALP staining and western blot. Then all these were reassessed after the ILK gene had been knocked down. The results showed that morphology and YAP location of hPDLSCs were different between matrix changed and unchanged groups; osteogenic genes expression, ALP staining and western blot also varied. After the ILK gene had been knocked down, the YAP location and osteogenic activity of hPDLSCs were significantly influenced. Thus, it could be concluded that mechanical memory exists in hPDLSCs; ILK is involved in this process.
Collapse
Affiliation(s)
- Chuan He
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Tairan Wang
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Yijie Wang
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Tongtong Xu
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Shuyang Zhao
- School of medicine, Xi'an Jiaotong University, Xi'an, China
| | - Haoyu Shi
- School of medicine, Xi'an Jiaotong University, Xi'an, China
| | - Rui Zou
- Dentofacial Development Management Center, Hospital of Stomatology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
218
|
Höhfeld J, Benzing T, Bloch W, Fürst DO, Gehlert S, Hesse M, Hoffmann B, Hoppe T, Huesgen PF, Köhn M, Kolanus W, Merkel R, Niessen CM, Pokrzywa W, Rinschen MM, Wachten D, Warscheid B. Maintaining proteostasis under mechanical stress. EMBO Rep 2021; 22:e52507. [PMID: 34309183 PMCID: PMC8339670 DOI: 10.15252/embr.202152507] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Cell survival, tissue integrity and organismal health depend on the ability to maintain functional protein networks even under conditions that threaten protein integrity. Protection against such stress conditions involves the adaptation of folding and degradation machineries, which help to preserve the protein network by facilitating the refolding or disposal of damaged proteins. In multicellular organisms, cells are permanently exposed to stress resulting from mechanical forces. Yet, for long time mechanical stress was not recognized as a primary stressor that perturbs protein structure and threatens proteome integrity. The identification and characterization of protein folding and degradation systems, which handle force-unfolded proteins, marks a turning point in this regard. It has become apparent that mechanical stress protection operates during cell differentiation, adhesion and migration and is essential for maintaining tissues such as skeletal muscle, heart and kidney as well as the immune system. Here, we provide an overview of recent advances in our understanding of mechanical stress protection.
Collapse
Affiliation(s)
- Jörg Höhfeld
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
| | - Dieter O Fürst
- Institute for Cell BiologyRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Sebastian Gehlert
- Institute of Cardiovascular Research and Sports MedicineGerman Sport UniversityCologneGermany
- Department for the Biosciences of SportsInstitute of Sports ScienceUniversity of HildesheimHildesheimGermany
| | - Michael Hesse
- Institute of Physiology I, Life & Brain CenterMedical FacultyRheinische Friedrich‐Wilhelms UniversityBonnGermany
| | - Bernd Hoffmann
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Thorsten Hoppe
- Institute for GeneticsCologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD) and CMMCUniversity of CologneCologneGermany
| | - Pitter F Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA3Forschungszentrum JülichJülichGermany
- CECADUniversity of CologneCologneGermany
| | - Maja Köhn
- Institute of Biology IIIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| | - Waldemar Kolanus
- LIMES‐InstituteRheinische Friedrich‐Wilhelms University BonnBonnGermany
| | - Rudolf Merkel
- Institute of Biological Information Processing, IBI‐2: MechanobiologyForschungszentrum JülichJülichGermany
| | - Carien M Niessen
- Department of Dermatology and CECADUniversity of CologneCologneGermany
| | | | - Markus M Rinschen
- Department of Biomedicine and Aarhus Institute of Advanced StudiesAarhus UniversityAarhusDenmark
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Dagmar Wachten
- Institute of Innate ImmunityUniversity Hospital BonnBonnGermany
| | - Bettina Warscheid
- Institute of Biology IIFaculty of Biology, and Signalling Research Centres BIOSS and CIBSSAlbert‐Ludwigs‐University FreiburgFreiburgGermany
| |
Collapse
|
219
|
Boyer O, Butler-Browne G, Chinoy H, Cossu G, Galli F, Lilleker JB, Magli A, Mouly V, Perlingeiro RCR, Previtali SC, Sampaolesi M, Smeets H, Schoewel-Wolf V, Spuler S, Torrente Y, Van Tienen F. Myogenic Cell Transplantation in Genetic and Acquired Diseases of Skeletal Muscle. Front Genet 2021; 12:702547. [PMID: 34408774 PMCID: PMC8365145 DOI: 10.3389/fgene.2021.702547] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/16/2021] [Indexed: 01/04/2023] Open
Abstract
This article will review myogenic cell transplantation for congenital and acquired diseases of skeletal muscle. There are already a number of excellent reviews on this topic, but they are mostly focused on a specific disease, muscular dystrophies and in particular Duchenne Muscular Dystrophy. There are also recent reviews on cell transplantation for inflammatory myopathies, volumetric muscle loss (VML) (this usually with biomaterials), sarcopenia and sphincter incontinence, mainly urinary but also fecal. We believe it would be useful at this stage, to compare the same strategy as adopted in all these different diseases, in order to outline similarities and differences in cell source, pre-clinical models, administration route, and outcome measures. This in turn may help to understand which common or disease-specific problems have so far limited clinical success of cell transplantation in this area, especially when compared to other fields, such as epithelial cell transplantation. We also hope that this may be useful to people outside the field to get a comprehensive view in a single review. As for any cell transplantation procedure, the choice between autologous and heterologous cells is dictated by a number of criteria, such as cell availability, possibility of in vitro expansion to reach the number required, need for genetic correction for many but not necessarily all muscular dystrophies, and immune reaction, mainly to a heterologous, even if HLA-matched cells and, to a minor extent, to the therapeutic gene product, a possible antigen for the patient. Finally, induced pluripotent stem cell derivatives, that have entered clinical experimentation for other diseases, may in the future offer a bank of immune-privileged cells, available for all patients and after a genetic correction for muscular dystrophies and other myopathies.
Collapse
Affiliation(s)
- Olivier Boyer
- Department of Immunology & Biotherapy, Rouen University Hospital, Normandy University, Inserm U1234, Rouen, France
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Hector Chinoy
- Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, The University of Manchester, Manchester, United Kingdom
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Berlin, Germany
- InSpe and Division of Neuroscience, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Francesco Galli
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - James B. Lilleker
- Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Alessandro Magli
- Department of Medicine, Lillehei Heart Institute, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Rita C. R. Perlingeiro
- Department of Medicine, Lillehei Heart Institute, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Stefano C. Previtali
- InSpe and Division of Neuroscience, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Hubert Smeets
- Department of Toxicogenomics, Maastricht University Medical Centre, Maastricht, Netherlands
- School for Mental Health and Neurosciences (MHeNS), Maastricht University, Maastricht, Netherlands
- School for Developmental Biology and Oncology (GROW), Maastricht University, Maastricht, Netherlands
| | - Verena Schoewel-Wolf
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Yvan Torrente
- Unit of Neurology, Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Florence Van Tienen
- Department of Toxicogenomics, Maastricht University Medical Centre, Maastricht, Netherlands
- School for Mental Health and Neurosciences (MHeNS), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
220
|
Abdul-Al M, Kyeremeh GK, Saeinasab M, Heidari Keshel S, Sefat F. Stem Cell Niche Microenvironment: Review. Bioengineering (Basel) 2021; 8:bioengineering8080108. [PMID: 34436111 PMCID: PMC8389324 DOI: 10.3390/bioengineering8080108] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
The cornea comprises a pool of self-regenerating epithelial cells that are crucial to preserving clarity and visibility. Limbal epithelial stem cells (LESCs), which live in a specialized stem cell niche (SCN), are crucial for the survival of the human corneal epithelium. They live at the bottom of the limbal crypts, in a physically enclosed microenvironment with a number of neighboring niche cells. Scientists also simplified features of these diverse microenvironments for more analysis in situ by designing and recreating features of different SCNs. Recent methods for regenerating the corneal epithelium after serious trauma, including burns and allergic assaults, focus mainly on regenerating the LESCs. Mesenchymal stem cells, which can transform into self-renewing and skeletal tissues, hold immense interest for tissue engineering and innovative medicinal exploration. This review summarizes all types of LESCs, identity and location of the human epithelial stem cells (HESCs), reconstruction of LSCN and artificial stem cells for self-renewal.
Collapse
Affiliation(s)
- Mohamed Abdul-Al
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD71DP, UK; (M.A.-A.); (G.K.K.)
| | - George Kumi Kyeremeh
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD71DP, UK; (M.A.-A.); (G.K.K.)
| | - Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 91779 48974, Iran;
| | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839 69411, Iran;
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD71DP, UK; (M.A.-A.); (G.K.K.)
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford BD71DP, UK
- Correspondence:
| |
Collapse
|
221
|
Dykstra PB, Dayanidhi S, Chambers HG, Lieber RL. Stretch-induced satellite cell deformation incontracturedmuscles in children with cerebral palsy. J Biomech 2021; 126:110635. [PMID: 34303895 DOI: 10.1016/j.jbiomech.2021.110635] [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: 04/28/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 10/20/2022]
Abstract
Satellite cells (SCs) are quiescent, adult skeletal muscle stem cells responsible for postnatal muscle growth and repair. Children with cerebral palsy (CP) have muscle contractures with reduced SC abundance, extracellular matrix abnormalities and reduced serial sarcomere number resulting in greatly increased in vivo sarcomere length, perhaps due to impaired sarcomere addition, compared to children with typical development (TD). Stretch is a strong activator of SCs that leads to addition of sarcomeres during bone-muscle growth. Mechanical loading and subsequent deformation of intracellular structures can lead to activation and proliferation, perhaps by cytoskeletal transmissions of extracellular mechanical signals to the nuclei. The primary aim of this study was to determine the effect of ex vivo stretch-induced sarcomere length change on SC deformation in children with CP and TD. Muscle biopsies were obtained from twelve children (7 CP, 5 TD) during surgery. Fiber bundles were labeled with fluorescent antibodies for Pax7 (SC), DRAQ5 (nuclei), and alpha-actinin (sarcomere protein). Fibers were stretched using a custom jig and imaged using confocal microscopy. SC nuclear length, height and aspect ratio underwent increased deformation with increasing sarcomere length (p < 0.05) in both groups. Slopes of association for SC nuclear length, aspect ratio and sarcomere lengths were similar between CP and TD. Our results indicate that SC in children with CP undergo similar deformation as TD across sarcomere lengths.
Collapse
Affiliation(s)
- Peter B Dykstra
- Department of Bioengineering, University of California, San Diego, CA, USA; Department of Orthopaedic Surgery, University of California, San Diego, CA, USA
| | - Sudarshan Dayanidhi
- Department of Orthopaedic Surgery, University of California, San Diego, CA, USA; Department of Veterans Affairs Medical Center, San Diego, CA, USA; Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Henry G Chambers
- Department of Orthopaedics, Rady Children's Hospital, San Diego, CA, USA
| | - Richard L Lieber
- Department of Bioengineering, University of California, San Diego, CA, USA; Department of Orthopaedic Surgery, University of California, San Diego, CA, USA; Department of Orthopaedics, Rady Children's Hospital, San Diego, CA, USA; Shirley Ryan AbilityLab, Chicago, IL, USA; Edward G Hines VA Medical Center, Maywood, IL, USA.
| |
Collapse
|
222
|
Shah V, Patel S, Shah J. Emerging Role of Piezo Ion Channels in Cardiovascular Development. Dev Dyn 2021; 251:276-286. [PMID: 34255896 DOI: 10.1002/dvdy.401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/12/2021] [Accepted: 07/09/2021] [Indexed: 12/23/2022] Open
Abstract
Mechanical cues are crucial for vascular development and the proper differentiation of various cell types. Piezo1 and Piezo2 are mechanically activated cationic channels expressed in various cell types, especially in vascular smooth muscle and endothelial cells. It is present as a transmembrane homotrimeric complex, regulating calcium influx. Local blood flow associated shear stress, in addition to blood pressure associated cell membrane stretching are key Piezo channel activators. There is rising proof, showcasing Piezo channels significance in myocytes, cardiac fibroblast, vascular tone maintenance, atherosclerosis, hypertension, NO generation, and baroreceptor reflex. Here, we review the role of Piezo channels in cardiovascular development and its associated clinical disorders. Also, emphasizing on Piezo channel modulators which might lead to novel therapies for cardiovascular diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Sandip Patel
- Department of Pharmacology, L.M. College of Pharmacy, Navrangpura, Ahmedabad, Gujarat, India
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
223
|
Soheilmoghaddam F, Rumble M, Cooper-White J. High-Throughput Routes to Biomaterials Discovery. Chem Rev 2021; 121:10792-10864. [PMID: 34213880 DOI: 10.1021/acs.chemrev.0c01026] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many existing clinical treatments are limited in their ability to completely restore decreased or lost tissue and organ function, an unenviable situation only further exacerbated by a globally aging population. As a result, the demand for new medical interventions has increased substantially over the past 20 years, with the burgeoning fields of gene therapy, tissue engineering, and regenerative medicine showing promise to offer solutions for full repair or replacement of damaged or aging tissues. Success in these fields, however, inherently relies on biomaterials that are engendered with the ability to provide the necessary biological cues mimicking native extracellular matrixes that support cell fate. Accelerating the development of such "directive" biomaterials requires a shift in current design practices toward those that enable rapid synthesis and characterization of polymeric materials and the coupling of these processes with techniques that enable similarly rapid quantification and optimization of the interactions between these new material systems and target cells and tissues. This manuscript reviews recent advances in combinatorial and high-throughput (HT) technologies applied to polymeric biomaterial synthesis, fabrication, and chemical, physical, and biological screening with targeted end-point applications in the fields of gene therapy, tissue engineering, and regenerative medicine. Limitations of, and future opportunities for, the further application of these research tools and methodologies are also discussed.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Madeleine Rumble
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| |
Collapse
|
224
|
Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol 2021; 22:465-482. [PMID: 33837369 DOI: 10.1038/s41580-021-00357-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/02/2023]
Abstract
Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.
Collapse
Affiliation(s)
- Alexander Rauch
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
225
|
Hu LY, Mileti CJ, Loomis T, Brashear SE, Ahmad S, Chellakudam RR, Wohlgemuth RP, Gionet-Gonzales MA, Leach JK, Smith LR. Skeletal muscle progenitors are sensitive to collagen architectural features of fibril size and cross linking. Am J Physiol Cell Physiol 2021; 321:C330-C342. [PMID: 34191625 DOI: 10.1152/ajpcell.00065.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Muscle stem cells (MuSCs) are essential for the robust regenerative capacity of skeletal muscle. However, in fibrotic environments marked by abundant collagen and altered collagen organization, the regenerative capability of MuSCs is diminished. MuSCs are sensitive to their extracellular matrix environment but their response to collagen architecture is largely unknown. The present study aimed to systematically test the effect of underlying collagen structures on MuSC functions. Collagen hydrogels were engineered with varied architectures: collagen concentration, cross linking, fibril size, and fibril alignment, and the changes were validated with second harmonic generation imaging and rheology. Proliferation and differentiation responses of primary mouse MuSCs and immortal myoblasts (C2C12s) were assessed using EdU assays and immunolabeling skeletal muscle myosin expression, respectively. Changing collagen concentration and the corresponding hydrogel stiffness did not have a significant influence on MuSC proliferation or differentiation. However, MuSC differentiation on atelocollagen gels, which do not form mature pyridinoline cross links, was increased compared with the cross-linked control. In addition, MuSCs and C2C12 myoblasts showed greater differentiation on gels with smaller collagen fibrils. Proliferation rates of C2C12 myoblasts were also higher on gels with smaller collagen fibrils, whereas MuSCs did not show a significant difference. Surprisingly, collagen alignment did not have significant effects on muscle progenitor function. This study demonstrates that MuSCs are capable of sensing their underlying extracellular matrix (ECM) structures and enhancing differentiation on substrates with less collagen cross linking or smaller collagen fibrils. Thus, in fibrotic muscle, targeting cross linking and fibril size rather than collagen expression may more effectively support MuSC-based regeneration.
Collapse
Affiliation(s)
- Lin-Ya Hu
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California
| | - Cassidy J Mileti
- Biomedical Engineering Graduate Group, University of California, Davis, California
| | - Taryn Loomis
- Biomedical Engineering Graduate Group, University of California, Davis, California
| | - Sarah E Brashear
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California
| | - Sarah Ahmad
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California
| | - Rosemary R Chellakudam
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California
| | - Ross P Wohlgemuth
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California
| | | | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, California.,Department of Orthopaedic Surgery, University of California, Davis, California
| | - Lucas R Smith
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California.,Department of Physical Medicine and Rehabilitation, University of California, Davis, California
| |
Collapse
|
226
|
MacDonald JA, Woods DC, Tilly JL. Biomechanical Strain Promotes the Differentiation of Murine Oogonial Stem Cells. Stem Cells Dev 2021; 30:749-757. [PMID: 34036812 DOI: 10.1089/scd.2021.0086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cells within tissues are routinely subjected to physiological stress and strain, arising from direct interactions with neighboring cells as well as with extracellular matrix components. Accordingly, there is tremendous interest in deciphering how cells sense, and respond to, changes in biomechanical forces. In this study, we explored the effects of mechanostimulation on the differentiation of mouse female germline or oogonial stem cells (OSCs) as a model for adult stem cell function. We report that increasing levels, or repeated application of a subthreshold fixed level, of radial strain to OSCs in culture significantly increased rates of in vitro oocyte formation as a measure of stem cell differentiation. These responses involved changes in F-actin-mediated cytoskeletal tension as well as in activation of intracellular signaling by Rho-associated protein kinase (ROCK) and Yes-associated protein (YAP) phosphorylation. In addition, application of mechanical strain to OSCs enhanced association of YAP with muscle-specific cytidine-adenosine-thymidine (MCAT) response elements in the promoter stimulated by retinoic acid gene 8 (Stra8), the transcriptional activation of which is required for germline meiotic commitment. These data indicate that biomechanical strain directly promotes the differentiation of adult female germline stem cells through a signaling pathway involving F-actin, ROCK, YAP, and Stra8.
Collapse
Affiliation(s)
- Julie A MacDonald
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Dori C Woods
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Jonathan L Tilly
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
227
|
Sağraç D, Şişli HB, Şenkal S, Hayal TB, Şahin F, Doğan A. Organoids in Tissue Transplantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:45-64. [PMID: 34164796 DOI: 10.1007/5584_2021_647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Improvements in stem cell-based research and genetic modification tools enable stem cell-based tissue regeneration applications in clinical therapies. Although inadequate cell numbers in culture, invasive isolation procedures, and poor survival rates after transplantation remain as major challenges, cell-based therapies are useful tools for tissue regeneration.Organoids hold a great promise for tissue regeneration, organ and disease modeling, drug testing, development, and genetic profiling studies. Establishment of 3D cell culture systems eliminates the disadvantages of 2D models in terms of cell adaptation and tissue structure and function. Organoids possess the capacity to mimic the specific features of tissue architecture, cell-type composition, and the functionality of real organs while preserving the advantages of simplified and easily accessible cell culture models. Thus, organoid technology might emerge as an alternative to cell and tissue transplantation. Although transplantation of various organoids in animal models has been demonstrated, liöitations related to vascularized structure formation, cell viability and functionality remain as obstacles in organoid-based transplantation therapies. Clinical applications of organoid-based transplantations might be possible in the near future, when limitations related to cell viability and tissue integration are solved. In this review, the literature was analyzed and discussed to explore the current status of organoid-based transplantation studies.
Collapse
Affiliation(s)
- Derya Sağraç
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Hatice Burcu Şişli
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Selinay Şenkal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Taha Bartu Hayal
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
228
|
Madl CM, Flaig IA, Holbrook CA, Wang YX, Blau HM. Biophysical matrix cues from the regenerating niche direct muscle stem cell fate in engineered microenvironments. Biomaterials 2021; 275:120973. [PMID: 34224984 DOI: 10.1016/j.biomaterials.2021.120973] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/30/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022]
Abstract
Skeletal muscle stem cells (MuSCs) are essential for efficacious muscle repair, making MuSCs promising therapeutic targets for tissue engineering and regenerative medicine. MuSCs are presented with a diverse and temporally defined set of cues from their microenvironment during regeneration that direct stem cell expansion, differentiation, and return to quiescence. Understanding the complex interplay among these biophysical and biochemical cues is necessary to develop therapies targeting or employing MuSCs. To probe the role of mechanical cues presented by the extracellular matrix, we leverage chemically defined hydrogel substrates with controllable stiffness and adhesive ligand composition to characterize the MuSC response to matrix cues presented during early and late phases of regeneration. We demonstrate that relatively soft hydrogels recapitulating healthy muscle stiffness promote MuSC activation and expansion, while relatively stiff hydrogels impair MuSC proliferation and arrest myogenic progression. These effects are seen on soft and stiff hydrogels presenting laminin-111 and exacerbated on hydrogels presenting RGD adhesive peptides. Soluble factors present in the MuSC niche during different phases of regeneration, prostaglandin E2 and oncostatin M, synergize with matrix-presented cues to enhance stem cell expansion on soft substrates and block myogenic progression on stiff substrates. To determine if temporally varied matrix stiffness reminiscent of the regenerating microenvironment alters MuSC fate, we developed a photoresponsive hydrogel system with accelerated reaction kinetics that can be rapidly softened on demand. MuSCs cultured on these materials revealed that the cellular response to a stiff microenvironment is fixed within the first three days of culture, as subsequent softening back to a healthy stiffness did not rescue MuSC proliferation or myogenic progression. These results highlight the importance of temporally controlled biophysical and biochemical cues in regulating MuSC fate that can be harnessed to improve regenerative medicine approaches to restore skeletal muscle tissue.
Collapse
Affiliation(s)
- Christopher M Madl
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Iris A Flaig
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Colin A Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology & Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
229
|
Hayward MK, Muncie JM, Weaver VM. Tissue mechanics in stem cell fate, development, and cancer. Dev Cell 2021; 56:1833-1847. [PMID: 34107299 PMCID: PMC9056158 DOI: 10.1016/j.devcel.2021.05.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022]
Abstract
Cells in tissues experience a plethora of forces that regulate their fate and modulate development and homeostasis. Cells sense mechanical cues through localized mechanoreceptors or by influencing cytoskeletal or plasma membrane organization. Cells translate force and modulate their behavior through a process termed mechanotransduction. Cells tune their tension upon exposure to chronic force by engaging cellular machinery that modulates actin tension, which in turn stimulates matrix remodeling and stiffening and alters cell-cell adhesions until cells achieve a state of tensional homeostasis. Loss of tensional homeostasis can be induced through oncogene activity and/or tissue fibrosis, accompanies tumor progression, and is associated with increased cancer risk. The mechanical stresses that develop in tumors can also foster the mesenchymal-like transdifferentiation of cells to induce a stem-like phenotype that contributes to their aggression, metastatic dissemination, and treatment resistance. Thus, strategies that ameliorate tumor mechanics may comprise an effective strategy to prevent aggressive tumor behavior.
Collapse
Affiliation(s)
- Mary-Kate Hayward
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences and Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; The Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
230
|
Stowers RS. Advances in Extracellular Matrix-Mimetic Hydrogels to Guide Stem Cell Fate. Cells Tissues Organs 2021; 211:703-720. [PMID: 34082418 DOI: 10.1159/000514851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/11/2021] [Indexed: 01/25/2023] Open
Abstract
In the fields of regenerative medicine and tissue engineering, stem cells offer vast potential for treating or replacing diseased and damaged tissue. Much progress has been made in understanding stem cell biology, yielding protocols for directing stem cell differentiation toward the cell type of interest for a specific application. One particularly interesting and powerful signaling cue is the extracellular matrix (ECM) surrounding stem cells, a network of biopolymers that, along with cells, makes up what we define as a tissue. The composition, structure, biochemical features, and mechanical properties of the ECM are varied in different tissues and developmental stages, and serve to instruct stem cells toward a specific lineage. By understanding and recapitulating some of these ECM signaling cues through engineered ECM-mimicking hydrogels, stem cell fate can be directed in vitro. In this review, we will summarize recent advances in material systems to guide stem cell fate, highlighting innovative methods to capture ECM functionalities and how these material systems can be used to provide basic insight into stem cell biology or make progress toward therapeutic objectives.
Collapse
Affiliation(s)
- Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
231
|
Wu YX, Ma H, Wang JL, Qu W. Production of chitosan scaffolds by lyophilization or electrospinning: which is better for peripheral nerve regeneration? Neural Regen Res 2021; 16:1093-1098. [PMID: 33269755 PMCID: PMC8224144 DOI: 10.4103/1673-5374.300463] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/24/2020] [Accepted: 09/20/2020] [Indexed: 11/17/2022] Open
Abstract
Both lyophilization and electrospinning are commonly used to make chitosan scaffolds. However, it remains unknown which method is better for cell growth. In this study, we established the following groups: (1) lyophilization group-chitosan scaffolds were prepared by lyophilization method and seeded with Schwann cells from Sprague-Dawley rats aged 3-5 days; (2) electrospinning group-chitosan scaffolds were prepared by electrospinning method and seeded with Schwann cells; (3) control group-Schwann cells were cultured on culture dishes. The growth of Schwann cells was evaluated by immunofluorescence and scanning electron microscopy. Western blot assay was performed to explore the mechanism of Schwann cell growth. Both materials were non-toxic and suitable for the growth of Schwann cells. The pores produced by electrospinning were much smaller than those produced by lyophilization. The proliferation rate and adhesion rate of Schwann cells in the electrospinning group were higher than those in the lyophilization group. Schwann cells cultured on electrospinning scaffolds formed a Bungner band-like structure, and a much greater amount of brain-derived neurotrophic factor was secreted, which can promote the growth of neurons. Our findings show that the chitosan scaffold prepared by the electrospinning method has a nanofiber structure that provides an extracellular matrix that is more favorable for cell-cell interactions. The electrospinning method is more suitable for nerve regeneration than the lyophilization method. This research was approved by the Medical Ethical Committee of Dalian Medical University (approval No. AEE1-2016-045) on March 3, 2016.
Collapse
Affiliation(s)
- Yu-Xuan Wu
- Department of Hand Microsurgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Hao Ma
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Lan Wang
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wei Qu
- Department of Hand Microsurgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
232
|
Morton AB, Jacobsen NL, Segal SS. Functionalizing biomaterials to promote neurovascular regeneration following skeletal muscle injury. Am J Physiol Cell Physiol 2021; 320:C1099-C1111. [PMID: 33852364 PMCID: PMC8285637 DOI: 10.1152/ajpcell.00501.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022]
Abstract
During embryogenesis, blood vessels and nerves develop with similar branching structure in response to shared signaling pathways guiding network growth. With both systems integral to physiological homeostasis, dual targeting of blood vessels and nerves to promote neurovascular regeneration following injury is an emerging therapeutic approach in biomedical engineering. A limitation to this strategy is that the nature of cross talk between emergent vessels and nerves during regeneration in an adult is poorly understood. Following peripheral nerve transection, intraneural vascular cells infiltrate the site of injury to provide a migratory pathway for mobilized Schwann cells of regenerating axons. As Schwann cells demyelinate, they secrete vascular endothelial growth factor, which promotes angiogenesis. Recent advances point to concomitant restoration of neurovascular architecture and function through simultaneous targeting of growth factors and guidance cues shared by both systems during regeneration. In the context of traumatic injury associated with volumetric muscle loss, we consider the nature of biomaterials used to engineer three-dimensional scaffolds, functionalization of scaffolds with molecular signals that guide and promote neurovascular growth, and seeding scaffolds with progenitor cells. Physiological success is defined by each tissue component of the bioconstruct (nerve, vessel, muscle) becoming integrated with that of the host. Advances in microfabrication, cell culture techniques, and progenitor cell biology hold great promise for engineering bioconstructs able to restore organ function after volumetric muscle loss.
Collapse
Affiliation(s)
- Aaron B Morton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Nicole L Jacobsen
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
233
|
Ausems CRM, van Engelen BGM, van Bokhoven H, Wansink DG. Systemic cell therapy for muscular dystrophies : The ultimate transplantable muscle progenitor cell and current challenges for clinical efficacy. Stem Cell Rev Rep 2021; 17:878-899. [PMID: 33349909 PMCID: PMC8166694 DOI: 10.1007/s12015-020-10100-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 01/07/2023]
Abstract
The intrinsic regenerative capacity of skeletal muscle makes it an excellent target for cell therapy. However, the potential of muscle tissue to renew is typically exhausted and insufficient in muscular dystrophies (MDs), a large group of heterogeneous genetic disorders showing progressive loss of skeletal muscle fibers. Cell therapy for MDs has to rely on suppletion with donor cells with high myogenic regenerative capacity. Here, we provide an overview on stem cell lineages employed for strategies in MDs, with a focus on adult stem cells and progenitor cells resident in skeletal muscle. In the early days, the potential of myoblasts and satellite cells was explored, but after disappointing clinical results the field moved to other muscle progenitor cells, each with its own advantages and disadvantages. Most recently, mesoangioblasts and pericytes have been pursued for muscle cell therapy, leading to a handful of preclinical studies and a clinical trial. The current status of (pre)clinical work for the most common forms of MD illustrates the existing challenges and bottlenecks. Besides the intrinsic properties of transplantable cells, we discuss issues relating to cell expansion and cell viability after transplantation, optimal dosage, and route and timing of administration. Since MDs are genetic conditions, autologous cell therapy and gene therapy will need to go hand-in-hand, bringing in additional complications. Finally, we discuss determinants for optimization of future clinical trials for muscle cell therapy. Joined research efforts bring hope that effective therapies for MDs are on the horizon to fulfil the unmet clinical need in patients.
Collapse
Affiliation(s)
- C Rosanne M Ausems
- Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
- Donders lnstitute for Brain Cognition and Behavior, Department of Neurology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Baziel G M van Engelen
- Donders lnstitute for Brain Cognition and Behavior, Department of Neurology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands.
| | - Derick G Wansink
- Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands.
| |
Collapse
|
234
|
Alarcin E, Bal-Öztürk A, Avci H, Ghorbanpoor H, Dogan Guzel F, Akpek A, Yesiltas G, Canak-Ipek T, Avci-Adali M. Current Strategies for the Regeneration of Skeletal Muscle Tissue. Int J Mol Sci 2021; 22:5929. [PMID: 34072959 PMCID: PMC8198586 DOI: 10.3390/ijms22115929] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic injuries, tumor resections, and degenerative diseases can damage skeletal muscle and lead to functional impairment and severe disability. Skeletal muscle regeneration is a complex process that depends on various cell types, signaling molecules, architectural cues, and physicochemical properties to be successful. To promote muscle repair and regeneration, various strategies for skeletal muscle tissue engineering have been developed in the last decades. However, there is still a high demand for the development of new methods and materials that promote skeletal muscle repair and functional regeneration to bring approaches closer to therapies in the clinic that structurally and functionally repair muscle. The combination of stem cells, biomaterials, and biomolecules is used to induce skeletal muscle regeneration. In this review, we provide an overview of different cell types used to treat skeletal muscle injury, highlight current strategies in biomaterial-based approaches, the importance of topography for the successful creation of functional striated muscle fibers, and discuss novel methods for muscle regeneration and challenges for their future clinical implementation.
Collapse
Affiliation(s)
- Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34854 Istanbul, Turkey;
| | - Ayca Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, 34010 Istanbul, Turkey;
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Hüseyin Avci
- Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Cellular Therapy and Stem Cell Research Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Translational Medicine Research and Clinical Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Hamed Ghorbanpoor
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
- Department of Biomedical Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Fatma Dogan Guzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
| | - Ali Akpek
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Gözde Yesiltas
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Tuba Canak-Ipek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| |
Collapse
|
235
|
Escobar H, Krause A, Keiper S, Kieshauer J, Müthel S, de Paredes MG, Metzler E, Kühn R, Heyd F, Spuler S. Base editing repairs an SGCA mutation in human primary muscle stem cells. JCI Insight 2021; 6:145994. [PMID: 33848270 PMCID: PMC8262330 DOI: 10.1172/jci.insight.145994] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/07/2021] [Indexed: 11/28/2022] Open
Abstract
Skeletal muscle can regenerate from muscle stem cells and their myogenic precursor cell progeny, myoblasts. However, precise gene editing in human muscle stem cells for autologous cell replacement therapies of untreatable genetic muscle diseases has not yet been reported. Loss-of-function mutations in SGCA, encoding α-sarcoglycan, cause limb-girdle muscular dystrophy 2D/R3, an early-onset, severe, and rapidly progressive form of muscular dystrophy affecting both male and female patients. Patients suffer from muscle degeneration and atrophy affecting the limbs, respiratory muscles, and heart. We isolated human muscle stem cells from 2 donors, with the common SGCA c.157G>A mutation affecting the last coding nucleotide of exon 2. We found that c.157G>A is an exonic splicing mutation that induces skipping of 2 coregulated exons. Using adenine base editing, we corrected the mutation in the cells from both donors with > 90% efficiency, thereby rescuing the splicing defect and α-sarcoglycan expression. Base-edited patient cells regenerated muscle and contributed to the Pax7+ satellite cell compartment in vivo in mouse xenografts. Here, we provide the first evidence to our knowledge that autologous gene–repaired human muscle stem cells can be harnessed for cell replacement therapies of muscular dystrophies.
Collapse
Affiliation(s)
- Helena Escobar
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Anne Krause
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sandra Keiper
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Berlin, Germany
| | - Janine Kieshauer
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Stefanie Müthel
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Manuel García de Paredes
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany
| | - Eric Metzler
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralf Kühn
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Florian Heyd
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany
| |
Collapse
|
236
|
Ni Y, Zhou X, Yang J, Shi H, Li H, Zhao X, Ma X. The Role of Tumor-Stroma Interactions in Drug Resistance Within Tumor Microenvironment. Front Cell Dev Biol 2021; 9:637675. [PMID: 34095111 PMCID: PMC8173135 DOI: 10.3389/fcell.2021.637675] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/19/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer cells resistance to various therapies remains to be a key challenge nowadays. For a long time, scientists focused on tumor cells themselves for the mechanisms of acquired drug resistance. However, recent evidence showed that tumor microenvironment (TME) is essential for regulating immune escape, drug resistance, progression and metastasis of malignant cells. Reciprocal interactions between cancer cells and non-malignant cells within this milieu often reshape the TME and promote drug resistance. Therefore, advanced knowledge about these sophisticated interactions is significant for the design of effective therapeutic approaches. In this review, we highlight cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), T-regulatory lymphocytes (Tregs), mesenchymal stem cells (MSCs), cancer-associated adipocytes (CAAs), and tumor endothelial cells (TECs) existing in TME, as well as their multiple cross-talk with tumor cells, which eventually endows tumor cells with therapeutic resistance.
Collapse
Affiliation(s)
- Yanghong Ni
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xiaoting Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jia Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Houhui Shi
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Hongyi Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China.,Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
237
|
Dey K, Roca E, Ramorino G, Sartore L. Progress in the mechanical modulation of cell functions in tissue engineering. Biomater Sci 2021; 8:7033-7081. [PMID: 33150878 DOI: 10.1039/d0bm01255f] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, mechanics at multiple stages-nucleus to cell to ECM-underlie multiple physiological and pathological functions from its development to reproduction to death. Under this inspiration, substantial research has established the role of multiple aspects of mechanics in regulating fundamental cellular processes, including spreading, migration, growth, proliferation, and differentiation. However, our understanding of how these mechanical mechanisms are orchestrated or tuned at different stages to maintain or restore the healthy environment at the tissue or organ level remains largely a mystery. Over the past few decades, research in the mechanical manipulation of the surrounding environment-known as substrate or matrix or scaffold on which, or within which, cells are seeded-has been exceptionally enriched in the field of tissue engineering and regenerative medicine. To do so, traditional tissue engineering aims at recapitulating key mechanical milestones of native ECM into a substrate for guiding the cell fate and functions towards specific tissue regeneration. Despite tremendous progress, a big puzzle that remains is how the cells compute a host of mechanical cues, such as stiffness (elasticity), viscoelasticity, plasticity, non-linear elasticity, anisotropy, mechanical forces, and mechanical memory, into many biological functions in a cooperative, controlled, and safe manner. High throughput understanding of key cellular decisions as well as associated mechanosensitive downstream signaling pathway(s) for executing these decisions in response to mechanical cues, solo or combined, is essential to address this issue. While many reports have been made towards the progress and understanding of mechanical cues-particularly, substrate bulk stiffness and viscoelasticity-in regulating the cellular responses, a complete picture of mechanical cues is lacking. This review highlights a comprehensive view on the mechanical cues that are linked to modulate many cellular functions and consequent tissue functionality. For a very basic understanding, a brief discussion of the key mechanical players of ECM and the principle of mechanotransduction process is outlined. In addition, this review gathers together the most important data on the stiffness of various cells and ECM components as well as various tissues/organs and proposes an associated link from the mechanical perspective that is not yet reported. Finally, beyond addressing the challenges involved in tuning the interplaying mechanical cues in an independent manner, emerging advances in designing biomaterials for tissue engineering are also explored.
Collapse
Affiliation(s)
- Kamol Dey
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Bangladesh
| | | | | | | |
Collapse
|
238
|
Culturing Keratinocytes on Biomimetic Substrates Facilitates Improved Epidermal Assembly In Vitro. Cells 2021; 10:cells10051177. [PMID: 34066027 PMCID: PMC8151809 DOI: 10.3390/cells10051177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/29/2021] [Accepted: 05/09/2021] [Indexed: 12/18/2022] Open
Abstract
Mechanotransduction is defined as the ability of cells to sense mechanical stimuli from their surroundings and translate them into biochemical signals. Epidermal keratinocytes respond to mechanical cues by altering their proliferation, migration, and differentiation. In vitro cell culture, however, utilises tissue culture plastic, which is significantly stiffer than the in vivo environment. Current epidermal models fail to consider the effects of culturing keratinocytes on plastic prior to setting up three-dimensional cultures, so the impact of this non-physiological exposure on epidermal assembly is largely overlooked. In this study, primary keratinocytes cultured on plastic were compared with those grown on 4, 8, and 50 kPa stiff biomimetic hydrogels that have similar mechanical properties to skin. Our data show that keratinocytes cultured on biomimetic hydrogels exhibited major changes in cellular architecture, cell density, nuclear biomechanics, and mechanoprotein expression, such as specific Linker of Nucleoskeleton and Cytoskeleton (LINC) complex constituents. Mechanical conditioning of keratinocytes on 50 kPa biomimetic hydrogels improved the thickness and organisation of 3D epidermal models. In summary, the current study demonstrates that the effects of extracellular mechanics on keratinocyte cell biology are significant and therefore should be harnessed in skin research to ensure the successful production of physiologically relevant skin models.
Collapse
|
239
|
Westman AM, Peirce SM, Christ GJ, Blemker SS. Agent-based model provides insight into the mechanisms behind failed regeneration following volumetric muscle loss injury. PLoS Comput Biol 2021; 17:e1008937. [PMID: 33970905 PMCID: PMC8110270 DOI: 10.1371/journal.pcbi.1008937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle possesses a remarkable capacity for repair and regeneration following a variety of injuries. When successful, this highly orchestrated regenerative process requires the contribution of several muscle resident cell populations including satellite stem cells (SSCs), fibroblasts, macrophages and vascular cells. However, volumetric muscle loss injuries (VML) involve simultaneous destruction of multiple tissue components (e.g., as a result of battlefield injuries or vehicular accidents) and are so extensive that they exceed the intrinsic capability for scarless wound healing and result in permanent cosmetic and functional deficits. In this scenario, the regenerative process fails and is dominated by an unproductive inflammatory response and accompanying fibrosis. The failure of current regenerative therapeutics to completely restore functional muscle tissue is not surprising considering the incomplete understanding of the cellular mechanisms that drive the regeneration response in the setting of VML injury. To begin to address this profound knowledge gap, we developed an agent-based model to predict the tissue remodeling response following surgical creation of a VML injury. Once the model was able to recapitulate key aspects of the tissue remodeling response in the absence of repair, we validated the model by simulating the tissue remodeling response to VML injury following implantation of either a decellularized extracellular matrix scaffold or a minced muscle graft. The model suggested that the SSC microenvironment and absence of pro-differentiation SSC signals were the most important aspects of failed muscle regeneration in VML injuries. The major implication of this work is that agent-based models may provide a much-needed predictive tool to optimize the design of new therapies, and thereby, accelerate the clinical translation of regenerative therapeutics for VML injuries.
Collapse
Affiliation(s)
- Amanda M. Westman
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
| | - George J. Christ
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| | - Silvia S. Blemker
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Ophthalmology, University of Virginia, Charlottesville, Virginia, United States of America
- Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail: (GJC); (SSB)
| |
Collapse
|
240
|
Norman MDA, Ferreira SA, Jowett GM, Bozec L, Gentleman E. Measuring the elastic modulus of soft culture surfaces and three-dimensional hydrogels using atomic force microscopy. Nat Protoc 2021; 16:2418-2449. [PMID: 33854255 PMCID: PMC7615740 DOI: 10.1038/s41596-021-00495-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 01/05/2021] [Indexed: 02/02/2023]
Abstract
Growing interest in exploring mechanically mediated biological phenomena has resulted in cell culture substrates and 3D matrices with variable stiffnesses becoming standard tools in biology labs. However, correlating stiffness with biological outcomes and comparing results between research groups is hampered by variability in the methods used to determine Young's (elastic) modulus, E, and by the inaccessibility of relevant mechanical engineering protocols to most biology labs. Here, we describe a protocol for measuring E of soft 2D surfaces and 3D hydrogels using atomic force microscopy (AFM) force spectroscopy. We provide instructions for preparing hydrogels with and without encapsulated live cells, and provide a method for mounting samples within the AFM. We also provide details on how to calibrate the instrument, and give step-by-step instructions for collecting force-displacement curves in both manual and automatic modes (stiffness mapping). We then provide details on how to apply either the Hertz or the Oliver-Pharr model to calculate E, and give additional instructions to aid the user in plotting data distributions and carrying out statistical analyses. We also provide instructions for inferring differential matrix remodeling activity in hydrogels containing encapsulated single cells or organoids. Our protocol is suitable for probing a range of synthetic and naturally derived polymeric hydrogels such as polyethylene glycol, polyacrylamide, hyaluronic acid, collagen, or Matrigel. Although sample preparation timings will vary, a user with introductory training to AFM will be able to use this protocol to characterize the mechanical properties of two to six soft surfaces or 3D hydrogels in a single day.
Collapse
Affiliation(s)
- Michael D. A. Norman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
| | - Silvia A. Ferreira
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
| | - Geraldine M. Jowett
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
| | - Laurent Bozec
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
- London Centre for Nanotechnology, London WC1H 0AH, UK
| |
Collapse
|
241
|
Tong Z, Jin L, Oliveira JM, Reis RL, Zhong Q, Mao Z, Gao C. Adaptable hydrogel with reversible linkages for regenerative medicine: Dynamic mechanical microenvironment for cells. Bioact Mater 2021; 6:1375-1387. [PMID: 33210030 PMCID: PMC7658331 DOI: 10.1016/j.bioactmat.2020.10.029] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Hydrogels are three-dimensional platforms that serve as substitutes for native extracellular matrix. These materials are starting to play important roles in regenerative medicine because of their similarities to native matrix in water content and flexibility. It would be very advantagoues for researchers to be able to regulate cell behavior and fate with specific hydrogels that have tunable mechanical properties as biophysical cues. Recent developments in dynamic chemistry have yielded designs of adaptable hydrogels that mimic dynamic nature of extracellular matrix. The current review provides a comprehensive overview for adaptable hydrogel in regenerative medicine as follows. First, we outline strategies to design adaptable hydrogel network with reversible linkages according to previous findings in supramolecular chemistry and dynamic covalent chemistry. Next, we describe the mechanism of dynamic mechanical microenvironment influence cell behaviors and fate, including how stress relaxation influences on cell behavior and how mechanosignals regulate matrix remodeling. Finally, we highlight techniques such as bioprinting which utilize adaptable hydrogel in regenerative medicine. We conclude by discussing the limitations and challenges for adaptable hydrogel, and we present perspectives for future studies.
Collapse
Affiliation(s)
- Zongrui Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017, Barco GMR, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017, Barco GMR, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Qi Zhong
- Key Laboratory of Advanced Textile Materials & Manufacturing Technology, Ministry of Education, National Base for International Science and Technology Cooperation in Textiles and Consumer-Goods Chemistry, Zhejiang Sci-Tech University, 310018, Hangzhou, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
242
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
243
|
Kjell J, Fischer-Sternjak J, Thompson AJ, Friess C, Sticco MJ, Salinas F, Cox J, Martinelli DC, Ninkovic J, Franze K, Schiller HB, Götz M. Defining the Adult Neural Stem Cell Niche Proteome Identifies Key Regulators of Adult Neurogenesis. Cell Stem Cell 2021; 26:277-293.e8. [PMID: 32032526 PMCID: PMC7005820 DOI: 10.1016/j.stem.2020.01.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/24/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022]
Abstract
The mammalian brain contains few niches for neural stem cells (NSCs) capable of generating new neurons, whereas other regions are primarily gliogenic. Here we leverage the spatial separation of the sub-ependymal zone NSC niche and the olfactory bulb, the region to which newly generated neurons from the sub-ependymal zone migrate and integrate, and present a comprehensive proteomic characterization of these regions in comparison to the cerebral cortex, which is not conducive to neurogenesis and integration of new neurons. We find differing compositions of regulatory extracellular matrix (ECM) components in the neurogenic niche. We further show that quiescent NSCs are the main source of their local ECM, including the multi-functional enzyme transglutaminase 2, which we show is crucial for neurogenesis. Atomic force microscopy corroborated indications from the proteomic analyses that neurogenic niches are significantly stiffer than non-neurogenic parenchyma. Together these findings provide a powerful resource for unraveling unique compositions of neurogenic niches.
Collapse
Affiliation(s)
- Jacob Kjell
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany
| | - Judith Fischer-Sternjak
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany
| | - Amelia J Thompson
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| | - Christian Friess
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany
| | - Matthew J Sticco
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Favio Salinas
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Jürgen Cox
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - David C Martinelli
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Jovica Ninkovic
- Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany; Division of Cell Biology and Anatomy, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; SYNERGY, Excellence Cluster Systems Neurology, Ludwig-Maximilians-Universitaet, Muenchen, Germany
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| | - Herbert B Schiller
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany; Institute of Lung Biology and Disease, Member of the German Center for Lung Research, Helmholtz Zentrum Muenchen, Germany
| | - Magdalena Götz
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany; SYNERGY, Excellence Cluster Systems Neurology, Ludwig-Maximilians-Universitaet, Muenchen, Germany.
| |
Collapse
|
244
|
Fernández-Garibay X, Ortega MA, Cerro-Herreros E, Comelles J, Martínez E, Artero R, Fernández-Costa JM, Ramón-Azcón J. Bioengineered in vitro3D model of myotonic dystrophy type 1 human skeletal muscle. Biofabrication 2021; 13. [PMID: 33836519 DOI: 10.1088/1758-5090/abf6ae] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common hereditary myopathy in the adult population. The disease is characterized by progressive skeletal muscle degeneration that produces severe disability. At present, there is still no effective treatment for DM1 patients, but the breakthroughs in understanding the molecular pathogenic mechanisms in DM1 have allowed the testing of new therapeutic strategies. Animal models andin vitrotwo-dimensional cell cultures have been essential for these advances. However, serious concerns exist regarding how faithfully these models reproduce the biological complexity of the disease. Biofabrication tools can be applied to engineer human three-dimensional (3D) culture systems that complement current preclinical research models. Here, we describe the development of the firstin vitro3D model of DM1 human skeletal muscle. Transdifferentiated myoblasts from patient-derived fibroblasts were encapsulated in micromolded gelatin methacryloyl-carboxymethyl cellulose methacrylate hydrogels through photomold patterning on functionalized glass coverslips. These hydrogels present a microstructured topography that promotes myoblasts alignment and differentiation resulting in highly aligned myotubes from both healthy and DM1 cells in a long-lasting cell culture. The DM1 3D microtissues recapitulate the molecular alterations detected in patient biopsies. Importantly, fusion index analyses demonstrate that 3D micropatterning significantly improved DM1 cell differentiation into multinucleated myotubes compared to standard cell cultures. Moreover, the characterization of the 3D cultures of DM1 myotubes detects phenotypes as the reduced thickness of myotubes that can be used for drug testing. Finally, we evaluated the therapeutic effect of antagomiR-23b administration on bioengineered DM1 skeletal muscle microtissues. AntagomiR-23b treatment rescues both molecular DM1 hallmarks and structural phenotype, restoring myotube diameter to healthy control sizes. Overall, these new microtissues represent an improvement over conventional cell culture models and can be used as biomimetic platforms to establish preclinical studies for myotonic dystrophy.
Collapse
Affiliation(s)
- Xiomara Fernández-Garibay
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - María A Ortega
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Estefanía Cerro-Herreros
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Jordi Comelles
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain
| | - Elena Martínez
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Department of Electronics and Biomedical Engineering, University of Barcelona (UB), c/Martí i Franquès 1-11, E08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, E28029 Madrid, Spain
| | - Rubén Artero
- University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Translational Genomics Group, Incliva Health Research Institute, Dr Moliner 50, E46100 Burjassot, Valencia, Spain.,Joint Unit Incliva- CIPF, Dr Moliner 50, E46100 Burjassot, Valencia, Spain
| | - Juan M Fernández-Costa
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain
| | - Javier Ramón-Azcón
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), c/Baldiri Reixac 10-12, E08028 Barcelona, Spain.,Institució Catalana de Reserca I Estudis Avançats (ICREA), Passeig de Lluís Companys, 23, E08010 Barcelona, Spain
| |
Collapse
|
245
|
Melino S, Seliktar D. A hydrogel reveals an elusive cancer stem cell. Cell Death Dis 2021; 12:415. [PMID: 33879776 PMCID: PMC8058059 DOI: 10.1038/s41419-021-03696-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Sonia Melino
- Department of Chemical Science and Technologies, University of Rome "Tor Vergata", Rome, Italy
| | - Dror Seliktar
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
246
|
Theret M, Rossi FMV, Contreras O. Evolving Roles of Muscle-Resident Fibro-Adipogenic Progenitors in Health, Regeneration, Neuromuscular Disorders, and Aging. Front Physiol 2021; 12:673404. [PMID: 33959042 PMCID: PMC8093402 DOI: 10.3389/fphys.2021.673404] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Normal skeletal muscle functions are affected following trauma, chronic diseases, inherited neuromuscular disorders, aging, and cachexia, hampering the daily activities and quality of life of the affected patients. The maladaptive accumulation of fibrous intramuscular connective tissue and fat are hallmarks of multiple pathologies where chronic damage and inflammation are not resolved, leading to progressive muscle replacement and tissue degeneration. Muscle-resident fibro-adipogenic progenitors are adaptable stromal cells with multilineage potential. They are required for muscle homeostasis, neuromuscular integrity, and tissue regeneration. Fibro-adipogenic progenitors actively regulate and shape the extracellular matrix and exert immunomodulatory functions via cross-talk with multiple other residents and non-resident muscle cells. Remarkably, cumulative evidence shows that a significant proportion of activated fibroblasts, adipocytes, and bone-cartilage cells, found after muscle trauma and disease, descend from these enigmatic interstitial progenitors. Despite the profound impact of muscle disease on human health, the fibrous, fatty, and ectopic bone tissues' origins are poorly understood. Here, we review the current knowledge of fibro-adipogenic progenitor function on muscle homeostatic integrity, regeneration, repair, and aging. We also discuss how scar-forming pathologies and disorders lead to dysregulations in their behavior and plasticity and how these stromal cells can control the onset and severity of muscle loss in disease. We finally explore the rationale of improving muscle regeneration by understanding and modulating fibro-adipogenic progenitors' fate and behavior.
Collapse
Affiliation(s)
- Marine Theret
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Biomedical Research Centre, Department of Medical Genetics, School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Osvaldo Contreras
- Departamento de Biología Celular y Molecular, Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, Australia
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| |
Collapse
|
247
|
Blackburn DM, Lazure F, Soleimani VD. SMART approaches for genome-wide analyses of skeletal muscle stem and niche cells. Crit Rev Biochem Mol Biol 2021; 56:284-300. [PMID: 33823731 DOI: 10.1080/10409238.2021.1908950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Darren M Blackburn
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
248
|
Ng S, Kurisawa M. Integrating biomaterials and food biopolymers for cultured meat production. Acta Biomater 2021; 124:108-129. [PMID: 33472103 DOI: 10.1016/j.actbio.2021.01.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
Cultured meat has recently achieved mainstream prominence due to the emergence of societal and industrial interest. In contrast to animal-based production of traditional meat, the cultured meat approach entails laboratory cultivation of engineered muscle tissue. However, bioengineers have hitherto engineered tissues to fulfil biomedical endpoints, and have had limited experience in engineering muscle tissue for its post-mortem traits, which broadly govern consumer definitions of meat quality. Furthermore, existing tissue engineering approaches face fundamental challenges in technical feasibility and industrial scalability for cultured meat production. This review discusses how animal-based meat production variables influence meat properties at both the molecular and functional level, and whether current cultured meat approaches recapitulate these properties. In addition, this review considers how conventional meat producers employ exogenous biopolymer-based meat ingredients and processing techniques to mimic desirable meat properties in meat products. Finally, current biomaterial strategies for engineering muscle and adipose tissue are surveyed in the context of emerging constraints that pertain to cultured meat production, such as edibility, sustainability and scalability, and potential areas for integrating biomaterials and food biopolymer approaches to address these constraints are discussed. STATEMENT OF SIGNIFICANCE: Laboratory-grown or cultured meat has gained increasing interest from industry and the public, but currently faces significant impediment to market feasibility. This is due to fundamental knowledge gaps in producing realistic meat tissues via conventional tissue engineering approaches, as well as translational challenges in scaling up these approaches in an efficient, sustainable and high-volume manner. By defining the molecular basis for desirable meat quality attributes, such as taste and texture, and introducing the fundamental roles of food biopolymers in mimicking these properties in conventional meat products, this review aims to bridge the historically disparate fields of meat science and biomaterials engineering in order to inspire potentially synergistic strategies that address some of these challenges.
Collapse
|
249
|
Nalbandian M, Zhao M, Sasaki-Honda M, Jonouchi T, Lucena-Cacace A, Mizusawa T, Yasuda M, Yoshida Y, Hotta A, Sakurai H. Characterization of hiPSC-Derived Muscle Progenitors Reveals Distinctive Markers for Myogenic Cell Purification Toward Cell Therapy. Stem Cell Reports 2021; 16:883-898. [PMID: 33798449 PMCID: PMC8072070 DOI: 10.1016/j.stemcr.2021.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 01/14/2023] Open
Abstract
The transplantation of muscle progenitor cells (MuPCs) differentiated from human induced pluripotent stem cells (hiPSCs) is a promising approach for treating skeletal muscle diseases such as Duchenne muscular dystrophy (DMD). However, proper purification of the MuPCs before transplantation is essential for clinical application. Here, by using MYF5 hiPSC reporter lines, we identified two markers for myogenic cell purification: CDH13, which purified most of the myogenic cells, and FGFR4, which purified a subset of MuPCs. Cells purified with each of the markers showed high efficiency for regeneration after transplantation and contributed to the restoration of dystrophin expression in DMD-immunodeficient model mice. Moreover, we found that MYF5 regulates CDH13 expression by binding to the promoter regions. These findings suggest that FGFR4 and CDH13 are strong candidates for the purification of hiPSC-derived MuPCs for therapeutical application. MYF5 and PAX7 mark different populations of hiPSC-MuPCs RNA-seq of MYF5+ cells reveals CDH13 and FGFR4 as hiPSC-MuPC markers CDH13+ and FGFR4+ hiPSC-MuPCs contribute to regeneration in mdx mice MYF5 regulates CDH13 expression by binding to its promoter region
Collapse
Affiliation(s)
- Minas Nalbandian
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mingming Zhao
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Mitsuru Sasaki-Honda
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuya Jonouchi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Antonio Lucena-Cacace
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takuma Mizusawa
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Masahiko Yasuda
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
250
|
Virdi JK, Pethe P. Biomaterials Regulate Mechanosensors YAP/TAZ in Stem Cell Growth and Differentiation. Tissue Eng Regen Med 2021; 18:199-215. [PMID: 33230800 PMCID: PMC8012461 DOI: 10.1007/s13770-020-00301-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/15/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
Tissue-resident stem cells are surrounded by a microenvironment known as 'stem cell niche' which is specific for each stem cell type. This niche comprises of cell-intrinsic and -extrinsic factors like biochemical and biophysical signals, which regulate stem cell characteristics and differentiation. Biochemical signals have been thoroughly studied however, the effect of biophysical signals on stem cell regulation is yet to be completely understood. Biomaterials have aided in addressing this issue since they can provide a defined and tuneable microenvironment resembling in vivo conditions. We review various biomaterials used in many studies which have shown a connection between biomaterial-generated mechanical signals and alteration in stem cell behaviour. Researchers probed to understand the mechanism of mechanotransduction and reported that the signals from the extracellular matrix regulate a transcription factor yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ), which is a downstream-regulator of the Hippo pathway and it transduces the mechanical signals inside the nucleus. We highlight the role of the YAP/TAZ as mechanotransducers in stem cell self-renewal and differentiation in response to substrate stiffness, also the possibility of mechanobiology as the emerging field of regenerative medicines and three-dimensional tissue printing.
Collapse
Affiliation(s)
- Jasmeet Kaur Virdi
- Department of Biological Science, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to-be) University, Mumbai, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International University, Lavale, Mulshi, Pune, 412115, India.
| |
Collapse
|