1
|
Wu T, Li Y, Liu Y, Chu CQ. Preclinical RA: How to halt its progression. Best Pract Res Clin Rheumatol 2025; 39:102030. [PMID: 39721896 DOI: 10.1016/j.berh.2024.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder with a complex pathogenesis that evolves through various stages before clinical symptoms emerge. This review outlines the natural history of RA, starting from genetic predisposition and environmental triggers to preclinical autoimmunity and subsequent joint inflammation. Key genetic factors interact with environmental elements like smoking and infections, producing autoantibodies such as anti-citrullinated protein antibodies (ACPA) and rheumatoid factor, which precede clinical manifestations by several years. The preclinical phases offer critical opportunities for intervention aiming at halting disease progression. Preventive strategies including lifestyle modifications, dietary interventions, and targeted immune modulation may halt the progression to clinical RA in those at-risk individuals.
Collapse
Affiliation(s)
- Tong Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Lecheng Hospital, Sichuan University, Boao, Hainan, 571435, China.
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA; Rheumatology Section, VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
2
|
Lu F, Liu Y, Dai Y, Zhang G, Tong Y. Preparation of nanosilver/polymer composites and evaluation of their antimicrobial and antitumor effect. RSC Adv 2025; 15:6357-6369. [PMID: 40008016 PMCID: PMC11855276 DOI: 10.1039/d4ra08108k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
In this study, a copolymer (PVA-g-PEG) of polyethylene glycol (PEG) and polyvinyl alcohol (PVA) was synthesized by grafting PEG chains onto PVA backbone. PVA-g-PEG was used as the carrier to prepare the silver nanoparticles/polymer composite (AgNPs/PVA-g-PEG) using a "one-pot" biological method in the presence of grape seeds extract as a reducing and stabilizing agent. In order to highlight the effect of the copolymer, the homo-polymers PVA and PEG were applied as the carriers to prepare the corresponding composites - AgNPs/PVA, and AgNPs/PEG, respectively using the same method. The prepared AgNPs/polymer products were characterized by UV absorption spectroscopy (UV-vis), scanning electron microscopy (SEM) and X-ray diffraction (XRD). The results show that the silver ions were successfully reduced by the grade seeds extract and the produced AgNPs are coated on the surface of AgNPs/PVA-g-PEG and AgNPs/PVA, but not for AgNPs/PEG. The prepared AgNPs are uniform and monodisperse, the particle size is small with mean diameter about 25.7 ± 2.3 nm and 54.2 ± 3.4 nm for AgNPs/PVA-g-PEG and AgNPs/PVA, respectively. The AgNPs/polymer composites exhibited superior antimicrobial effects against microorganisms (Escherichia coli and Staphylococcus aureus). AgNPs/PVA-g-PEG demonstrated a better performance than AgNPs/PVA. AgNPs/PVA-g-PEG had a minimum inhibitory concentration (MIC) of 1.3 μg mL-1 and a minimum inhibitory concentration (MBC) of 2.4 μg mL-1 against the microorganisms. For anti-tumor effect, AgNPs/PVA-g-PEG also demonstrated a high cytotoxicity to the colorectal cancerous cells HCT116 and SW620. The IC50 values of AgNPs/PVA-g-PEG for HCT116 and SW620 cell lines were 25.4 and 37.6 μg mL-1, respectively, suggesting a good anticancer activity. All above results indicate that AgNPs/PVA-g-PEG composites have a significant potential for the control of microorganisms and inhibition of cancer cells.
Collapse
Affiliation(s)
- FeiFei Lu
- College of Medicine and Biological Information Engineering, Northeastern University Shenyang Liaoning 110167 China
| | - Yuxin Liu
- Department of Nuclear Medicine, General Hospital of Northern Theater Command No. 83, Wenhua Road Shenyang 110016 China
| | - Yingxin Dai
- Department of Nuclear Medicine, General Hospital of Northern Theater Command No. 83, Wenhua Road Shenyang 110016 China
| | - Guoxu Zhang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command No. 83, Wenhua Road Shenyang 110016 China
| | - Yanan Tong
- Department of Nuclear Medicine, General Hospital of Northern Theater Command No. 83, Wenhua Road Shenyang 110016 China
| |
Collapse
|
3
|
Katoh K. Integrin and Its Associated Proteins as a Mediator for Mechano-Signal Transduction. Biomolecules 2025; 15:166. [PMID: 40001469 PMCID: PMC11853369 DOI: 10.3390/biom15020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/11/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Mechano-signal transduction is a process in which cells perceive extracellular mechanical signals, convert them into intracellular biochemical signals, and produce a response. Integrins are cell surface receptors that sense the extracellular mechanical cues and bind to the extracellular matrix (ECM). This binding induces integrin clustering and activation. Cytoplasmic tails of activated integrins interact and induce cytoskeleton tensions via several adaptor proteins. Integrins monitor extracellular stiffness via cytoskeleton tensions and modulate ECM stiffness via downstream signaling pathways regulating the expression of genes of ECM components. Integrin-mediated mechano-transduction is very crucial for the cell as it regulates the cell physiology both in normal and diseased conditions according to extracellular mechanical cues. It regulates cell proliferation, survival, and migration. Abnormal mechanical cues such as extreme and prolonged mechanical stress result in pathological conditions including fibrosis, cancers, skin, and autoimmune disorders. This paper aims to explore the role of integrins and their associated proteins in mechano-signal transduction. It highlights the integrins and their associated proteins as targets for therapy development. Furthermore, it also presents the challenges to the targeted drug development, which can be drug resistance and cytotoxicity. It is concluded in this paper that research on integrin-mediated mechano-signal transduction and its relationship with cell physiology and pathologies will be an important step towards the development of effective therapies.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, Tsukuba 305-8521, Japan
| |
Collapse
|
4
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
5
|
Latif A, Fisher LE, Dundas AA, Cuzzucoli Crucitti V, Imir Z, Lawler K, Pappalardo F, Muir BW, Wildman R, Irvine DJ, Alexander MR, Ghaemmaghami AM. Microparticles Decorated with Cell-Instructive Surface Chemistries Actively Promote Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2208364. [PMID: 36440539 DOI: 10.1002/adma.202208364] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/16/2022] [Indexed: 06/16/2023]
Abstract
Wound healing is a complex biological process involving close crosstalk between various cell types. Dysregulation in any of these processes, such as in diabetic wounds, results in chronic nonhealing wounds. Fibroblasts are a critical cell type involved in the formation of granulation tissue, essential for effective wound healing. 315 different polymer surfaces are screened to identify candidates which actively drive fibroblasts toward either pro- or antiproliferative functional phenotypes. Fibroblast-instructive chemistries are identified, which are synthesized into surfactants to fabricate easy to administer microparticles for direct application to diabetic wounds. The pro-proliferative microfluidic derived particles are able to successfully promote neovascularization, granulation tissue formation, and wound closure after a single application to the wound bed. These active novel bio-instructive microparticles show great potential as a route to reducing the burden of chronic wounds.
Collapse
Affiliation(s)
- Arsalan Latif
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Leanne E Fisher
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Adam A Dundas
- Faculty of Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | | | - Zeynep Imir
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Karen Lawler
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | | | - Benjamin W Muir
- Commonwealth Scientific & Industrial Research Organization, Canberra ACT 2601, Australia
| | - Ricky Wildman
- Faculty of Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Derek J Irvine
- Faculty of Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | | | | |
Collapse
|
6
|
Pallotta I, Stec MJ, Schriver B, Golann DR, Considine K, Su Q, Barahona V, Napolitano JE, Stanley S, Garcia M, Feric NT, Durney KM, Aschar‐Sobbi R, Bays N, Shavlakadze T, Graziano MP. Electrical stimulation of biofidelic engineered muscle enhances myotube size, force, fatigue resistance, and induces a fast-to-slow-phenotype shift. Physiol Rep 2024; 12:e70051. [PMID: 39384537 PMCID: PMC11464147 DOI: 10.14814/phy2.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 10/11/2024] Open
Abstract
Therapeutic development for skeletal muscle diseases is challenged by a lack of ex vivo models that recapitulate human muscle physiology. Here, we engineered 3D human skeletal muscle tissue in the Biowire II platform that could be maintained and electrically stimulated long-term. Increasing differentiation time enhanced myotube formation, modulated myogenic gene expression, and increased twitch and tetanic forces. When we mimicked exercise training by applying chronic electrical stimulation, the "exercised" skeletal muscle tissues showed increased myotube size and a contractility profile, fatigue resistance, and gene expression changes comparable to in vivo models of exercise training. Additionally, tissues also responded with expected physiological changes to known pharmacological treatment. To our knowledge, this is the first evidence of a human engineered 3D skeletal muscle tissue that recapitulates in vivo models of exercise. By recapitulating key features of human skeletal muscle, we demonstrated that the Biowire II platform may be used by the pharmaceutical industry as a model for identifying and optimizing therapeutic drug candidates that modulate skeletal muscle function.
Collapse
Affiliation(s)
| | | | | | | | | | - Qi Su
- Regeneron PharmaceuticalsTarrytownNew YorkUSA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Mercer IG, Yu K, Devanny AJ, Gordon MB, Kaufman LJ. Plasticity variable collagen-PEG interpenetrating networks modulate cell spreading. Acta Biomater 2024; 187:242-252. [PMID: 39218279 DOI: 10.1016/j.actbio.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The extracellular matrix protein collagen I has been used extensively in the field of biomaterials due to its inherent biocompatibility and unique viscoelastic and mechanical properties. Collagen I self-assembly into fibers and networks is environmentally sensitive to gelation conditions such as temperature, resulting in gels with distinct network architectures and mechanical properties. Despite this, collagen gels are not suitable for many applications given their relatively low storage modulus. We have prepared collagen-poly(ethylene glycol) [PEG] interpenetrating network (IPN) hydrogels to reinforce the collagen network, which also induces changes to network plasticity, a recent focus of study in cell-matrix interactions. Here, we prepare collagen/PEG IPNs, varying collagen concentration and collagen gelation temperature to assess changes in microarchitecture and mechanical properties of these networks. By tuning these parameters, IPNs with a range of stiffness, plasticity and pore size are obtained. Cell studies suggest that matrix plasticity is a key determinant of cell behavior, including cell elongation, on these gels. This work presents a natural/synthetic biocompatible matrix that retains the unique structural properties of collagen networks with increased storage modulus and tunable plasticity. The described IPN materials will be of use for applications in which control of cell spreading is desirable, as only minimal changes in sample preparation lead to changes in cell spreading and circularity. Additionally, this study contributes to our understanding of the connection between collagen self-assembly conditions and matrix structural and mechanical properties and presents them as useful tools for the design of other collagen based biomaterials. STATEMENT OF SIGNIFICANCE: We developed a collagen-poly(ethylene glycol) interpenetrating network (IPN) platform that retains native collagen architecture and biocompatibility but provides higher stiffness and tunable plasticity. With minor changes in collagen gelation temperature or concentration, IPN gels with a range of plasticity, storage modulus, and pore size can be obtained. The tunable plasticity of the gels is shown to modulate cell spreading, with a greater proportion of elongated cells on the most plastic of IPNs, supporting the assertion that matrix plasticity is a key determinant of cell spreading. The material can be of use for situations where control of cell spreading is desired with minimal intervention, and the findings herein may be used to develop similar collagen based IPN platforms.
Collapse
Affiliation(s)
- Iris G Mercer
- Department of Chemistry, Columbia University, New York, NY 10027, United States
| | - Karen Yu
- Department of Chemistry, Columbia University, New York, NY 10027, United States
| | - Alexander J Devanny
- Department of Chemistry, Columbia University, New York, NY 10027, United States
| | - Melissa B Gordon
- Department of Chemical and Biomolecular Engineering, Lafayette College, Easton, PA 18042, United States
| | - Laura J Kaufman
- Department of Chemistry, Columbia University, New York, NY 10027, United States.
| |
Collapse
|
8
|
Rodríguez-Mandujano L, Pimentel-Domínguez R, Tamariz E, Campos-Puente E, Giraldo-Betancur AL, Avila R. Fibrillogenesis in collagen hydrogels accelerated by carboxylated microbeads. Biomed Mater 2024; 19:045005. [PMID: 38688293 DOI: 10.1088/1748-605x/ad459a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 04/30/2024] [Indexed: 05/02/2024]
Abstract
Collagen type I is a material widely used for 3D cell culture and tissue engineering. Different architectures, such as gels, sponges, membranes, and nanofibers, can be fabricated with it. In collagen hydrogels, the formation of fibrils and fibers depends on various parameters, such as the source of collagen, pH, temperature, concentration, age, etc. In this work, we study the fibrillogenesis process in collagen type I hydrogels with different types of microbeads embedded, using optical techniques such as turbidity assay and confocal reflectance microscopy. We observe that microbeads embedded in the collagen matrix hydrogels modify the fibrillogenesis. Our results show that carboxylated fluorescent microbeads accelerate 3.6 times the gelation, while silica microbeads slow down the formation of collagen fibrils by a factor of 1.9, both compared to pure collagen hydrogels. Our observations suggest that carboxylate microbeads act as nucleation sites and the early collagen fibrils bind to the microbeads.
Collapse
Affiliation(s)
- Laura Rodríguez-Mandujano
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, 76230 Querétaro, Mexico
| | - Reinher Pimentel-Domínguez
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, 76230 Querétaro, Mexico
| | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico
| | - Edgar Campos-Puente
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, 76230 Querétaro, Mexico
| | - Astrid Lorena Giraldo-Betancur
- Centro de Investigación y de Estudios Avanzados del IPN, Unidad Querétaro, Libramiento Norponiente, #2000 C.P., 76230 Querétaro, Mexico
| | - Remy Avila
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, 76230 Querétaro, Mexico
| |
Collapse
|
9
|
Zhang B, Ma L, Tang LS, Song DW, Guo J, Zhang F, Xu X. In vitro and in vivo evaluation of a modified porcine acellular dermal matrix for soft tissue augmentation. J Biomater Appl 2023; 37:1497-1506. [PMID: 36469608 DOI: 10.1177/08853282221140667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To evaluate the effects of a modified porcine acellular dermal matrix (P-ADM), subepithelial connective tissue graft (SCTG) and other commercial bovine acellular dermal matrix membrane materials (B-ADM) on gingival soft tissue augmentation in the oral esthetic zone. MATERIAL AND METHODS The characteristics of P-ADM were observed by scanning electron microscope (SEM), Hematoxylin and eosin (H&E) and Masson's trichrome staining (Masson staining). The biocompatibility of P-ADM was verified by CCK8, phalloidin and living/dead cell staining. Beagle dog models were constructed and the thickness of gingiva was analyzed by the intraoral scanner. The morphology was observed by H&E and Masson staining. RESULTS Scanning electron microscopy, H&E and Masson staining showed that the P-ADM was mainly composed of collagen fibers, with no component of nuclear. The results of CCK8, phalloidin and living/dead cell staining indicated that the P-ADM had good cytocompatibility and no cytotoxicity. Human gingival fibroblasts were able to adhere and stretch on the surface of the material with pseudopodia. The SCTG group outperformed the B-ADM and P-ADM groups in terms of effectiveness, according to the analysis of digital oral scanning data at various time points following incremental soft tissue surgery. Compared with the B-ADM group, the effect of soft tissue increment was better in the P-ADM group. CONCLUSIONS P-ADM, as a biocompatible biomaterial, can be used as an alternative biomaterial for oral soft tissue thickening. However, the results of this study need to be verified by more clinical trials.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Health Care (Department of General Dentistry Ⅱ), School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases,12589 Jinan, China
| | - Li Ma
- Department of Health Care (Department of General Dentistry Ⅱ), School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases,12589 Jinan, China
| | - Lian Sheng Tang
- Shandong Provincial Key Laboratory of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Da Wei Song
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Jing Guo
- Endodontics, Taian Stomatology Hospital, Tai'an, China
| | - Fan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases12589, Jinan, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases12589, Jinan, China
| |
Collapse
|
10
|
Lu X, Han L, Kassab GS. Pulmonary Visceral Pleura Biomaterial: Elastin- and Collagen-Based Extracellular Matrix. Front Bioeng Biotechnol 2022; 10:796076. [PMID: 35433658 PMCID: PMC9006517 DOI: 10.3389/fbioe.2022.796076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Objective: The goal of the study is to determine the structural characteristics, mechanical properties, cytotoxicity, and biocompatibility of the pulmonary visceral pleura (PVP).Background: Collagen and elastin are the major components of the extracellular matrix. The PVP has an abundance of elastin and collagen that can serve as a potential biomaterial for clinical repair and reconstructions.Methods: The PVP was processed from swine and bovine lungs. Chemical analyses were used to determine collagen and elastin contents in the PVPs. Immunofluorescence microscopy was used to analyze the structure of the PVP. The stress–strain relationships and stress relaxation were determined by using the planar uniaxial test. The cytotoxicity of the PVP was tested in cultured cells. In in vivo evaluations, the PVP was implanted in the sciatic nerve and skin of rats.Results: Collagen and elastin contents are abundant in the PVP with larger proportions of elastin than in the bovine pericardium and porcine small intestinal submucosa. A microstructural analysis revealed that the elastin fibers were distributed throughout the PVP and the collagen was distributed mainly in the mesothelial basal lamina. The incremental moduli in stress–strain curves and relaxation moduli in the Maxwell–Wiechert model of PVP were approximately one-tenth of the bovine pericardium and small intestinal submucosa. The minimal cytotoxicity of the PVP was demonstrated. The axons proliferated in the PVP conduit guidance from proximal to distal sciatic nerves of rats. The neo-skin regenerated under the PVP skin substitute within 4 weeks.Conclusions: The PVP is composed of abundant collagen and elastin. The structural characteristics and mechanical compliance of the PVP render a suitable biological material for repair/reconstruction.
Collapse
|
11
|
Slater B, Li J, Indana D, Xie Y, Chaudhuri O, Kim T. Transient mechanical interactions between cells and viscoelastic extracellular matrix. SOFT MATTER 2021; 17:10274-10285. [PMID: 34137758 PMCID: PMC8695121 DOI: 10.1039/d0sm01911a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
During various physiological processes, such as wound healing and cell migration, cells continuously interact mechanically with a surrounding extracellular matrix (ECM). Contractile forces generated by the actin cytoskeleton are transmitted to a surrounding ECM, resulting in structural remodeling of the ECM. To better understand how matrix remodeling takes place, a myriad of in vitro experiments and simulations have been performed during recent decades. However, physiological ECMs are viscoelastic, exhibiting stress relaxation or creep over time. The time-dependent nature of matrix remodeling induced by cells remains poorly understood. Here, we employed a discrete model to investigate how the viscoelastic nature of ECMs affects matrix remodeling and stress profiles. In particular, we used explicit transient cross-linkers with varied density and unbinding kinetics to capture viscoelasticity unlike most of the previous models. Using this model, we quantified the time evolution of generation, propagation, and relaxation of stresses induced by a contracting cell in an ECM. It was found that matrix connectivity, regulated by fiber concentration and cross-linking density, significantly affects the magnitude and propagation of stress and subsequent matrix remodeling, as characterized by fiber displacements and local net deformation. In addition, we demonstrated how the base rate and force sensitivity of cross-linker unbinding regulate stress profiles and matrix remodeling. We verified simulation results using in vitro experiments performed with fibroblasts encapsulated in a three-dimensional collagen matrix. Our study provides key insights into the dynamics of physiologically relevant mechanical interactions between cells and a viscoelastic ECM.
Collapse
Affiliation(s)
- Brandon Slater
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN 47907, USA.
| | - Jing Li
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN 47907, USA.
| | - Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, 440 Escondido Mall, Stanford, CA, 94305, USA
| | - Yihao Xie
- School of Mechanical Engineering, Purdue University, 585 Purdue Mall, West Lafayette, IN 47907, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, 452 Escondido Mall, Stanford, CA, 94305, USA
| | - Taeyoon Kim
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN 47907, USA.
| |
Collapse
|
12
|
Køster D, Egedal JH, Lomholt S, Hvid M, Jakobsen MR, Müller-Ladner U, Eibel H, Deleuran B, Kragstrup TW, Neumann E, Nielsen MA. Phenotypic and functional characterization of synovial fluid-derived fibroblast-like synoviocytes in rheumatoid arthritis. Sci Rep 2021; 11:22168. [PMID: 34772990 PMCID: PMC8590001 DOI: 10.1038/s41598-021-01692-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
Fibroblast-like synoviocytes (FLS) play an important pathological role in persistent inflammatory joint diseases such as rheumatoid arthritis (RA). These cells have primarily been characterized in the RA synovial membrane. Here we aim to phenotypically and functionally characterize cultured synovial fluid-derived FLS (sfRA-FLS). Paired peripheral blood mononuclear cells (PBMC) and sfRA-FLS from patients with RA were obtained and monocultures of sfRA-FLS and autologous co-cultures of sfRA-FLS and PBMC were established. The in situ activated sfRA-FLS were CD34-, CD45-, Podoplanin+, Thymocyte differentiation antigen-1+. SfRA-FLS expressed uniform levels of NFкB-related pathway proteins and secreted several pro-inflammatory cytokines dominated by IL-6 and MCP-1. In a co-culture model with autologous PBMC, the ICAM-1 and HLA-DR expression on sfRA-FLS and secretion of IL-1β, IL-6, and MCP-1 increased. In vivo, human sfRA-FLS were cartilage invasive both at ipsilateral and contralateral implantation site. We conclude that, sfRA-FLS closely resemble the pathological sublining layer FLS subset in terms of surface protein expression, cytokine production and leukocyte cross-talk potential. Further, sfRA-FLS are comparable to tissue-derived FLS in their capabilities to invade cartilage at implantation sites but also spread tissue destruction to a distant site. Collectively, sfRA-FLS can serve as a an easy-to-obtain source of pathological sublining FLS in RA.
Collapse
Affiliation(s)
- Ditte Køster
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Johanne Hovgaard Egedal
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Søren Lomholt
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Malene Hvid
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin R Jakobsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
| | - Ulf Müller-Ladner
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Campus Kerckhoff, Bad Nauheim, Germany
| | - Hermann Eibel
- Department of Rheumatology and Clinical Immunology and Center for Chronic Immunodeficiency, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Tue Wenzel Kragstrup
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | - Elena Neumann
- Department of Internal Medicine and Rheumatology, Justus-Liebig-University Giessen, Campus Kerckhoff, Bad Nauheim, Germany
| | - Morten Aagaard Nielsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus, Denmark.
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
13
|
Exogenous extracellular matrix proteins decrease cardiac fibroblast activation in stiffening microenvironment through CAPG. J Mol Cell Cardiol 2021; 159:105-119. [PMID: 34118218 PMCID: PMC10066715 DOI: 10.1016/j.yjmcc.2021.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/20/2022]
Abstract
Controlling fibrosis is an essential part of regenerating the post-ischemic heart. In the post-ischemic heart, fibroblasts differentiate to myofibroblasts that produce collagen-rich matrix to physically stabilize the infarct area. Infarct models in adult mice result in permanent scarring unlike newborn animals which fully regenerate. Decellularized extracellular matrix (dECM) hydrogels derived from early-aged hearts have been shown to be a transplantable therapy that preserves heart function and stimulates cardiomyocyte proliferation and vascularization. In this study, we investigate the anti-fibrotic effects of injectable dECM hydrogels in a cardiac explant model in the context of age-associated tissue compliance. Treatments with adult and fetal dECM hydrogels were tested for molecular effects on cardiac fibroblast activation and fibrosis. Altered sensitivity of fibroblasts to the mechanosignaling of the remodeling microenvironment was evaluated by manipulating the native extracellular matrix in explants and also with elastomeric substrates in the presence of dECM hydrogels. The injectable fetal dECM hydrogel treatment decreases fibroblast activation and contractility and lowers the stiffness-mediated increases in fibroblast activation observed in stiffened explants. The anti-fibrotic effect of dECM hydrogel is most observable at highest stiffness. Experiments with primary cells on elastomeric substrates with dECM treatment support this phenomenon. Transcriptome analysis indicated that dECM hydrogels affect cytoskeleton related signaling including Macrophage capping protein (CAPG) and Leupaxin (LPXN). CAPG was down-regulated by the fetal dECM hydrogel. LPXN expression was decreased by stiffening the explants; however, this effect was reversed by dECM hydrogel treatment. Pharmacological disruption of cytoskeleton polymerization lowered fibroblast activation and CAPG levels. Knocking down CAPG expression with siRNA inhibited fibroblast activation and collagen deposition. Collectively, fibroblast activation is dependent on cooperative action of extracellular molecular signals and mechanosignaling by cytoskeletal integrity.
Collapse
|
14
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
15
|
Kim JY, Yang KM, Youn JH, Park H, Hahn HM, Lee IJ. In Vitro Analysis of Histology, Mechanics, and Safety of Radiation-free Pre-hydrated Human Acellular Dermal Matrix. J Breast Cancer 2020; 23:635-646. [PMID: 33408889 PMCID: PMC7779726 DOI: 10.4048/jbc.2020.23.e64] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/28/2020] [Indexed: 11/30/2022] Open
Abstract
Purpose Acellular dermal matrix (ADM) supports tissue expanders or implants in implant-based breast reconstruction. The characteristics of ADM tissue are defined by the manufacturing procedure, such as decellularization, preservation, and sterilization, and are directly related to clinical outcomes. This study aimed to compare the properties of a new pre-hydrated-ADM (H-ADM-low) obtained using a decellularization reagent reduction process with a low concentration of detergent with those of radiation-sterilized H-ADM and freeze-dried ADM (FD-ADM). Methods ADMs were evaluated in terms of structure, mechanical quality, and cytotoxicity using histochemical staining, tensile strength testing, and in vitro cell viability analysis. Results The tissue structure of H-ADM-low (CGDERM ONE-STEP) was similar to that of native skin despite complete decellularization. By contrast, in FD-ADM, the tissue structure was damaged by the freeze-drying process, and radiation-sterilized H-ADM showed a compact fibrillar arrangement. Furthermore, matrix components such as collagen and elastin were preserved in H-ADM-low, whereas a loss of elastin fibers with fragmented distribution was observed in radiation-sterilized H-ADMs. H-ADM-low's tensile strength (58.84 MPa) was significantly greater than that of FD-ADM (38.60 MPa) and comparable with that of radiation-sterilized H-ADMs. The residual detergent content in H-ADM-low (47.45 mg/L) was 2.67-fold lower than that of H-ADM decellularized with a conventional detergent concentration (126.99 mg/mL), and this finding was consistent with the cell viability results (90.7% and 70.7%, respectively), indicating that H-ADM-low has very low cytotoxicity. Conclusions H-ADM-low produced through aseptic processes retains the original tissue structure, demonstrates excellent mechanical properties, and does not affect cell viability. Therefore, this newer H-ADM is suitable for use in implant-based breast reconstruction.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Surgery, Ajou University Hospital, Suwon, Korea
| | - Kyung Min Yang
- Department of Plastic and Reconstructive Surgery, Ajou University School of Medicine, Suwon, Korea
| | | | | | - Hyung Min Hahn
- Department of Plastic and Reconstructive Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Il Jae Lee
- Department of Plastic and Reconstructive Surgery, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
16
|
Lai Benjamin FL, Lu Rick X, Hu Y, Davenport HL, Dou W, Wang EY, Radulovich N, Tsao MS, Sun Y, Radisic M. Recapitulating pancreatic tumor microenvironment through synergistic use of patient organoids and organ-on-a-chip vasculature. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2000545. [PMID: 33692660 PMCID: PMC7939064 DOI: 10.1002/adfm.202000545] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Tumor progression relies heavily on the interaction between the neoplastic epithelial cells and their surrounding stromal partners. This cell cross-talk affects stromal development, and ultimately the heterogeneity impacts drug efflux and efficacy. To mimic this evolving paradigm, we have micro-engineered a three-dimensional (3D) vascularized pancreatic adenocarcinoma tissue in a tri-culture system composed of patient derived pancreatic organoids, primary human fibroblasts and endothelial cells on a perfusable InVADE platform situated in a 96-well plate. Uniquely, through synergistic engineering we combined the benefits of cellular fidelity of patient tumor derived organoids with the addressability of a plastic organ-on-a-chip platform. Validation of this platform included demonstrating the growth of pancreatic tumor organoids by monitoring the change in metabolic activity of the tissue. Investigation of tumor microenvironmental behavior highlighted the role of fibroblasts in symbiosis with patient organoid cells, resulting in a six-fold increase of collagen deposition and a corresponding increase in tissue stiffness in comparison to fibroblast free controls. The value of a perfusable vascular network was evident in drug screening, as perfusion of gemcitabine into a stiffened matrix did not show the dose-dependent effects on tumor viability as those under static conditions. These findings demonstrate the importance of studying the dynamic synergistic relationship between patient cells with stromal fibroblasts, in a 3D perfused vascular network, to accurately understand and recapitulate the tumor microenvironment.
Collapse
Affiliation(s)
- F L Lai Benjamin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - X Lu Rick
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Yangshuo Hu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Huyer Locke Davenport
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Wenkun Dou
- Material Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Erika Y Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Nikolina Radulovich
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ming S Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yu Sun
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Material Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
17
|
González-Larraza PG, López-Goerne TM, Padilla-Godínez FJ, González-López MA, Hamdan-Partida A, Gómez E. IC 50 Evaluation of Platinum Nanocatalysts for Cancer Treatment in Fibroblast, HeLa, and DU-145 Cell Lines. ACS OMEGA 2020; 5:25381-25389. [PMID: 33043218 PMCID: PMC7542800 DOI: 10.1021/acsomega.0c03759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 09/09/2020] [Indexed: 05/27/2023]
Abstract
Cancer is a major public health problem being one of the main causes of morbidity and mortality today. Recent advances in catalytic nanomedicine have offered new cancer therapies based on the administration of nanoparticles (NPs) of platinum (Pt) dispersed in catalytic mesoporous nanomaterials (titania, TiO2) with highly selective cytotoxic properties and no adverse effects. A half maximal inhibitory concentration (IC50) study was carried out in cancerous cell lines (HeLa, DU-145, and fibroblasts) to evaluate the cytotoxic effect of different nanomaterials [Pt/TiO2, TiO2, and Pt(acac)2] synthesized by the sol-gel method at concentrations 0-1000 μg/mL. The assays showed that IC50 values for Pt in functionalized TiO2 (NPt) in HeLa (53.74 ± 2.95 μg/mL) and DU-145 (75.07 ± 5.48 μg/mL) were lower than those of pure TiO2 (74.29 ± 8.95 and 82.02 ± 6.03 μg/mL, respectively). Pt(acac)2 exhibited no cytotoxicity. Normal cells (fibroblasts) treated with NPt exhibited no significant growth inhibition, suggesting the high selectivity of the compound for cancerous cells only. TiO2 and NPt were identified as antineoplastic compounds in vitro. Pt(acac)2 is not recommendable because of the low cytotoxicity observed.
Collapse
Affiliation(s)
- Pamela G. González-Larraza
- Department of Health Care, Autonomous Metropolitan
University Xochimilco, Coyoacan, Mexico City 04960, Mexico
| | - Tessy M. López-Goerne
- Department of Health Care, Autonomous Metropolitan
University Xochimilco, Coyoacan, Mexico City 04960, Mexico
| | - Francisco J. Padilla-Godínez
- Department of Health Care, Autonomous Metropolitan
University Xochimilco, Coyoacan, Mexico City 04960, Mexico
- Department of Mathematics and Physics, Western Institute of Technology and Higher Education, San Pedro Tlaquepaque, Jalisco 45604, Mexico
| | - Marco A. González-López
- Department of Health Care, Autonomous Metropolitan
University Xochimilco, Coyoacan, Mexico City 04960, Mexico
| | - Aida Hamdan-Partida
- Department of Health Care, Autonomous Metropolitan
University Xochimilco, Coyoacan, Mexico City 04960, Mexico
| | - Esteban Gómez
- AG Nanooptik, Humboldt-Universtät zu Berlin, Berlin 10117, Germany
| |
Collapse
|
18
|
Bretherton R, Bugg D, Olszewski E, Davis J. Regulators of cardiac fibroblast cell state. Matrix Biol 2020; 91-92:117-135. [PMID: 32416242 PMCID: PMC7789291 DOI: 10.1016/j.matbio.2020.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/13/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023]
Abstract
Fibroblasts are the primary regulator of cardiac extracellular matrix (ECM). In response to disease stimuli cardiac fibroblasts undergo cell state transitions to a myofibroblast phenotype, which underlies the fibrotic response in the heart and other organs. Identifying regulators of fibroblast state transitions would inform which pathways could be therapeutically modulated to tactically control maladaptive extracellular matrix remodeling. Indeed, a deeper understanding of fibroblast cell state and plasticity is necessary for controlling its fate for therapeutic benefit. p38 mitogen activated protein kinase (MAPK), which is part of the noncanonical transforming growth factor β (TGFβ) pathway, is a central regulator of fibroblast to myofibroblast cell state transitions that is activated by chemical and mechanical stress signals. Fibroblast intrinsic signaling, local and global cardiac mechanics, and multicellular interactions individually and synergistically impact these state transitions and hence the ECM, which will be reviewed here in the context of cardiac fibrosis.
Collapse
Affiliation(s)
- Ross Bretherton
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Darrian Bugg
- Department of Pathology, University of Washington, 850 Republican, #343, Seattle, WA 98109, United States
| | - Emily Olszewski
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States; Department of Pathology, University of Washington, 850 Republican, #343, Seattle, WA 98109, United States; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98109, United States; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, United States.
| |
Collapse
|
19
|
Coupling of Fibrin Reorganization and Fibronectin Patterning by Corneal Fibroblasts in Response to PDGF BB and TGFβ1. Bioengineering (Basel) 2020; 7:bioengineering7030089. [PMID: 32784578 PMCID: PMC7552779 DOI: 10.3390/bioengineering7030089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 11/17/2022] Open
Abstract
We previously reported that corneal fibroblasts within 3D fibrin matrices secrete, bind, and organize fibronectin into tracks that facilitate cell spreading and migration. Other cells use these fibronectin tracks as conduits, which leads to the development of an interconnected cell/fibronectin network. In this study, we investigate how cell-induced reorganization of fibrin correlates with fibronectin track formation in response to two growth factors present during wound healing: PDGF BB, which stimulates cell spreading and migration; and TGFβ1, which stimulates cellular contraction and myofibroblast transformation. Both PDGF BB and TGFβ1 stimulated global fibrin matrix contraction (p < 0.005); however, the cell and matrix patterning were different. We found that, during PDGF BB-induced cell spreading, fibronectin was organized simultaneously with the generation of tractional forces at the leading edge of pseudopodia. Over time this led to the formation of an interconnected network consisting of cells, fibronectin and compacted fibrin tracks. Following culture in TGFβ1, cells were less motile, produced significant local fibrin reorganization, and formed fewer cellular connections as compared to PDGF BB (p < 0.005). Although bands of compacted fibrin tracks developed in between neighboring cells, fibronectin labeling was not generally present along these tracks, and the correlation between fibrin and fibronectin labeling was significantly less than that observed in PDGF BB (p < 0.001). Taken together, our results show that cell-induced extracellular matrix (ECM) reorganization can occur independently from fibronectin patterning. Nonetheless, both events seem to be coordinated, as corneal fibroblasts in PDGF BB secrete and organize fibronectin as they preferentially spread along compacted fibrin tracks between cells, producing an interconnected network in which cells, fibronectin and compacted fibrin tracks are highly correlated. This mechanism of patterning could contribute to the formation of organized cellular networks that have been observed following corneal injury and refractive surgery.
Collapse
|
20
|
Tang VW. Collagen, stiffness, and adhesion: the evolutionary basis of vertebrate mechanobiology. Mol Biol Cell 2020; 31:1823-1834. [PMID: 32730166 PMCID: PMC7525820 DOI: 10.1091/mbc.e19-12-0709] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023] Open
Abstract
The emergence of collagen I in vertebrates resulted in a dramatic increase in the stiffness of the extracellular environment, supporting long-range force propagation and the development of low-compliant tissues necessary for the development of vertebrate traits including pressurized circulation and renal filtration. Vertebrates have also evolved integrins that can bind to collagens, resulting in the generation of higher tension and more efficient force transmission in the extracellular matrix. The stiffer environment provides an opportunity for the vertebrates to create new structures such as the stress fibers, new cell types such as endothelial cells, new developmental processes such as neural crest delamination, and new tissue organizations such as the blood-brain barrier. Molecular players found only in vertebrates allow the modification of conserved mechanisms as well as the design of novel strategies that can better serve the physiological needs of the vertebrates. These innovations collectively contribute to novel morphogenetic behaviors and unprecedented increases in the complexities of tissue mechanics and functions.
Collapse
Affiliation(s)
- Vivian W. Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana–Champaign, Urbana, IL 61801
| |
Collapse
|
21
|
Shakiba D, Alisafaei F, Savadipour A, Rowe RA, Liu Z, Pryse KM, Shenoy VB, Elson EL, Genin GM. The Balance between Actomyosin Contractility and Microtubule Polymerization Regulates Hierarchical Protrusions That Govern Efficient Fibroblast-Collagen Interactions. ACS NANO 2020; 14:7868-7879. [PMID: 32286054 DOI: 10.1021/acsnano.9b09941] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Fibroblasts undergo a critical transformation from an initially inactive state to a morphologically different and contractile state after several hours of being embedded within a physiologically relevant three-dimensional (3D) fibrous collagen-based extracellular matrix (ECM). However, little is known about the critical mechanisms by which fibroblasts adapt themselves and their microenvironment in the earliest stage of cell-matrix interaction. Here, we identified the mechanisms by which fibroblasts interact with their 3D collagen fibrous matrices in the early stages of cell-matrix interaction and showed that fibroblasts use energetically efficient hierarchical micro/nano-scaled protrusions in these stages as the primary means for the transformation and adaptation. We found that actomyosin contractility in these protrusions in the early stages of cell-matrix interaction restricts the growth of microtubules by applying compressive forces on them. Our results show that actomyosin contractility and microtubules work in concert in the early stages of cell-matrix interaction to adapt fibroblasts and their microenvironment to one another. These early stage interactions result in responses to disruption of the microtubule network and/or actomyosin contractility that are opposite to well-known responses to late-stage disruption and reveal insight into the ways that cells adapt themselves and their ECM recursively.
Collapse
Affiliation(s)
- Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| | - Farid Alisafaei
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alireza Savadipour
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| | - Roger A Rowe
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| | - Zhangao Liu
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| | - Kenneth M Pryse
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| | - Vivek B Shenoy
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Elliot L Elson
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Guy M Genin
- NSF Science and Technology Center for Engineering Mechanobiology and Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri 63130 United States
| |
Collapse
|
22
|
The Mechanical Microenvironment in Breast Cancer. Cancers (Basel) 2020; 12:cancers12061452. [PMID: 32503141 PMCID: PMC7352870 DOI: 10.3390/cancers12061452] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 01/22/2023] Open
Abstract
Mechanotransduction is the interpretation of physical cues by cells through mechanosensation mechanisms that elegantly translate mechanical stimuli into biochemical signaling pathways. While mechanical stress and their resulting cellular responses occur in normal physiologic contexts, there are a variety of cancer-associated physical cues present in the tumor microenvironment that are pathological in breast cancer. Mechanistic in vitro data and in vivo evidence currently support three mechanical stressors as mechanical modifiers in breast cancer that will be the focus of this review: stiffness, interstitial fluid pressure, and solid stress. Increases in stiffness, interstitial fluid pressure, and solid stress are thought to promote malignant phenotypes in normal breast epithelial cells, as well as exacerbate malignant phenotypes in breast cancer cells.
Collapse
|
23
|
Vaughan MB, Xu G, Morris TL, Kshetri P, Herwig JX. Predictable fibroblast tension generation by measuring compaction of anchored collagen matrices using microscopy and optical coherence tomography. Cell Adh Migr 2019; 13:303-314. [PMID: 31331232 PMCID: PMC6650198 DOI: 10.1080/19336918.2019.1644855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The anchored fibroblast-populated collagen matrix (aFPCM) is an appropriate model to study fibrocontractive disease mechanisms. Our goal was to determine if aFPCM height reduction (compaction) during development is sufficient to predict tension generation. Compaction was quantified daily by both traditional light microscopy and an optical coherence tomography (OCT) system. Contraction in aFPCM was revealed by releasing them from anchorage. We found that aFPCM contraction increase was correlated to the compaction increase. Cytochalasin D treatment reversibly inhibited compaction. Therefore, we demonstrated that aFPCM height reduction efficiently measures compaction, contraction, and relative maturity of the collagen matrix during development or treatment. In addition, we showed that OCT is suitable for effectively imaging the cross-sectional morphology of the aFPCM in culture. This study will pave the way for more efficient studies on the mechanisms of (and treatments that target) migration and contraction in wound healing and Dupuytren’s contracture in a tissue environment.
Collapse
Affiliation(s)
- Melville B Vaughan
- a Department of Biology, University of Central Oklahoma , Edmond , OK , USA.,b Center for Interdisciplinary Biomedical Education and Research (CIBER), University of Central Oklahoma, 100 N. University Drive , Edmond , OK
| | - Gang Xu
- b Center for Interdisciplinary Biomedical Education and Research (CIBER), University of Central Oklahoma, 100 N. University Drive , Edmond , OK.,c Department of Engineering and Physics, University of Central Oklahoma , Edmond , OK , USA
| | - Tracy L Morris
- b Center for Interdisciplinary Biomedical Education and Research (CIBER), University of Central Oklahoma, 100 N. University Drive , Edmond , OK.,d Department of Mathematics and Statistics, University of Central Oklahoma , Edmond , OK , USA
| | - Pratiksha Kshetri
- a Department of Biology, University of Central Oklahoma , Edmond , OK , USA
| | - Jing X Herwig
- a Department of Biology, University of Central Oklahoma , Edmond , OK , USA
| |
Collapse
|
24
|
Elson EL, Qian H, Fee JA, Wakatsuki T. A model for positive feedback control of the transformation of fibroblasts to myofibroblasts. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 144:30-40. [PMID: 30174171 PMCID: PMC11033709 DOI: 10.1016/j.pbiomolbio.2018.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/31/2018] [Accepted: 08/13/2018] [Indexed: 12/22/2022]
Abstract
The phenotypic conversion of normal fibroblasts to myofibroblasts is central to normal wound healing and to pathological fibrosis that can occur in the heart and many other tissues. The transformation occurs in two stages. The first stage is driven mainly by mechanical changes such as increased stiffness of the heart due to hypertension and cellular contractility. The second stage requires both increasing stiffness and biochemical factors such as the growth factor, TGFβ. As more and more cells convert from weakly contractile fibroblasts to strongly contractile myofibroblasts, the stiffness of the ventricular muscle increases. We propose a simple model for the establishment of non-equilibrium steady states with different compositions of fibroblasts and myofibroblasts. Under some conditions a positive feedback loop resulting from the increasing stiffness caused by increasing numbers of myofibroblasts can produce a bifurcation between steady states with low and high myofibroblast content. We illustrate the large mechanical differences between normal fibroblasts and myofibroblasts with measurements in engineered tissue constructs.
Collapse
Affiliation(s)
- Elliot L Elson
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, School of Medicine, Campus Box 8231, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA.
| | - Hong Qian
- Department of Applied Mathematics, University of Washington, Lewis Hall 201 Box 353925, Seattle, WA, 98195, USA
| | - Judy A Fee
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, School of Medicine, Campus Box 8231, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA
| | | |
Collapse
|
25
|
Pinet K, McLaughlin KA. Mechanisms of physiological tissue remodeling in animals: Manipulating tissue, organ, and organism morphology. Dev Biol 2019; 451:134-145. [DOI: 10.1016/j.ydbio.2019.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/21/2022]
|
26
|
Hoffmann GA, Wong JY, Smith ML. On Force and Form: Mechano-Biochemical Regulation of Extracellular Matrix. Biochemistry 2019; 58:4710-4720. [PMID: 31144496 DOI: 10.1021/acs.biochem.9b00219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The extracellular matrix is well-known for its structural role in supporting cells and tissues, and its important biochemical role in providing signals to cells has increasingly become apparent. These structural and biochemical roles are closely coupled through mechanical forces: the biochemistry of the extracellular matrix determines its mechanical properties, mechanical forces control release or display of biochemical signals from the extracellular matrix, and the mechanical properties of the matrix in turn influence the mechanical set point at which signals are sent. In this Perspective, we explain how the extracellular matrix is regulated by strain and mechanical forces. We show the impact of biochemistry and mechanical forces on in vivo assembly of extracellular matrix and illustrate how matrix can be generated in vitro using a variety of methods. We cover how the matrix can be characterized in terms of mechanics, composition, and conformation to determine its properties and to predict interactions. Finally, we explore how extracellular matrix remodeling, ligand binding, and hemostasis are regulated by mechanical forces. These recently discovered mechano-biochemical interactions have important functions in wound healing and disease progression. It is likely that mechanically altered extracellular matrix interactions are a commonly recurring theme, but due to limited tools to generate extracellular matrix fibers in vitro and lack of high-throughput methods to detect these interactions, it is hypothesized that many of these interactions have yet to be discovered.
Collapse
Affiliation(s)
- Gwendolyn A Hoffmann
- Department of Biomedical Engineering , Boston University , 44 Cummington Mall , Boston , Massachusetts 02215 , United States
| | - Joyce Y Wong
- Department of Biomedical Engineering , Boston University , 44 Cummington Mall , Boston , Massachusetts 02215 , United States
| | - Michael L Smith
- Department of Biomedical Engineering , Boston University , 44 Cummington Mall , Boston , Massachusetts 02215 , United States
| |
Collapse
|
27
|
Zhang W, Xin Y, Yin B, Ye GL, Wang JX, Shen JF, Li L, Yang QH. Synthesis and properties of crosslinked carboxymethyl chitosan and its hemostatic and wound healing effects on liver injury of rats. J Biomater Appl 2019; 34:442-450. [PMID: 31137997 DOI: 10.1177/0885328219852888] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Wei Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Yu Xin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Bin Yin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Guan L Ye
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Jun X Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Jian F Shen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Lei Li
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Qi H Yang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| |
Collapse
|
28
|
Chia CY, Medeiros AD, Corraes ADMS, Manso JEF, Silva CSCD, Takiya CM, Vanz RL. Healing effect of andiroba-based emulsion in cutaneous wound healing via modulation of inflammation and transforming growth factor beta 31. Acta Cir Bras 2018; 33:1000-1015. [PMID: 30517327 DOI: 10.1590/s0102-865020180110000007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To evaluate the effects and mechanisms of andiroba-based emulsion (ABE) topical treatment on full-thickness cutaneous wounds in rats. METHODS The wounds were harvested on days 3, 7, 15, and 20 post-surgery. Wound contraction rate, quantitative immunohistochemistry [macrophages, myofibroblasts, capillaries, collagens (col) I and III, transforming growth factor β3β (TGFβ3)], and tensile strength were assessed. RESULTS Treated wounds were smaller, contracted earlier and had increased angiogenesis, fewer CD68+ and M2 macrophages on days 7 and 15, but higher on day 20. Myofibroblasts appeared on days 3 to 7 in untreated wounds and on days 7 to 15 in treated wounds. TGFβ3 levels were higher in the treated wounds, less dense collagen fibers, lower col I/III ratios and a higher tensile strength. CONCLUSION These results demonstrate the important anti-inflammatory role of treatment and the associated modulation of macrophages, myofibroblasts, and TGFβ3 levels. Collagen fibers in the treated wounds were more organized and less dense, similar to unwounded skin, which likely contributed to the higher tensile strength.
Collapse
Affiliation(s)
- Chang Yung Chia
- MD, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, and Immunopathology Laboratory, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Brazil. Conception and design of the study, analysis and interpretation of data, statistics analysis, technical procedures, manuscript writing, critical revision
| | - Andréia Dantas Medeiros
- PhD, Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro-RJ, Brazil. Technical procedures, manuscript writing, critical revision, final approval
| | - André de Menezes Silva Corraes
- PhD, Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro-RJ, Brazil. Technical procedures, manuscript writing, critical revision, final approval
| | - José Eduardo Ferreira Manso
- PhD, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, UFRJ, Rio de Janeiro-RJ, Brazil. Design of the study, analysis and interpretation of data, final approval
| | - César Silveira Claudio da Silva
- PhD, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, UFRJ, Rio de Janeiro-RJ, Brazil. Design of the study, analysis and interpretation of data, final approval
| | - Christina Maeda Takiya
- PhD, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, and Immunopathology Laboratory, Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro-RJ, Brazil. Design of the study, analysis and interpretation of data, immunohistochemical examinations, statistics analysis, manuscript writing, critical revision, final approval
| | - Ricardo Luís Vanz
- MD, Postgraduate Program in Surgical Science, Department of Surgery, School of Medicine, UFRJ, Rio de Janeiro-RJ, Brazil. Manuscript writing, critical revision, final approval
| |
Collapse
|
29
|
Kwak BS, Choi W, Jeon JW, Won JI, Sung GY, Kim B, Sung JH. In vitro 3D skin model using gelatin methacrylate hydrogel. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2018.05.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
|
31
|
Barnes LA, Marshall CD, Leavitt T, Hu MS, Moore AL, Gonzalez JG, Longaker MT, Gurtner GC. Mechanical Forces in Cutaneous Wound Healing: Emerging Therapies to Minimize Scar Formation. Adv Wound Care (New Rochelle) 2018; 7:47-56. [PMID: 29392093 PMCID: PMC5792236 DOI: 10.1089/wound.2016.0709] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/15/2016] [Indexed: 12/25/2022] Open
Abstract
Significance: Excessive scarring is major clinical and financial burden in the United States. Improved therapies are necessary to reduce scarring, especially in patients affected by hypertrophic and keloid scars. Recent Advances: Advances in our understanding of mechanical forces in the wound environment enable us to target mechanical forces to minimize scar formation. Fetal wounds experience much lower resting stress when compared with adult wounds, and they heal without scars. Therapies that modulate mechanical forces in the wound environment are able to reduce scar size. Critical Issues: Increased mechanical stresses in the wound environment induce hypertrophic scarring via activation of mechanotransduction pathways. Mechanical stimulation modulates integrin, Wingless-type, protein kinase B, and focal adhesion kinase, resulting in cell proliferation and, ultimately, fibrosis. Therefore, the development of therapies that reduce mechanical forces in the wound environment would decrease the risk of developing excessive scars. Future Directions: The development of novel mechanotherapies is necessary to minimize scar formation and advance adult wound healing toward the scarless ideal. Mechanotransduction pathways are potential targets to reduce excessive scar formation, and thus, continued studies on therapies that utilize mechanical offloading and mechanomodulation are needed.
Collapse
Affiliation(s)
- Leandra A. Barnes
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Clement D. Marshall
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Tripp Leavitt
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael S. Hu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
- Department of Surgery, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | | | - Jennifer G. Gonzalez
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
32
|
Parviz M, Toshniwal P, Viola HM, Hool LC, Fear PMW, Wood FM, Gaus K, Iyer KS, Gooding JJ. Real-Time Bioimpedance Sensing of Antifibrotic Drug Action in Primary Human Cells. ACS Sens 2017; 2:1482-1490. [PMID: 28871791 DOI: 10.1021/acssensors.7b00442] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Fibrotic diseases are among the most serious health issues with severe burdens due to their chronic nature and a large number of patients suffering from the debilitating effects and long-term sequelae. Collagenase treatment is a nonsurgical option but has limited results. To date, there is no potent noninvasive solution for fibrosis. Part of the reason for this is the lack of appropriate in vitro live cell screening tools to assess the efficacy of new therapeutical agents. Here, we demonstrate the utility of a cell-based electrochemical impedance biosensor platform to screen the efficacy of potential antifibrotic compounds. The platform employs a label-free and noninvasive strategy to detect the progression of fibrosis and the potency of the antifibrotic molecules in real-time. The fundamental principle that governs this novel system is that dynamic changes in cell shape and adhesion during fibrosis can be measured accurately by monitoring the changes in the impedance. This is achieved by growing the cells on a transparent interdigitated indium tin oxide (ITO) electrodes. It was demonstrated by monitoring the efficacy of a model antifibrotic compound, PXS64, on cells collected from patients with Dupuytren's contracture. We confirmed the validity of the developed biochemical impedance biosensor as an tool for in vitro screening of antifibrotic compounds and provided quantitative information on subcellular influences of the examined chemical molecules using correlative microscopy analyses that monitor the average cell area, cell morphology, and the amount and directionality of the deposited extracellular matrix protein collagen and measurement of cytosolic Ca2+ changes.
Collapse
Affiliation(s)
| | | | | | - Livia C. Hool
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
| | - P. Mark W. Fear
- Burns
Service of Western Australia, Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Fiona M. Wood
- Burns
Service of Western Australia, Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | | | | | | |
Collapse
|
33
|
Owen LM, Adhikari AS, Patel M, Grimmer P, Leijnse N, Kim MC, Notbohm J, Franck C, Dunn AR. A cytoskeletal clutch mediates cellular force transmission in a soft, three-dimensional extracellular matrix. Mol Biol Cell 2017; 28:1959-1974. [PMID: 28592635 PMCID: PMC5541846 DOI: 10.1091/mbc.e17-02-0102] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/26/2017] [Accepted: 05/31/2017] [Indexed: 01/08/2023] Open
Abstract
The ability of cells to impart forces and deformations on their surroundings underlies cell migration and extracellular matrix (ECM) remodeling and is thus an essential aspect of complex, metazoan life. Previous work has resulted in a refined understanding, commonly termed the molecular clutch model, of how cells adhering to flat surfaces such as a microscope coverslip transmit cytoskeletally generated forces to their surroundings. Comparatively less is known about how cells adhere to and exert forces in soft, three-dimensional (3D), and structurally heterogeneous ECM environments such as occur in vivo. We used time-lapse 3D imaging and quantitative image analysis to determine how the actin cytoskeleton is mechanically coupled to the surrounding matrix for primary dermal fibroblasts embedded in a 3D fibrin matrix. Under these circumstances, the cytoskeletal architecture is dominated by contractile actin bundles attached at their ends to large, stable, integrin-based adhesions. Time-lapse imaging reveals that α-actinin-1 puncta within actomyosin bundles move more quickly than the paxillin-rich adhesion plaques, which in turn move more quickly than the local matrix, an observation reminiscent of the molecular clutch model. However, closer examination did not reveal a continuous rearward flow of the actin cytoskeleton over slower moving adhesions. Instead, we found that a subset of stress fibers continuously elongated at their attachment points to integrin adhesions, providing stable, yet structurally dynamic coupling to the ECM. Analytical modeling and numerical simulation provide a plausible physical explanation for this result and support a picture in which cells respond to the effective stiffness of local matrix attachment points. The resulting dynamic equilibrium can explain how cells maintain stable, contractile connections to discrete points within ECM during cell migration, and provides a plausible means by which fibroblasts contract provisional matrices during wound healing.
Collapse
Affiliation(s)
- Leanna M Owen
- Biophysics, Stanford University, Stanford, CA 94305
- Chemical Engineering, Stanford University, Stanford, CA 94305
| | | | - Mohak Patel
- School of Engineering, Brown University, Providence, RI 02912
| | - Peter Grimmer
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI 53711
| | | | - Min Cheol Kim
- Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI 53711
| | | | | |
Collapse
|
34
|
Lee JS, Tae SS, Kim DY, Han SK, Kim WK, Dhong ES. Do IL-3/GM-CSF effect on the myofibroblastic differentiation of human adipose derived stromal cells? Exp Cell Res 2017; 355:67-82. [PMID: 28377320 DOI: 10.1016/j.yexcr.2017.03.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Capsular contracture is an incurable complication after silicone-based implant surgery. Myofibroblast is the predominant cell in the contracted capsule. We hypothesized that human adipose derive stromal cells (hASCs) together with fibroblast may show a similar phenotypic characteristics of myofibroblast after the treatment of inflammatory cytokines in vitro. MATERIALS AND METHODS Interleukin 3 (IL-3) and granulocyte macrophage colony stimulating factor (GM-CSF) were treated in the culture of hASCs and HDFs. Lyn peptide inhibitor was applied as an inhibitor. The changes of cell surface markers (CD105, CD73, CD34, CD45, CD31, CD325 and CD146) were assessed. The expression of various cytokines related to wound contraction were tested such as TGF-β, α-SMA, HGF, FGF, ENT-1, and TSP-1. Myo-D, α-SMA, and glial fibrillary acidic protein (GFAP) were evaluated by blotting and immunocytochemical staining. The collagen-gel contraction assay was performed for the functional contraction of myofibroblastic phenotype. RESULTS The expression of α-SMA, Myo-D and GFAP after the treatment of IL-3/GM-CSF showed similar results in hASCs and HDFs. Enhanced expression of TGF- β was observed in HDFs and the increase of ENT-1 and TSP-1 was significant in hASCs. Collagen-gel with HDFs contracted significantly within 24h after the treatment of IL-3/GM-CSF, and the contraction was inhibited by Lyn peptide inhibitor. But in hASCs, the gel-contraction was not significant. CONCLUSION IL-3/ GM-CSF effected on the myofibroblastic differentiation of hASCs as well as it did on HDFs. But hASCs did not show the phenotypic gel-contraction within 24h.
Collapse
Affiliation(s)
- Jae-Sun Lee
- Department of Plastic Surgery, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Son-Seung Tae
- Department of Plastic Surgery, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Deok-Yeol Kim
- Department of Plastic Surgery, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Seung-Kyu Han
- Department of Plastic Surgery, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Woo-Kyung Kim
- Department of Plastic Surgery, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Eun-Sang Dhong
- Department of Plastic Surgery, Korea University, Guro Hospital, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Menon SN, Hall CL, McCue SW, McElwain DLS. A model for one-dimensional morphoelasticity and its application to fibroblast-populated collagen lattices. Biomech Model Mechanobiol 2017; 16:1743-1763. [PMID: 28523375 DOI: 10.1007/s10237-017-0917-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 05/03/2017] [Indexed: 11/26/2022]
Abstract
The mechanical behaviour of solid biological tissues has long been described using models based on classical continuum mechanics. However, the classical continuum theories of elasticity and viscoelasticity cannot easily capture the continual remodelling and associated structural changes in biological tissues. Furthermore, models drawn from plasticity theory are difficult to apply and interpret in this context, where there is no equivalent of a yield stress or flow rule. In this work, we describe a novel one-dimensional mathematical model of tissue remodelling based on the multiplicative decomposition of the deformation gradient. We express the mechanical effects of remodelling as an evolution equation for the effective strain, a measure of the difference between the current state and a hypothetical mechanically relaxed state of the tissue. This morphoelastic model combines the simplicity and interpretability of classical viscoelastic models with the versatility of plasticity theory. A novel feature of our model is that while most models describe growth as a continuous quantity, here we begin with discrete cells and develop a continuum representation of lattice remodelling based on an appropriate limit of the behaviour of discrete cells. To demonstrate the utility of our approach, we use this framework to capture qualitative aspects of the continual remodelling observed in fibroblast-populated collagen lattices, in particular its contraction and its subsequent sudden re-expansion when remodelling is interrupted.
Collapse
Affiliation(s)
- Shakti N Menon
- The Institute of Mathematical Sciences, CIT Campus, Taramani, Chennai, 600113, India
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, 4001, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, 4001, Australia
| | - Cameron L Hall
- Mathematics Applications Consortium with Science and Industry, University of Limerick, Castletroy, Limerick, V94 T9PX, Ireland
- Oxford Centre for Industrial and Applied Mathematics, Mathematical Institute, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
| | - Scott W McCue
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, 4001, Australia.
| | - D L Sean McElwain
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, 4001, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, 4001, Australia
| |
Collapse
|
36
|
Myofibroblast repair mechanisms post-inflammatory response: a fibrotic perspective. Inflamm Res 2016; 66:451-465. [PMID: 28040859 DOI: 10.1007/s00011-016-1019-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/10/2016] [Accepted: 12/15/2016] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Fibrosis is a complex chronic disease characterized by a persistent repair response. Its pathogenesis is poorly understood but it is typically the result of chronic inflammation and maintained with the required activity of transforming growth factor-β (TGFβ) and extracellular matrix (ECM) tension, both of which drive fibroblasts to transition into a myofibroblast phenotype. FINDINGS As the effector cells of repair, myofibroblasts migrate to the site of injury to deposit excessive amounts of matrix proteins and stimulate high levels of contraction. Myofibroblast activity is a decisive factor in whether a tissue is properly repaired by controlled wound healing or rendered fibrotic by deregulated repair. Extensive studies have documented the various contributing factors to an abrogated repair response. Though these fibrotic factors are known, very little is understood about the opposing antifibrotic molecules that assist in a successful repair, such as prostaglandin E2 (PGE2) and ECM retraction. The following review will discuss the general development of fibrosis through the transformation of myofibroblasts, focusing primarily on the prominent profibrotic pathways of TGFβ and ECM tension and antifibrotic pathways of PGE2 and ECM retraction. CONCLUSIONS The idea is to understand the ways in which the cell, after an injury and inflammatory response, normally controls its repair mechanisms through its homeostatic regulators so as to mimic them therapeutically to control abnormal pathways.
Collapse
|
37
|
Mueller S. Does pressure cause liver cirrhosis? The sinusoidal pressure hypothesis. World J Gastroenterol 2016; 22:10482-10501. [PMID: 28082801 PMCID: PMC5192260 DOI: 10.3748/wjg.v22.i48.10482] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/10/2016] [Accepted: 11/28/2016] [Indexed: 02/06/2023] Open
Abstract
Independent of their etiology, all chronic liver diseases ultimately lead to liver cirrhosis, which is a major health problem worldwide. The underlying molecular mechanisms are still poorly understood and no efficient treatment strategies are available. This paper introduces the sinusoidal pressure hypothesis (SPH), which identifies an elevated sinusoidal pressure (SP) as cause of fibrosis. SPH has been mainly derived from recent studies on liver stiffness. So far, pressure changes have been exclusively seen as a consequence of cirrhosis. According to the SPH, however, an elevated SP is the major upstream event that initiates fibrosis via biomechanic signaling by stretching of perisinusoidal cells such as hepatic stellate cells or fibroblasts (SPH part I: initiation). Fibrosis progression is determined by the degree and time of elevated SP. The SPH predicts that the degree of extracellular matrix eventually matches SP with critical thresholds > 12 mmHg and > 4 wk. Elevated arterial flow and final arterialization of the cirrhotic liver represents the self-perpetuating key event exposing the low-pressure-organ to pathologically high pressures (SPH part II: perpetuation). It also defines the “point of no return” where fibrosis progression becomes irreversible. The SPH is able to explain the macroscopic changes of cirrhotic livers and the uniform fibrotic response to various etiologies. It also opens up new views on the role of fat and disease mechanisms in other organs. The novel concept will hopefully stimulate the search for new treatment strategies.
Collapse
|
38
|
Hogrebe NJ, Reinhardt JW, Gooch KJ. Biomaterial microarchitecture: a potent regulator of individual cell behavior and multicellular organization. J Biomed Mater Res A 2016; 105:640-661. [DOI: 10.1002/jbm.a.35914] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 08/17/2016] [Accepted: 09/02/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Nathaniel J. Hogrebe
- Department of Biomedical EngineeringThe Ohio State University270 Bevis Hall 1080 Carmack RdColumbus Ohio43210
| | - James W. Reinhardt
- Department of Biomedical EngineeringThe Ohio State University270 Bevis Hall 1080 Carmack RdColumbus Ohio43210
| | - Keith J. Gooch
- Department of Biomedical EngineeringThe Ohio State University270 Bevis Hall 1080 Carmack RdColumbus Ohio43210
- The Ohio State University, Davis Heart Lung Research Institute473 W 12th AveColumbus Ohio43210
| |
Collapse
|
39
|
Nilsen TJ, Dasgupta A, Huang YC, Wilson H, Chnari E. Do Processing Methods Make a Difference in Acellular Dermal Matrix Properties? Aesthet Surg J 2016; 36:S7-S22. [PMID: 27697888 DOI: 10.1093/asj/sjw163] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The use of acellular dermal matrices (ADMs) has become the standard of practice in many reconstructive and aesthetic surgical applications. Different methods used to prepare the allograft tissue for surgical use can alter the ADMs natural properties. Aseptic processing has been shown to retain the natural properties of ADMs more favorably than terminally sterilized ADMs. Terminal sterilization has been historically linked to alteration of biological materials. In vitro work was conducted to compare ADM processing methods. OBJECTIVES Characterize aseptically processed ADMs and compare cell-matrix interaction characteristics to terminally sterilized ADMs. METHODS Two aseptically processed ADMs, FlexHD Pliable and BellaDerm, were characterized via histological evaluation, biomechanical integrity, enzymatic degradation, and in vitro cell studies. FlexHD Pliable was compared to Alloderm Ready-to-Use (RTU). RESULTS Histological evaluation revealed that FlexHD Pliable had a uniform, open structure compared to BellaDerm. Mechanical characterization demonstrated that BellaDerm had higher strength and stiffness compared to FlexHD Pliable, which maintained higher elasticity. Immunohistochemical analysis verified that key matrix proteins remained intact after aseptic processing. Cell studies found that fibroblasts attached more readily, and proliferated faster on FlexHD Pliable compared to BellaDerm. Additionally, fibroblasts infiltrated into FlexHD Pliable from both sides and on the dermal side in BellaDerm and produced an abundance of multi-layered matrix proteins (collagen, fibronectin) when compared to AlloDerm RTU which was sparse. CONCLUSIONS Aseptically processed FlexHD Pliable and BellaDerm provide a suitable, biocompatible option for tissue repair and regeneration in aesthetic and reconstructive surgical applications.
Collapse
Affiliation(s)
- Todd J Nilsen
- Mr Nilsen is a Senior Engineer, Dr Dasgupta is a Senior Scientist, and Dr Chnari is an Associate Director, Research and Development, Wound Care, and General and Plastic Surgery; and Dr Huang is a Staff Scientist, Research and Development and Allograft Materials Research, Musculoskeletal Transplant Foundation, Edison, NJ. Dr Wilson is a Clinical Assistant Professor of Plastic Surgery, Liberty University College of Osteopathic Medicine, Lynchburg, VA
| | - Anouska Dasgupta
- Mr Nilsen is a Senior Engineer, Dr Dasgupta is a Senior Scientist, and Dr Chnari is an Associate Director, Research and Development, Wound Care, and General and Plastic Surgery; and Dr Huang is a Staff Scientist, Research and Development and Allograft Materials Research, Musculoskeletal Transplant Foundation, Edison, NJ. Dr Wilson is a Clinical Assistant Professor of Plastic Surgery, Liberty University College of Osteopathic Medicine, Lynchburg, VA
| | - Yen-Chen Huang
- Mr Nilsen is a Senior Engineer, Dr Dasgupta is a Senior Scientist, and Dr Chnari is an Associate Director, Research and Development, Wound Care, and General and Plastic Surgery; and Dr Huang is a Staff Scientist, Research and Development and Allograft Materials Research, Musculoskeletal Transplant Foundation, Edison, NJ. Dr Wilson is a Clinical Assistant Professor of Plastic Surgery, Liberty University College of Osteopathic Medicine, Lynchburg, VA
| | - Henry Wilson
- Mr Nilsen is a Senior Engineer, Dr Dasgupta is a Senior Scientist, and Dr Chnari is an Associate Director, Research and Development, Wound Care, and General and Plastic Surgery; and Dr Huang is a Staff Scientist, Research and Development and Allograft Materials Research, Musculoskeletal Transplant Foundation, Edison, NJ. Dr Wilson is a Clinical Assistant Professor of Plastic Surgery, Liberty University College of Osteopathic Medicine, Lynchburg, VA
| | - Evangelia Chnari
- Mr Nilsen is a Senior Engineer, Dr Dasgupta is a Senior Scientist, and Dr Chnari is an Associate Director, Research and Development, Wound Care, and General and Plastic Surgery; and Dr Huang is a Staff Scientist, Research and Development and Allograft Materials Research, Musculoskeletal Transplant Foundation, Edison, NJ. Dr Wilson is a Clinical Assistant Professor of Plastic Surgery, Liberty University College of Osteopathic Medicine, Lynchburg, VA
| |
Collapse
|
40
|
Jimenez Valencia AM, Wu PH, Yogurtcu ON, Rao P, DiGiacomo J, Godet I, He L, Lee MH, Gilkes D, Sun SX, Wirtz D. Collective cancer cell invasion induced by coordinated contractile stresses. Oncotarget 2016; 6:43438-51. [PMID: 26528856 PMCID: PMC4791242 DOI: 10.18632/oncotarget.5874] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/20/2015] [Indexed: 12/17/2022] Open
Abstract
The physical underpinnings of fibrosarcoma cell dissemination from a tumor in a surrounding collagen-rich matrix are poorly understood. Here we show that a tumor spheroid embedded in a 3D collagen matrix exerts large contractile forces on the matrix before invasion. Cell invasion is accompanied by complex spatially and temporally dependent patterns of cell migration within and at the surface of the spheroids that are fundamentally different from migratory patterns of individual fibrosarcoma cells homogeneously distributed in the same type of matrix. Cells display a continuous transition from a round morphology at the spheroid core, to highly aligned elongated morphology at the spheroid periphery, which depends on both β1-integrin-based cell-matrix adhesion and myosin II/ROCK-based cell contractility. This isotropic-to-anisotropic transition corresponds to a shift in migration, from a slow and unpolarized movement at the core, to a fast, polarized and persistent one at the periphery. Our results also show that the ensuing collective invasion of fibrosarcoma cells is induced by anisotropic contractile stresses exerted on the surrounding matrix.
Collapse
Affiliation(s)
- Angela M Jimenez Valencia
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Osman N Yogurtcu
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Pranay Rao
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Josh DiGiacomo
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Inês Godet
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Lijuan He
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Meng-Horng Lee
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Daniele Gilkes
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Sean X Sun
- Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland, 21218, USA.,Department of Oncology and Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland, 21218, USA
| |
Collapse
|
41
|
Zhao M, Altankov G, Grabiec U, Bennett M, Salmeron-Sanchez M, Dehghani F, Groth T. Molecular composition of GAG-collagen I multilayers affects remodeling of terminal layers and osteogenic differentiation of adipose-derived stem cells. Acta Biomater 2016; 41:86-99. [PMID: 27188244 DOI: 10.1016/j.actbio.2016.05.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/03/2016] [Accepted: 05/13/2016] [Indexed: 12/15/2022]
Abstract
UNLABELLED The effect of molecular composition of multilayers, by pairing type I collagen (Col I) with either hyaluronic acid (HA) or chondroitin sulfate (CS) was studied regarding the osteogenic differentiation of adhering human adipose-derived stem cells (hADSCs). Polyelectrolyte multilayer (PEM) formation was based primarily on ion pairing and on additional intrinsic cross-linking through imine bond formation with Col I replacing native by oxidized HA (oHA) or CS (oCS). Significant amounts of Col I fibrils were found on both native and oxidized CS-based PEMs, resulting in higher water contact angles and surface potential under physiological condition, while much less organized Col I was detected in either HA-based multilayers, which were more hydrophilic and negatively charged. An important finding was that hADSCs remodeled Col I at the terminal layers of PEMs by mechanical reorganization and pericellular proteolytic degradation, being more pronounced on CS-based PEMs. This was in accordance with the higher quantity of Col I deposition in this system, accompanied by more cell spreading, focal adhesions (FA) formation and significant α2β1 integrin recruitment compared to HA-based PEMs. Both CS-based PEMs caused also an increased fibronectin (FN) secretion and cell growth. Furthermore, significant calcium phosphate deposition, enhanced ALP, Col I and Runx2 expression were observed in hADSCs on CS-based PEMs, particularly on oCS-containing one. Overall, multilayer composition can be used to direct cell-matrix interactions, and hence stem cell fates showing for the first time that PEMs made of biogenic polyelectrolytes undergo significant remodeling of terminal protein layers, which seems to enable cells to form a more adequate extracellular matrix-like environment. STATEMENT OF SIGNIFICANCE Natural polymer derived polyelectrolyte multilayers (PEMs) have been recently applied to adjust biomaterials to meet specific tissue demands. However, the effect of molecular composition of multilayers on both surface properties and cellular response, especially the fate of human adipose derived stem cells (hADSCs) upon osteogenic differentiation has not been studied extensively, yet. In addition, no studies exist that investigate a potential cell-dependent remodeling of PEMs made of extracellular matrix (ECM) components like collagens and glycosaminoglycans (GAGs). Furthermore, there is no knowledge whether the ability of cells to remodel PEM components may provide an added value regarding cell growth and differentiation. Finally, it has not been explored yet, how intrinsic cross-linking of ECM derived polyelectrolytes that improve the stability of PEMs will affect the differentiation potential of hADSCs. The current work aims to address these questions and found that the type of GAG has a strong effect on properties of multilayers and osteogenic differentiation of hADSCs. Additionally, we also show for the first time that PEMs made of biogenic polyelectrolytes undergo significant remodeling of terminal layers as completely new finding, which allows cells to form an ECM-like environment supporting differentiation upon osteogenic lineage. The finding of this work may open new avenues of application of PEM systems made by layer by layer (LbL) technique in tissue engineering and regenerative medicine.
Collapse
|
42
|
Magnesium Modifies the Structural Features of Enzymatically Mineralized Collagen Gels Affecting the Retraction Capabilities of Human Dermal Fibroblasts Embedded within This 3D System. MATERIALS 2016; 9:ma9060477. [PMID: 28773595 PMCID: PMC5456744 DOI: 10.3390/ma9060477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022]
Abstract
Mineralized collagen gels have been developed as in vitro models to better understand the mechanisms regulating the calcification process and the behavior of a variety of cell types. The vast majority of data are related to stem cells and to osteoblast-like cells, whereas little information is available for dermal fibroblasts, although these cells have been associated with ectopic calcification and consequently to a number of pathological conditions. Therefore, we developed and characterized an enzymatically mineralized collagen gel in which fibroblasts were encapsulated within the 3D structure. MgCl2 was also added during gel polymerization, given its role as (i) modulator of ectopic calcification; (ii) component of biomaterials used for bone replacement; and (iii) constituent of pathological mineral deposits. Results demonstrate that, in a short time, an enzymatically mineralized collagen gel can be prepared in which mineral deposits and viable cells are homogeneously distributed. MgCl2 is present in mineral deposits and significantly affects collagen fibril assembly and organization. Consequently, cell shape and the ability of fibroblasts to retract collagen gels were modified. The development of three-dimensional (3D) mineralized collagen matrices with both different structural features and mineral composition together with the use of fibroblasts, as a prototype of soft connective tissue mesenchymal cells, may pave new ways for the study of ectopic calcification.
Collapse
|
43
|
Zhou W, Stukel JM, Cebull HL, Willits RK. Tuning the Mechanical Properties of Poly(Ethylene Glycol) Microgel-Based Scaffolds to Increase 3D Schwann Cell Proliferation. Macromol Biosci 2016; 16:535-44. [PMID: 26726886 DOI: 10.1002/mabi.201500336] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/30/2015] [Indexed: 12/14/2022]
Abstract
2D in vitro studies have demonstrated that Schwann cells prefer scaffolds with mechanical modulus approximately 10× higher than the modulus preferred by nerves, limiting the ability of many scaffolds to promote both neuron extension and Schwann cell proliferation. Therefore, the goals of this work are to develop and characterize microgel-based scaffolds that are tuned over the stiffness range relevant to neural tissue engineering and investigate Schwann cell morphology, viability, and proliferation within 3D scaffolds. Using thiol-ene reaction, microgels with surface thiols are produced and crosslinked into hydrogels using a multiarm vinylsulfone (VS). By varying the concentration of VS, scaffold stiffness ranges from 0.13 to 0.76 kPa. Cell morphology in all groups demonstrates that cells are able to spread and interact with the scaffold through day 5. Although the viability in all groups is high, proliferation of Schwann cells within the scaffold of G* = 0.53 kPa is significantly higher than other groups. This result is ≈ 5× lower than previously reported optimal stiffnesses on 2D surfaces, demonstrating the need for correlation of 3D cell response to mechanical modulus. As proliferation is the first step in Schwann cell integration into peripheral nerve conduits, these scaffolds demonstrate that the stiffness is a critical parameter to optimizing the regenerative process.
Collapse
Affiliation(s)
- Wenda Zhou
- Biomedical Engineering, The University of Akron, Akron, OH, 44325-0302, USA
| | - Jessica M Stukel
- Biomedical Engineering, The University of Akron, Akron, OH, 44325-0302, USA
| | - Hannah L Cebull
- Biomedical Engineering, The University of Akron, Akron, OH, 44325-0302, USA
| | | |
Collapse
|
44
|
Fernández J, Larrañaga A, Etxeberria A, Sarasua JR. Ethylene brassylate-co-δ-hexalactone biobased polymers for application in the medical field: synthesis, characterization and cell culture studies. RSC Adv 2016. [DOI: 10.1039/c6ra01065b] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Biodegradable polyesters based on ethylene brassylate and δ-hexalactone were synthesized by ring-opening polymerization and thoroughly characterized.
Collapse
Affiliation(s)
- Jorge Fernández
- Department of Mining-Metallurgy Engineering and Materials Science
- POLYMAT
- University of the Basque Country (UPV/EHU)
- School of Engineering
- 48013 Bilbao
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science
- POLYMAT
- University of the Basque Country (UPV/EHU)
- School of Engineering
- 48013 Bilbao
| | - Agustin Etxeberria
- Department of Polymer Science and Technology
- POLYMAT
- University of the Basque Country (UPV/EHU)
- 20018 Donostia-San Sebastian
- Spain
| | - Jose-Ramon Sarasua
- Department of Mining-Metallurgy Engineering and Materials Science
- POLYMAT
- University of the Basque Country (UPV/EHU)
- School of Engineering
- 48013 Bilbao
| |
Collapse
|
45
|
Agarwal V, Toshniwal P, Smith NE, Smith NM, Li B, Clemons TD, Byrne LT, Kakulas F, Wood FM, Fear M, Corry B, Swaminathan Iyer K. Enhancing the efficacy of cation-independent mannose 6-phosphate receptor inhibitors by intracellular delivery. Chem Commun (Camb) 2016; 52:327-30. [DOI: 10.1039/c5cc06826f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Intracellular delivery of M6P/IGFII receptor inhibitors exhibits better efficacy than extracellular inhibitors to regulate TGFβ1 mediated upregulation of profibrotic marker, collagen I.
Collapse
|
46
|
Changede R, Xu X, Margadant F, Sheetz MP. Nascent Integrin Adhesions Form on All Matrix Rigidities after Integrin Activation. Dev Cell 2015; 35:614-621. [PMID: 26625956 DOI: 10.1016/j.devcel.2015.11.001] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 10/26/2015] [Accepted: 11/02/2015] [Indexed: 10/24/2022]
Abstract
Integrin adhesions assemble and mature in response to ligand binding and mechanical factors, but the molecular-level organization is not known. We report that ∼100-nm clusters of ∼50 β3-activated integrins form very early adhesions under a wide variety of conditions on RGD surfaces. These adhesions form similarly on fluid and rigid substrates, but most adhesions are transient on rigid substrates. Without talin or actin polymerization, few early adhesions form, but expression of either the talin head or rod domain in talin-depleted cells restores early adhesion formation. Mutation of the integrin binding site in the talin rod decreases cluster size. We suggest that the integrin clusters constitute universal early adhesions and that they are the modular units of cell matrix adhesions. They require the association of activated integrins with cytoplasmic proteins, in particular talin and actin, and cytoskeletal contraction on them causes adhesion maturation for cell motility and growth.
Collapse
Affiliation(s)
- Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Xiaochun Xu
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Felix Margadant
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411; Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
47
|
Pinto V, Mohammadi H, Lee W, Cheung A, McCulloch C. PAK1 is involved in sensing the orientation of collagen stiffness gradients in mouse fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2526-38. [DOI: 10.1016/j.bbamcr.2015.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 05/01/2015] [Accepted: 05/19/2015] [Indexed: 01/13/2023]
|
48
|
Arora PD, Wang Y, Bresnick A, Janmey PA, McCulloch CA. Flightless I interacts with NMMIIA to promote cell extension formation, which enables collagen remodeling. Mol Biol Cell 2015; 26:2279-97. [PMID: 25877872 PMCID: PMC4462945 DOI: 10.1091/mbc.e14-11-1536] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 04/09/2015] [Indexed: 01/14/2023] Open
Abstract
The role of the actin-capping protein flightless I in collagen remodeling by mouse fibroblasts is examined. Flightless and nonmuscle myosin IIA cooperate to enable collagen phagocytosis. We examined the role of the actin-capping protein flightless I (FliI) in collagen remodeling by mouse fibroblasts. FliI-overexpressing cells exhibited reduced spreading on collagen but formed elongated protrusions that stained for myosin10 and fascin and penetrated pores of collagen-coated membranes. Inhibition of Cdc42 blocked formation of cell protrusions. In FliI-knockdown cells, transfection with constitutively active Cdc42 did not enable protrusion formation. FliI-overexpressing cells displayed increased uptake and degradation of exogenous collagen and strongly compacted collagen fibrils, which was blocked by blebbistatin. Mass spectrometry analysis of FliI immunoprecipitates showed that FliI associated with nonmuscle myosin IIA (NMMIIA), which was confirmed by immunoprecipitation. GFP-FliI colocalized with NMMIIA at cell protrusions. Purified FliI containing gelsolin-like domains (GLDs) 1–6 capped actin filaments efficiently, whereas FliI GLD 2–6 did not. Binding assays showed strong interaction of purified FliI protein (GLD 1–6) with the rod domain of NMMIIA (kD = 0.146 μM), whereas FliI GLD 2–6 showed lower binding affinity (kD = 0.8584 μM). Cells expressing FliI GLD 2–6 exhibited fewer cell extensions, did not colocalize with NMMIIA, and showed reduced collagen uptake compared with cells expressing FliI GLD 1–6. We conclude that FliI interacts with NMMIIA to promote cell extension formation, which enables collagen remodeling in fibroblasts.
Collapse
Affiliation(s)
- Pamma D Arora
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Yongqiang Wang
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Anne Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461
| | - Paul A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Christopher A McCulloch
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 3E2, Canada
| |
Collapse
|
49
|
Broussard JA, Diggins NL, Hummel S, Georgescu W, Quaranta V, Webb DJ. Automated analysis of cell-matrix adhesions in 2D and 3D environments. Sci Rep 2015; 5:8124. [PMID: 25630460 PMCID: PMC4309964 DOI: 10.1038/srep08124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/06/2015] [Indexed: 01/01/2023] Open
Abstract
Cell-matrix adhesions are of great interest because of their contribution to numerous biological processes, including cell migration, differentiation, proliferation, survival, tissue morphogenesis, wound healing, and tumorigenesis. Adhesions are dynamic structures that are classically defined on two-dimensional (2D) substrates, though the need to analyze adhesions in more physiologic three-dimensional (3D) environments is being increasingly recognized. However, progress has been greatly hampered by the lack of available tools to analyze adhesions in 3D environments. To address this need, we have developed a platform for the automated analysis, segmentation, and tracking of adhesions (PAASTA) based on an open source MATLAB framework, CellAnimation. PAASTA enables the rapid analysis of adhesion dynamics and many other adhesion characteristics, such as lifetime, size, and location, in 3D environments and on traditional 2D substrates. We manually validate PAASTA and utilize it to quantify rate constants for adhesion assembly and disassembly as well as adhesion lifetime and size in 3D matrices. PAASTA will be a valuable tool for characterizing adhesions and for deciphering the molecular mechanisms that regulate adhesion dynamics in 3D environments.
Collapse
Affiliation(s)
- Joshua A Broussard
- Department of Biological Sciences and Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235
| | - Nicole L Diggins
- Department of Biological Sciences and Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235
| | - Stephen Hummel
- Center for Cancer Systems Biology at Vanderbilt, Vanderbilt University, Nashville, Tennessee 37235
| | - Walter Georgescu
- 1] Center for Cancer Systems Biology at Vanderbilt, Vanderbilt University, Nashville, Tennessee 37235 [2] Vanderbilt Institute for Integrative Biosystems Research and Education (VIBRE), Vanderbilt University, Nashville, Tennessee 37235 [3] Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235
| | - Vito Quaranta
- 1] Center for Cancer Systems Biology at Vanderbilt, Vanderbilt University, Nashville, Tennessee 37235 [2] Vanderbilt Institute for Integrative Biosystems Research and Education (VIBRE), Vanderbilt University, Nashville, Tennessee 37235 [3] Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee 37235
| | - Donna J Webb
- 1] Department of Biological Sciences and Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235 [2] Vanderbilt Institute for Integrative Biosystems Research and Education (VIBRE), Vanderbilt University, Nashville, Tennessee 37235 [3] Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee 37235
| |
Collapse
|
50
|
Park S, Seawright A, Park S, Craig Dutton J, Grinnell F, Han B. Preservation of tissue microstructure and functionality during freezing by modulation of cytoskeletal structure. J Mech Behav Biomed Mater 2015; 45:32-44. [PMID: 25679482 DOI: 10.1016/j.jmbbm.2015.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 01/13/2015] [Accepted: 01/16/2015] [Indexed: 02/06/2023]
Abstract
Cryopreservation is one of the key enabling technologies for tissue engineering and regenerative medicine, which can provide reliable long-term storage of engineered tissues (ETs) without losing their functionality. However, it is still extremely difficult to design and develop cryopreservation protocols guaranteeing the post-thaw tissue functionality. One of the major challenges in cryopreservation is associated with the difficulty of identifying effective and less toxic cryoprotective agents (CPAs) to guarantee the post-thaw tissue functionality. In this study, thus, a hypothesis was tested that the modulation of the cytoskeletal structure of cells embedded in the extracellular matrix (ECM) can mitigate the freezing-induced changes of the functionality and can reduce the amount of CPA necessary to preserve the functionality of ETs during cryopreservation. In order to test this hypothesis, we prepared dermal equivalents by seeding fibroblasts in type I collagen matrices resulting in three different cytoskeletal structures. These ETs were exposed to various freeze/thaw (F/T) conditions with and without CPAs. The freezing-induced cell-fluid-matrix interactions and subsequent functional properties of the ETs were assessed. The results showed that the cytoskeletal structure and the use of CPA were strongly correlated to the preservation of the post-thaw functional properties. As the cytoskeletal structure became stronger via stress fiber formation, the ET's functionality was preserved better. It also reduced the necessary CPA concentration to preserve the post-thaw functionality. However, if the extent of the freezing-induced cell-fluid-matrix interaction was too excessive, the cytoskeletal structure was completely destroyed and the beneficial effects became minimal.
Collapse
Affiliation(s)
- Seungman Park
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Angela Seawright
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Sinwook Park
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - J Craig Dutton
- Department of Aerospace Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Frederick Grinnell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|