1
|
Landau S, Kieda J, Khosravi R, Okhovatian S, Ramsay K, Liu C, Shakeri A, Zhao Y, Shen K, Bar-Am O, Levenberg S, Tsai S, Radisic M. Cell driven elastomeric particle packing in composite bioinks for engineering and implantation of stable 3D printed structures. Bioact Mater 2025; 44:411-427. [PMID: 39525804 PMCID: PMC11550138 DOI: 10.1016/j.bioactmat.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Geometric and structural integrity often deteriorate in 3D printed cell-laden constructs over time due to cellular compaction and hydrogel shrinkage. This study introduces a new approach that synergizes the advantages of cell compatibility of biological hydrogels and mechanical stability of elastomeric polymers for structure fidelity maintenance upon stereolithography and extrusion 3D printing. Enabling this advance is the composite bioink, formulated by integrating elastomeric microparticles from poly(octamethylene maleate (anhydride) citrate) (POMaC) into biologically derived hydrogels (fibrin, gelatin methacryloyl (GelMA), and alginate). The composite bioink enhanced the elasticity and plasticity of the 3D printed constructs, effectively mitigating tissue compaction and swelling. It exhibited a low shear modulus and a rapid crosslinking time, along with a high ultimate compressive strength and resistance to deformation from cellular forces and physical handling; this was attributed to packing and stress dissipation of elastomeric particles, which was confirmed via mathematical modelling. Enhanced functional assembly and stability of human iPSC-derived cardiac tissues and primary vasculature proved the utility of the composite bioink in tissue engineering. In vivo implantation studies revealed that constructs containing POMaC particles exhibited improved resilience against host tissue stress, enhanced angiogenesis, and infiltration of pro-reparative macrophages.
Collapse
Affiliation(s)
- Shira Landau
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Jennifer Kieda
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Ramak Khosravi
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Sargol Okhovatian
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Kaitlyn Ramsay
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Chuan Liu
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Amid Shakeri
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Yimu Zhao
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
- Acceleration Consortium, University of Toronto, Toronto, ON, Canada
| | - Karen Shen
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Orit Bar-Am
- Faculty of Biomedical Engineering, Technion, Haifa, IL, Israel
| | | | - Scott Tsai
- Toronto Metropolitan University, Department of Mechanical, Industrial, and Mechatronics Engineering, Toronto, ON, Canada
- Toronto Metropolitan University and Unity Health Toronto, Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, ON, Canada
| | - Milica Radisic
- University of Toronto, Institute of Biomedical Engineering, Toronto, ON, Canada
- University Health Network, Toronto General Hospital Research Institute, Toronto, ON, Canada
- University of Toronto, Terrence Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
- University of Toronto, Department of Chemical Engineering and Applied Chemistry, Toronto, ON, Canada
- Acceleration Consortium, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Viti F, Pramotton FM, Martufi M, Magrassi R, Pedemonte N, Nizzari M, Zanacchi FC, De Michele B, Alampi M, Zambito M, Santamaria G, Bajetto A, Sardar S, Tomati V, Gandullia P, Giampietro C, Florio T, Beltrame F, Vassalli M, Ceccherini I. Patient's dermal fibroblasts as disease markers for visceral myopathy. BIOMATERIALS ADVANCES 2023; 148:213355. [PMID: 36893487 DOI: 10.1016/j.bioadv.2023.213355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/09/2023] [Accepted: 02/19/2023] [Indexed: 06/18/2023]
Abstract
Visceral myopathy (VSCM) is a rare genetic disease, orphan of pharmacological therapy. VSCM diagnosis is not always straightforward due to symptomatology similarities with mitochondrial or neuronal forms of intestinal pseudo-obstruction. The most prevalent form of VSCM is associates with variants in the gene ACTG2, encoding the protein gamma-2 actin. Overall, VSCM is a mechano-biological disorder, in which different genetic variants lead to similar alterations to the contractile phenotype of enteric smooth muscles, resulting in the emergence of life-threatening symptoms. In this work we analyzed the morpho-mechanical phenotype of human dermal fibroblasts from patients affected with VSCM, demonstrating that they retain a clear signature of the disease when compared with different controls. We evaluated several biophysical traits of fibroblasts, and we show that a measure of cellular traction forces can be used as a non-specific biomarker of the disease. We propose that a simple assay based on traction forces could be designed to provide a valuable support for clinical decision or pre-clinical research.
Collapse
Affiliation(s)
- Federica Viti
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy.
| | - Francesca Micaela Pramotton
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, Ueberlandstrasse 129, 8600 Dübendorf, Switzerland; ETH Zurich, The Institute for Mechanical Systems, Leonhardstrasse 21, 8092 Zürich, Switzerland
| | - Michela Martufi
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy; Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy
| | - Raffaella Magrassi
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy
| | - Nicoletta Pedemonte
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Mario Nizzari
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy
| | | | - Benedetta De Michele
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy
| | - Manuela Alampi
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Viale Causa, 13, 16145 Genova, Italy
| | - Martina Zambito
- Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy
| | - Giuseppe Santamaria
- UOSD Laboratorio di Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Adriana Bajetto
- Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy
| | - Sabah Sardar
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, Oakfield avenue, G128LT Glasgow, UK
| | - Valeria Tomati
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Paolo Gandullia
- UOC Pediatric Gastroenterology and Digestive Endoscopy, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Costanza Giampietro
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, Ueberlandstrasse 129, 8600 Dübendorf, Switzerland; ETH Zurich, The Institute for Mechanical Systems, Leonhardstrasse 21, 8092 Zürich, Switzerland
| | - Tullio Florio
- Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy; IRCCS Ospedale Policlinico San Martino, Largo rosanna benzi 10, 16132 Genova, Italy
| | - Francesco Beltrame
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Viale Causa, 13, 16145 Genova, Italy
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, Oakfield avenue, G128LT Glasgow, UK
| | - Isabella Ceccherini
- UOSD Laboratorio di Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| |
Collapse
|
3
|
Klumm MJ, Heim C, Fiegle DJ, Weyand M, Volk T, Seidel T. Long-Term Cultivation of Human Atrial Myocardium. Front Physiol 2022; 13:839139. [PMID: 35283779 PMCID: PMC8905341 DOI: 10.3389/fphys.2022.839139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Organotypic culture of human ventricular myocardium is emerging in basic and translational cardiac research. However, few institutions have access to human ventricular tissue, whereas atrial tissue is more commonly available and important for studying atrial physiology. This study presents a method for long-term cultivation of beating human atrial myocardium. After written informed consent, tissues from the right-atrial appendage were obtained from patients with sinus rhythm undergoing open heart surgery with cardiopulmonary bypass. Trabeculae (pectinate muscles) prepared from the samples were installed into cultivation chambers at 37°C with a diastolic preload of 500 μN. After 2 days with 0.5 Hz pacing, stimulation frequency was set to 1 Hz. Contractile force was monitored continuously. Beta-adrenergic response, refractory period (RP) and maximum captured frequency (fmax) were assessed periodically. After cultivation, viability and electromechanical function were investigated, as well as the expression of several genes important for intracellular Ca2+ cycling and electrophysiology. Tissue microstructure was analyzed by confocal microscopy. We cultivated 19 constantly beating trabeculae from 8 patient samples for 12 days and 4 trabeculae from 3 specimen for 21 days. Functional parameters were compared directly after installation (0 d) with those after 12 d in culture. Contraction force was 384 ± 69 μN at 0 d and 255 ± 90 μN at 12 d (p = 0.8, n = 22), RP 480 ± 97 ms and 408 ± 78 ms (p = 0.3, n = 9), fmax 3.0 ± 0.5 Hz and 3.8 ± 0.5 Hz (p = 0.18, n = 9), respectively. Application of 100 nM isoprenaline to 11 trabeculae at 7 d increased contraction force from 168 ± 35 μN to 361 ± 60 μN (p < 0.01), fmax from 6.4 ± 0.6 Hz to 8.5 ± 0.4 Hz (p < 0.01) and lowered RP from 319 ± 22 ms to 223 ± 15 ms. CACNA1c (L-type Ca2+ channel subunit) and GJA1 (connexin-43) mRNA expressions were not significantly altered at 12 d vs 0 d, while ATP2A (SERCA) and KCNJ4 (Kir2.3) were downregulated, and KCNJ2 (Kir2.1) was upregulated. Simultaneous Ca2+ imaging and force recording showed preserved excitation-contraction coupling in cultivated trabeculae. Confocal microscopy indicated preserved cardiomyocyte structure, unaltered amounts of extracellular matrix and gap junctions. MTT assays confirmed viability at 12 d. We established a workflow that allows for stable cultivation and functional analysis of beating human atrial myocardium for up to 3 weeks. This method may lead to novel insights into the physiology and pathophysiology of human atrial myocardium.
Collapse
Affiliation(s)
- Maximilian J Klumm
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dominik J Fiegle
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tilmann Volk
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Seidel
- Institute of Cellular and Molecular Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
Dittloff KT, Spanghero E, Solís C, Banach K, Russell B. Transthyretin deposition alters cardiomyocyte sarcomeric architecture, calcium transients, and contractile force. Physiol Rep 2022; 10:e15207. [PMID: 35262277 PMCID: PMC8906053 DOI: 10.14814/phy2.15207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023] Open
Abstract
Age-related wild-type transthyretin amyloidosis (wtATTR) is characterized by systemic deposition of amyloidogenic fibrils of misfolded transthyretin (TTR) in the connective tissue of many organs. In the heart, this leads to age-related heart failure with preserved ejection fraction (HFpEF). The hypothesis tested is that TTR deposited in vitro disrupts cardiac myocyte cell-to-cell and cell-to-matrix adhesion complexes, resulting in altered calcium handling, force generation, and sarcomeric disorganization. Human iPSC-derived cardiomyocytes and neonatal rat ventricular myocytes (NRVMs), when grown on TTR-coated polymeric substrata mimicking the stiffness of the healthy human myocardium (10 kPa), had decreased contraction and relaxation velocities as well as decreased force production measured using traction force microscopy. Both NRVMs and adult mouse atrial cardiomyocytes had altered calcium kinetics with prolonged transients when cultured on TTR fibril-coated substrates. Furthermore, NRVMs grown on stiff (~GPa), flat or microgrooved substrates coated with TTR fibrils exhibited significantly decreased intercellular electrical coupling as shown by FRAP dynamics of cells loaded with the gap junction-permeable dye calcein-AM, along with decreased gap junction content as determined by quantitative connexin 43 staining. Significant sarcomeric disorganization and loss of sarcomere content, with increased ubiquitin localization to the sarcomere, were seen in NRVMs on various TTR fibril-coated substrata. TTR presence decreased intercellular mechanical junctions as evidenced by quantitative immunofluorescence staining of N-cadherin and vinculin. Current therapies for wtATTR are cost-prohibitive and only slow the disease progression; therefore, better understanding of cardiomyocyte maladaptation induced by TTR amyloid may identify novel therapeutic targets.
Collapse
Affiliation(s)
- Kyle T. Dittloff
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Emanuele Spanghero
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Christopher Solís
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Kathrin Banach
- Department of Internal Medicine/CardiologyRush University Medical CenterChicagoIllinoisUSA
| | - Brenda Russell
- Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
- Department of Biomedical EngineeringUniversity of Illinois at ChicagoChicagoIllinoisUSA
| |
Collapse
|
5
|
Dittloff KT, Iezzi A, Zhong JX, Mohindra P, Desai TA, Russell B. Transthyretin amyloid fibrils alter primary fibroblast structure, function, and inflammatory gene expression. Am J Physiol Heart Circ Physiol 2021; 321:H149-H160. [PMID: 34018852 DOI: 10.1152/ajpheart.00073.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Age-related wild-type transthyretin amyloidosis (wtATTR) is characterized by systemic deposition of amyloidogenic fibrils of misfolded transthyretin (TTR) in the connective tissue of many organs. In the heart, this leads to cardiac dysfunction, which is a significant cause of age-related heart failure. The hypothesis tested is that TTR affects cardiac fibroblasts in ways that may contribute to fibrosis. When primary cardiac fibroblasts were cultured on TTR-deposited substrates, the F-actin cytoskeleton was disorganized, focal adhesion formation was decreased, and nuclear shape was flattened. Fibroblasts had faster collective and single-cell migration velocities on TTR-deposited substrates. In addition, fibroblasts cultured on microposts with TTR deposition had reduced attachment and increased proliferation above untreated. Transcriptomic and proteomic analyses of fibroblasts grown on glass covered with TTR showed significant upregulation of inflammatory genes after 48 h, indicative of progression in TTR-based diseases. Together, results suggest that TTR deposited in tissue extracellular matrix may affect the structure, function, and gene expression of cardiac fibroblasts. As therapies for wtATTR are cost-prohibitive and only slow disease progression, better understanding of cellular maladaptation may elucidate novel therapeutic targets.NEW & NOTEWORTHY Transthyretin (TTR) cardiac amyloidosis involves deposition of fibrils of misfolded TTR in the aging human heart, leading to cardiac dysfunction and heart failure. Our novel in vitro studies show that TTR fibrils alter primary cardiac fibroblast cytoskeletal and nuclear structure and focal adhesion formation. Furthermore, both fibrillar and tetrameric TTR significantly increased cellular migration velocity and caused upregulation of inflammatory genes determined by transcriptomic RNA and protein analysis. These findings may suggest new therapeutic approaches.
Collapse
Affiliation(s)
- Kyle T Dittloff
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Antonio Iezzi
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Justin X Zhong
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, California.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| | - Priya Mohindra
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, California.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| | - Tejal A Desai
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, California.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California.,Department of Bioengineering, University of California, Berkeley, California
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
6
|
Zhang Y, An J, Liu M, Li N, Wang W, Yao H, Li N, Yang X, Sun Y, Xu N, Wu L. Efficient isolation, culture, purification, and stem cell expression profiles of primary tumor cells derived from uterine cervical squamous cell carcinoma. Am J Reprod Immunol 2020; 84:e13251. [PMID: 32315465 DOI: 10.1111/aji.13251] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/10/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022] Open
Abstract
PROBLEM Since not too many human uterus cervical squamous cell carcinoma (CSCC) cell lines in existence, efficient isolation, culture, and purification protocols for primary CSCC cells were optimized as a tool for the study of uterus CSCC. METHOD OF STUDY The protocols for partial multiple enzymatic digestion and explant cell culture were combined and then the resulting mixed cell component cultures were purified by magnetic-activated cell sorting. Colony-forming assay was utilized for detection of cell carcinogenesis potential, and immunofluorescence was used to detect protein expression of CSCC. Finally, flow cytometry (FCM) was performed to analyze cancer stem cells (CSCs) phenotypic markers as well as programmed cell death ligand 1(PD-L 1). RESULTS Freshly isolated cells containing tumor cells and cancer-associated fibroblasts (CAFs) efficiently proliferate to 85% confluence on a 6 cm petri dish in 5-7 days. Anti-epithelial cell adhesion molecule antibody (EpCAM) microbeads were used to successfully separate a homogeneous subpopulation of epithelial tumor cells. Both EpCAM+ and EpCAM- cell subpopulations were able to be passaged more than 30 times. Proportions of tumor cell populations expressed CSCs markers such as CD133, CD24, aldehyde dehydrogenase 1 (ALDH1), and CD44. The vimentin+ & EpCAM- population, defined with CAFs, could express CD146 mesenchymal stem cells marker. Meanwhile, PD-L 1 was identified in most subpopulation of CD44+ cells at low passage numbers. CONCLUSION Efficient isolation, culture, and purification protocols for primary CSCC cells were successfully built. Additionally, the profiling of CSCs cell markers might provide promising therapeutic targets and clinic strategies.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jusheng An
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenpeng Wang
- Department of colorectal oncology, Tianjin Medical University Cancer Hospital, Tianjin, China
| | - Hongwen Yao
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Nan Li
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xi Yang
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yangchun Sun
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lingying Wu
- Department of Gynecologic Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Magdaleno F, Blajszczak CC, Charles-Niño CL, Guadrón-Llanos AM, Vázquez-Álvarez AO, Miranda-Díaz AG, Nieto N, Islas-Carbajal MC, Rincón-Sánchez AR. Aminoguanidine reduces diabetes-associated cardiac fibrosis. Exp Ther Med 2019; 18:3125-3138. [PMID: 31572553 DOI: 10.3892/etm.2019.7921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
Aminoguanidine (AG) inhibits advanced glycation end products (AGEs) and advanced oxidation protein products (AOPP) accumulated as a result of excessive oxidative stress in diabetes. However, the molecular mechanism by which AG reduces AGE-associated damage in diabetes is not well understood. Thus, we investigated whether AG supplementation mitigates oxidative-associated cardiac fibrosis in rats with type 2 diabetes mellitus (T2DM). Forty-five male Wistar rats were divided into three groups: Control, T2DM and T2DM+AG. Rats were fed with a high-fat, high-carbohydrate diet (HFCD) for 2 weeks and rendered diabetic using low-dose streptozotocin (STZ) (20 mg/kg), and one group was treated with AG (20 mg/kg) up to 25 weeks. In vitro experiments were performed in primary rat myofibroblasts to confirm the antioxidant and antifibrotic effects of AG and to determine if blocking the receptor for AGEs (RAGE) prevents the fibrogenic response in myofibroblasts. Diabetic rats exhibited an increase in cardiac fibrosis resulting from HFCD and STZ injections. By contrast, AG treatment significantly reduced cardiac fibrosis, α-smooth muscle actin (αSMA) and oxidative-associated Nox4 and Nos2 mRNA expression. In vitro challenge of myofibroblasts with AG under T2DM conditions reduced intra- and extracellular collagen type I expression and Pdgfb, Tgfβ1 and Col1a1 mRNAs, albeit with similar expression of Tnfα and Il6 mRNAs. This was accompanied by reduced phosphorylation of ERK1/2 and SMAD2/3 but not of AKT1/2/3 and STAT pathways. RAGE blockade further attenuated collagen type I expression in AG-treated myofibroblasts. Thus, AG reduces oxidative stress-associated cardiac fibrosis by reducing pERK1/2, pSMAD2/3 and collagen type I expression via AGE/RAGE signaling in T2DM.
Collapse
Affiliation(s)
- Fernando Magdaleno
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico.,Department of Pathology, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | | | - Claudia Lisette Charles-Niño
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico.,Department of Microbiology and Pathology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Alma Marlene Guadrón-Llanos
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Alan Omar Vázquez-Álvarez
- Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Alejandra Guillermina Miranda-Díaz
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Natalia Nieto
- Department of Pathology, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - María Cristina Islas-Carbajal
- Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| | - Ana Rosa Rincón-Sánchez
- Institute of Molecular Biology and Gene Therapy, Department of Molecular Biology and Genomics, University Center of Health Sciences, Guadalajara University, Guadalajara, Jalisco 44340, Mexico
| |
Collapse
|
8
|
Le LV, Mkrtschjan MA, Russell B, Desai TA. Hang on tight: reprogramming the cell with microstructural cues. Biomed Microdevices 2019; 21:43. [PMID: 30955102 PMCID: PMC6791714 DOI: 10.1007/s10544-019-0394-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cells interact intimately with complex microdomains in their extracellular matrix (ECM) and maintain a delicate balance of mechanical forces through mechanosensitive cellular components. Tissue injury results in acute degradation of the ECM and disruption of cell-ECM contacts, manifesting in loss of cytoskeletal tension, leading to pathological cell transformation and the onset of disease. Recently, microscale hydrogel constructs have been developed to provide cells with microdomains to form focal adhesion binding sites, which enable restoration of cytoskeletal tension. These synthetic anchors can recapitulate the complex 3D architecture of the native ECM to provide microtopographical cues. The mechanical deformation of proteins at the cell surface can activate signaling cascades to modulate downstream gene-level transcription, making this a unique materials-based approach for reprogramming cell behavior. An overview of the mechanisms underlying these mechanosensitive interactions in fibroblasts, stem and other cell types is provided to review their effects on cellular reprogramming. Recent investigations on the fabrication, functionalization and implementation of these materials and microtopographical features for drug testing and therapeutic applications are discussed.
Collapse
Affiliation(s)
- Long V Le
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th St Rm 204, San Francisco, CA, 94158, USA
| | - Michael A Mkrtschjan
- Department of Bioengineering, University of Illinois, Chicago, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois, Chicago, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th St Rm 204, San Francisco, CA, 94158, USA.
| |
Collapse
|
9
|
Mkrtschjan MA, Gaikwad SB, Kappenman KJ, Solís C, Dommaraju S, Le LV, Desai TA, Russell B. Lipid signaling affects primary fibroblast collective migration and anchorage in response to stiffness and microtopography. J Cell Physiol 2017; 233:3672-3683. [PMID: 29034471 DOI: 10.1002/jcp.26236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
Cell migration is regulated by several mechanotransduction pathways, which consist of sensing and converting mechanical microenvironmental cues to internal biochemical cellular signals, such as protein phosphorylation and lipid signaling. While there has been significant progress in understanding protein changes in the context of mechanotransduction, lipid signaling is more difficult to investigate. In this study, physical cues of stiffness (10, 100, 400 kPa, and glass), and microrod or micropost topography were manipulated in order to reprogram primary fibroblasts and assess the effects of lipid signaling on the actin cytoskeleton. In an in vitro wound closure assay, primary cardiac fibroblast migration velocity was significantly higher on soft polymeric substrata. Modulation of PIP2 availability through neomycin treatment nearly doubled migration velocity on 10 kPa substrata, with significant increases on all stiffnesses. The distance between focal adhesions and the lamellar membrane (using wortmannin treatment to increase PIP2 via PI3K inhibition) was significantly shortest compared to untreated fibroblasts grown on the same surface. PIP2 localized to the leading edge of migrating fibroblasts more prominently in neomycin-treated cells. The membrane-bound protein, lamellipodin, did not vary under any condition. Additionally, fifteen micron-high micropost topography, which blocks migration, concentrates PIP2 near to the post. Actin dynamics within stress fibers, measured by fluorescence recovery after photobleaching, was not significantly different with stiffness, microtopography, nor with drug treatment. PIP2-modulating drugs delivered from microrod structures also affected migration velocity. Thus, manipulation of the microenvironment and lipid signaling regulatory drugs might be beneficial in improving therapeutics geared toward wound healing.
Collapse
Affiliation(s)
- Michael A Mkrtschjan
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Snehal B Gaikwad
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Kevin J Kappenman
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Christopher Solís
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Sagar Dommaraju
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Long V Le
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California
| | - Brenda Russell
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois.,Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
10
|
Dynamics of spatiotemporal line defects and chaos control in complex excitable systems. Sci Rep 2017; 7:7757. [PMID: 28798384 PMCID: PMC5552747 DOI: 10.1038/s41598-017-08011-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/30/2017] [Indexed: 12/01/2022] Open
Abstract
Spatiotemporal pattern formation governs dynamics and functions in various biological systems. In the heart, excitable waves can form complex oscillatory and chaotic patterns even at an abnormally higher frequency than normal heart beats, which increase the risk of fatal heart conditions by inhibiting normal blood circulation. Previous studies suggested that line defects (nodal lines) play a critical role in stabilizing those undesirable patterns. However, it remains unknown if the line defects are static or dynamically changing structures in heart tissue. Through in vitro experiments of heart tissue observation, we reveal the spatiotemporal dynamics of line defects in rotating spiral waves. We combined a novel signaling over-sampling technique with a multi-dimensional Fourier analysis, showing that line defects can translate, merge, collapse and form stable singularities with even and odd parity while maintaining a stable oscillation of the spiral wave in the tissue. These findings provide insights into a broad class of complex periodic systems, with particular impact to the control and understanding of heart diseases.
Collapse
|
11
|
Cerium Chloride Application Promotes Wound Healing and Cell Proliferation in Human Foreskin Fibroblasts. MATERIALS 2017; 10:ma10060573. [PMID: 28772932 PMCID: PMC5552080 DOI: 10.3390/ma10060573] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/15/2017] [Accepted: 05/18/2017] [Indexed: 11/17/2022]
Abstract
This study investigated the effect of cerium chloride (CeCl₃) on cell migration and gene expression of human foreskin fibroblasts (HFF). HFF were exposed to three different CeCl₃ solutions (1%, 5% and 10%, w/v %) for three different time durations (1, 5 and 10 min). 72 h after exposure to CeCl₃, cell viability was assessed by MTT test. A scratch-wounded assay determined the cell migration and the width of the wound, measured at 24 h. Gene expression patterns for cyclins B1, D1 and E1 were analyzed by RT-PCR (p < 0.05, t-test). The viability proliferation increased at 1- and 5-min exposures for all CeCl₃ concentrations, in contrast to no treatment (p < 0.05 at 24 h). No influence of CeCl₃ was found after 10 min. The scratch assay showed increased cell migration up to 60% at 1 and 5 min after 24 h at 5% and 10%. Cyclin B1, D1 and E1 all showed upregulation, confirming an increase in cell proliferation. This study demonstrates that exposure time and concentration of CeCl₃ may have a positive effect on fibroblast viability and migration. Application of CeCl₃ may be beneficial as a cell-stimulating agent leading to therapeutic tissue fibrosis or more resistant tissue around teeth, when warranted, during different periodontal therapies.
Collapse
|
12
|
Li J, Mkrtschjan MA, Lin YH, Russell B. Variation in stiffness regulates cardiac myocyte hypertrophy via signaling pathways. Can J Physiol Pharmacol 2016; 94:1178-1186. [PMID: 27486838 DOI: 10.1139/cjpp-2015-0578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Much diseased human myocardial tissue is fibrotic and stiff, which increases the work that the ventricular myocytes must perform to maintain cardiac output. The hypothesis tested is that the increased load due to greater stiffness of the substrata drives sarcomere assembly of cells, thus strengthening them. Neonatal rat ventricular myocytes (NRVM) were cultured on polyacrylamide or polydimethylsiloxane substrates with stiffness of 10 kPa, 100 kPa, or 400 kPa, or glass with stiffness of 61.9 GPa. Cell size increased with stiffness. Two signaling pathways were explored, phosphorylation of focal adhesion kinase (p-FAK) and lipids by phosphatidylinositol 4,5-bisphosphate (PIP2). Subcellular distributions of both were determined in the sarcomeric fraction by antibody localization, and total amounts were measured by Western or dot blotting, respectively. More p-FAK and PIP2 distributed to the sarcomeres of NRVM grown on stiffer substrates. Actin assembly involves the actin capping protein Z (CapZ). Both actin and CapZ dynamic exchange were significantly increased on stiffer substrates when assessed by fluorescence recovery after photobleaching (FRAP) of green fluorescent protein tags. Blunting of actin FRAP by FAK inhibition implicates linkage from mechano-signalling pathways to cell growth. Thus, increased stiffness of cardiac disease can be modeled with polymeric materials to understand how the microenvironment regulates cardiac hypertrophy.
Collapse
Affiliation(s)
- Jieli Li
- a Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL 60612, USA
| | - Michael A Mkrtschjan
- b Department of Bioengineering, University of Illinois at Chicago, 851 South Morgan Street, Chicago, IL 60607, USA
| | - Ying-Hsi Lin
- a Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL 60612, USA
| | - Brenda Russell
- a Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL 60612, USA.,b Department of Bioengineering, University of Illinois at Chicago, 851 South Morgan Street, Chicago, IL 60607, USA
| |
Collapse
|
13
|
Lin YH, Warren CM, Li J, McKinsey TA, Russell B. Myofibril growth during cardiac hypertrophy is regulated through dual phosphorylation and acetylation of the actin capping protein CapZ. Cell Signal 2016; 28:1015-24. [PMID: 27185186 DOI: 10.1016/j.cellsig.2016.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 01/08/2023]
Abstract
The mechanotransduction signaling pathways initiated in heart muscle by increased mechanical loading are known to lead to long-term transcriptional changes and hypertrophy, but the rapid events for adaptation at the sarcomeric level are not fully understood. The goal of this study was to test the hypothesis that actin filament assembly during cardiomyocyte growth is regulated by post-translational modifications (PTMs) of CapZβ1. In rapidly hypertrophying neonatal rat ventricular myocytes (NRVMs) stimulated by phenylephrine (PE), two-dimensional gel electrophoresis (2DGE) of CapZβ1 revealed a shift toward more negative charge. Consistent with this, mass spectrometry identified CapZβ1 phosphorylation on serine-204 and acetylation on lysine-199, two residues which are near the actin binding surface of CapZβ1. Ectopic expression of dominant negative PKCɛ (dnPKCɛ) in NRVMs blunted the PE-induced increase in CapZ dynamics, as evidenced by the kinetic constant (Kfrap) of fluorescence recovery after photobleaching (FRAP), and concomitantly reduced phosphorylation and acetylation of CapZβ1. Furthermore, inhibition of class I histone deacetylases (HDACs) increased lysine-199 acetylation on CapZβ1, which increased Kfrap of CapZ and stimulated actin dynamics. Finally, we show that PE treatment of NRVMs results in decreased binding of HDAC3 to myofibrils, suggesting a signal-dependent mechanism for the regulation of sarcomere-associated CapZβ1 acetylation. Taken together, this dual regulation through phosphorylation and acetylation of CapZβ1 provides a novel model for the regulation of myofibril growth during cardiac hypertrophy.
Collapse
Affiliation(s)
- Ying-Hsi Lin
- Department of Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612-7342, United States; Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612-7342, United States
| | - Chad M Warren
- Department of Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612-7342, United States; Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612-7342, United States
| | - Jieli Li
- Department of Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612-7342, United States; Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612-7342, United States
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology and Center for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045-0508, United States
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612-7342, United States; Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612-7342, United States.
| |
Collapse
|
14
|
Lin YH, Swanson ER, Li J, Mkrtschjan MA, Russell B. Cyclic mechanical strain of myocytes modifies CapZβ1 post translationally via PKCε. J Muscle Res Cell Motil 2015; 36:329-37. [PMID: 26429793 DOI: 10.1007/s10974-015-9420-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/07/2015] [Indexed: 11/27/2022]
Abstract
The heart is exquisitely sensitive to mechanical stimuli and adapts to increased demands for work by enlarging the cardiomyocytes. In order to determine links between mechano-transduction mechanisms and hypertrophy, neonatal rat ventricular myocytes (NRVM) were subjected to physiologic strain for analysis of the dynamics of the actin capping protein, CapZ, and its post-translational modifications (PTM). CapZ binding rates were assessed after strain by fluorescence recovery after photobleaching (FRAP) of green fluorescent protein (GFP) expressed by a GFP-CapZβ1 adenovirus. To assess the role of the protein kinase C epsilon isoform (PKCε), rest or cyclic strain were combined with specific PKCε activation by constitutively active PKCε, or by inhibition with dominant negative PKCε (dnPKCε) expression. Significant increases of CapZ FRAP kinetics with strain were blunted by dnPKCε, suggesting that PKCε is involved in mechano-transduction signaling. Similar combinations of strain and PKC regulation in NRVMs were studied by PTM profiles of CapZβ1 using quantitative two-dimensional gel electrophoresis. The significantly increased charge on CapZ seen with mechanical strain was reversed by the addition of dnPKCε. Potential clinical relevance was confirmed in vivo by PTMs of CapZ in the failing heart of one-year old transgenic mice over-expressing PKCε. Furthermore, with strain there was significant PKCε translocation to the Z-disc and co-localization with CapZβ1 or α-actinin, which was quantified on confocal images. A hypothetical model is presented proposing that one destination of the mechanotransduction signaling pathways might be for PTMs of CapZ thereby regulating actin capping and filament assembly.
Collapse
Affiliation(s)
- Ying-Hsi Lin
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, MC 901, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Erik R Swanson
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, MC 901, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Jieli Li
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, MC 901, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Michael A Mkrtschjan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, MC 901, 835 S. Wolcott, Chicago, IL, 60612, USA.,Department of Bioengineering, College of Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, MC 901, 835 S. Wolcott, Chicago, IL, 60612, USA. .,Department of Bioengineering, College of Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
15
|
Owen JH, Graham MP, Chinn SB, Darr OF, Chepeha DB, Wolf GT, Bradford CR, Carey TE, Prince MEP. Novel method of cell line establishment utilizing fluorescence-activated cell sorting resulting in 6 new head and neck squamous cell carcinoma lines. Head Neck 2015; 38 Suppl 1:E459-67. [PMID: 25677579 DOI: 10.1002/hed.24019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/09/2014] [Accepted: 02/06/2015] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The purpose of this study was to present the establishment of new cell lines, which is important to cancer research. METHODS Six new head and neck squamous cell carcinoma cell lines were established using a novel fluorescence-activated cell sorting (FACS) method in order to overcome the barrier of fibroblast overgrowth and the susceptibility of primary tumors to fail in vitro. RESULTS Antibodies chosen for specific targeting of epithelial cells and fibroblasts successfully separated cells for line establishment in 6 of 12 attempts, providing an alternative method of establishing head and neck squamous cell carcinoma cell lines. Each attempt at cell line establishment resulted in an epithelial carcinoma population, which was genotyped and catalogued as a unique cell line, and a corresponding fibroblast population. CONCLUSION The selection of antibody markers could be optimized to aid in the establishment of any cancer cell line derived from any tumor tissue; this method is not limited to head and neck cancer. © 2015 Wiley Periodicals, Inc. Head Neck 38: E459-E467, 2016.
Collapse
Affiliation(s)
- John Henry Owen
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Martin P Graham
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Steven B Chinn
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Owen F Darr
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Douglas B Chepeha
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Gregory T Wolf
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Carol R Bradford
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Thomas E Carey
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| | - Mark E P Prince
- Department of Otolaryngology/Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Vandergriff AC, Hensley MT, Cheng K. Isolation and cryopreservation of neonatal rat cardiomyocytes. J Vis Exp 2015:52726. [PMID: 25938862 PMCID: PMC4541493 DOI: 10.3791/52726] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Cell culture has become increasingly important in cardiac research, but due to the limited proliferation of cardiomyocytes, culturing cardiomyocytes is difficult and time consuming. The most commonly used cells are neonatal rat cardiomyocytes (NRCMs), which require isolation every time cells are needed. The birth of the rats can be unpredictable. Cryopreservation is proposed to allow for cells to be stored until needed, yet freezing/thawing methods for primary cardiomyocytes are challenging due to the sensitivity of the cells. Using the proper cryoprotectant, dimethyl sulfoxide (DMSO), cryopreservation was achieved. By slowly extracting the DMSO while thawing the cells, cultures were obtained with viable NRCMs. NRCM phenotype was verified using immunocytochemistry staining for α-sarcomeric actinin. In addition, cells also showed spontaneous contraction after several days in culture. Cell viability after thawing was acceptable at 40-60%. In spite of this, the methods outlined allow one to easily cryopreserve and thaw NRCMs. This gives researchers a greater amount of flexibility in planning experiments as well as reducing the use of animals.
Collapse
Affiliation(s)
- Adam C Vandergriff
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University
| | - Michael Taylor Hensley
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University; The Cyrus Tang Hematology Center, Soochow University;
| |
Collapse
|
17
|
Arif IS, Hooper CL, Greco F, Williams AC, Boateng SY. Increasing doxorubicin activity against breast cancer cells using PPARγ-ligands and by exploiting circadian rhythms. Br J Pharmacol 2015; 169:1178-88. [PMID: 23578093 DOI: 10.1111/bph.12202] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/25/2013] [Accepted: 04/09/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Doxorubicin is effective against breast cancer, but its major side effect is cardiotoxicity. The aim of this study was to determine whether the efficacy of doxorubicin on cancer cells could be increased in combination with PPARγ agonists or chrono-optimization by exploiting the diurnal cycle. EXPERIMENTAL APPROACH We determined cell toxicity using MCF-7 cancer cells, neonatal rat cardiac myocytes and fibroblasts in this study. KEY RESULTS Doxorubicin damages the contractile filaments of cardiac myocytes and affects cardiac fibroblasts by significantly inhibiting collagen production and proliferation at the level of the cell cycle. Cyclin D1 protein levels decreased significantly following doxorubicin treatment indicative of a G1/S arrest. PPARγ agonists with doxorubicin increased the toxicity to MCF-7 cancer cells without affecting cardiac cells. Rosiglitazone and ciglitazone both enhanced anti-cancer activity when combined with doxorubicin (e.g. 50% cell death for doxorubicin at 0.1 μM compared to 80% cell death when combined with rosiglitazone). Thus, the therapeutic dose of doxorubicin could be reduced by 20-fold through combination with the PPARγ agonists, thereby reducing adverse effects on the heart. The presence of melatonin also significantly increased doxorubicin toxicity, in cardiac fibroblasts (1 μM melatonin) but not in MCF-7 cells. CONCLUSIONS AND IMPLICATIONS Our data show, for the first time, that circadian rhythms play an important role in doxorubicin toxicity in the myocardium; doxorubicin should be administered mid-morning, when circulating levels of melatonin are low, and in combination with rosiglitazone to increase therapeutic efficacy in cancer cells while reducing the toxic effects on the heart.
Collapse
Affiliation(s)
- I S Arif
- School of Pharmacy, University of Reading, Reading, UK
| | | | | | | | | |
Collapse
|
18
|
Prabhakaran MP, Vatankhah E, Kai D, Ramakrishna S. Methods for Nano/Micropatterning of Substrates: Toward Stem Cells Differentiation. INT J POLYM MATER PO 2014. [DOI: 10.1080/00914037.2014.945207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
19
|
Montani C, Steimberg N, Boniotti J, Biasiotto G, Zanella I, Diafera G, Biunno I, Caimi L, Mazzoleni G, Di Lorenzo D. Fibroblasts maintained in 3 dimensions show a better differentiation state and higher sensitivity to estrogens. Toxicol Appl Pharmacol 2014; 280:421-33. [DOI: 10.1016/j.taap.2014.08.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 07/24/2014] [Accepted: 08/12/2014] [Indexed: 01/07/2023]
|
20
|
Cardiomyocyte subdomain contractility arising from microenvironmental stiffness and topography. Biomech Model Mechanobiol 2014; 14:589-602. [PMID: 25273278 DOI: 10.1007/s10237-014-0624-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 09/19/2014] [Indexed: 01/27/2023]
Abstract
Cellular structure and function are interdependent. To understand this relationship in beating heart cells, individual neonatal rat ventricular myocytes (NRVMs) were analyzed one and 3 days after plating when cultured on different stiffness (100, 400 kPa) and surface structures (flat or [Formula: see text] high, [Formula: see text] diameter, microposts spaced [Formula: see text] apart) manufactured from polydimethylsiloxane. Myofibril structure seen by immunohistochemistry was organized in three dimensions when NRVMs were attached to microposts. On day three, paxillin distribution near the post serving as cellular anchorage was quantified on both soft posts (12.04 % of total voxel count) and stiff posts (8.16 %). Living NRVMs were analyzed using line scans for sarcomeric shortening and shortening velocity, and traction force microscopy for surface stress and surface tension. One day after plating, NRVMs shortened more on soft posts ([Formula: see text] at [Formula: see text]) compared to either soft flat ([Formula: see text] at [Formula: see text]), stiff posts ([Formula: see text] at [Formula: see text]) or stiff flat ([Formula: see text] at [Formula: see text]). NRVMs have decreased shortening and shortening velocity on soft posts ([Formula: see text] at [Formula: see text]) compared to soft flat ([Formula: see text] at [Formula: see text]) substrates. The surface stress and surface tension increased over time for both soft post ([Formula: see text] and [Formula: see text] to [Formula: see text] and [Formula: see text]) and flat ([Formula: see text] and [Formula: see text] to [Formula: see text] and [Formula: see text]) substrates. Paxillin displacement during contraction on day three was significantly greater in NRVMs attached to soft posts [Formula: see text] compared to flat [Formula: see text] substrates. The volume and time creating four-dimensional data, interpreted by structural engineering theory, demonstrate subdomain structure is maintained by the counterbalance between the external load acting upon and the internal forces generated by the cardiomyocyte. These findings provide further insight into localized regulation of cellular mechanical function.
Collapse
|
21
|
Doroudian G, Pinney J, Ayala P, Los T, Desai TA, Russell B. Sustained delivery of MGF peptide from microrods attracts stem cells and reduces apoptosis of myocytes. Biomed Microdevices 2014; 16:705-15. [PMID: 24908137 PMCID: PMC4418932 DOI: 10.1007/s10544-014-9875-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Local release of drugs may have many advantages for tissue repair but also presents major challenges. Bioengineering approaches allow microstructures to be fabricated that contain bioactive peptides for sustained local delivery. Heart tissue damage is associated with local increases in mechano growth factor (MGF), a member of the IGF-1 family. The E domain of MGF peptide is anti-apoptotic and a stem cell homing factor. The objectives of this study were to fabricate a microrod delivery device of poly (ethylene glycol) dimethacrylate (PEGDMA) hydrogel loaded with MGF peptide and to determine the elution profile and bioactivity of MGF. The injectable microrods are 30 kPa stiffness and 15 μm widths by 100 μm lengths, chosen to match heart stiffness and myocyte size. Successful encapsulation of native MGF peptide within microrods was achieved with delivery of MGF for 2 weeks, as measured by HPLC. Migration of human mesenchymal stem cells (hMSCs) increased with MGF microrod treatment (1.72 ± 0.23, p < 0.05). Inhibition of the apoptotic pathway in neonatal rat ventricular myocytes was induced by 8 h of hypoxia (1 % O2). Protection from apoptosis by MGF microrod treatment was shown by the TUNEL assay and increased Bcl-2 expression (2 ± 0.19, p < 0.05). Microrods without MGF regulated the cytoskeleton, adhesion, and proliferation of hMSCs, and MGF had no effect on these properties. Therefore, the combination microdevice provided both the mechanical cues and 2-week MGF bioactivity to reduce apoptosis and recruit stem cells, suggesting potential use of MGF microrods for cardiac regeneration therapy in vivo.
Collapse
Affiliation(s)
- Golnar Doroudian
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - James Pinney
- Department of Physiology and Division of Bioengineering, University of California at San Francisco, San Francisco, CA, USA
| | - Perla Ayala
- Department of Physiology and Division of Bioengineering, University of California at San Francisco, San Francisco, CA, USA
| | - Tamara Los
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| | - Tejal A. Desai
- Department of Physiology and Division of Bioengineering, University of California at San Francisco, San Francisco, CA, USA
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| |
Collapse
|
22
|
Li J, Tanhehco EJ, Russell B. Actin dynamics is rapidly regulated by the PTEN and PIP2 signaling pathways leading to myocyte hypertrophy. Am J Physiol Heart Circ Physiol 2014; 307:H1618-25. [PMID: 25260617 DOI: 10.1152/ajpheart.00393.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Mature cardiac myocytes are terminally differentiated, and the heart has limited capacity to replace lost myocytes. Thus adaptation of myocyte size plays an important role in the determination of cardiac function. The hypothesis tested is that regulation of the dynamic exchange of actin leads to cardiac hypertrophy. ANG II was used as a hypertrophic stimulant in mouse heart and neonatal rat ventricular myocytes (NRVMs) in culture for assessment of a mechanism for regulation of actin dynamics by phosphatidylinositol 4,5-bisphosphate (PIP2). Actin dynamics in NRVMs rapidly increased in a PIP2-dependent manner, measured by imaging and fluorescence recovery after photobleaching (FRAP). A significant increase in PIP2 levels was found by immunoblotting in both adult mouse heart tissue and cultured NRVMs. Inhibition of phosphatase and tensin homolog (PTEN) in NRVMs markedly blunted ANG II-induced increases in actin dynamics, the PIP2 level, and cell size. Furthermore, PTEN activity was dramatically upregulated in ANG II-treated NRVMs but downregulated when PTEN inhibitors were used. The time course of the rise in the PIP2 level was inversely related to the fall in the PIP3 level, which was significant by 30 min in ANG II-treated NRVMs. However, significant translocation of PTEN to the plasma membrane occurred by 10 min, suggesting a crucial initial step for PTEN for the cellular responses to ANG II. In conclusion, PTEN and PIP2 signaling may play an important role in myocyte hypertrophy by the regulation of actin filament dynamics, which is induced by ANG II stimulation.
Collapse
Affiliation(s)
- Jieli Li
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Elaine J Tanhehco
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
23
|
Pinney JR, Melkus G, Cerchiari A, Hawkins J, Desai TA. Novel functionalization of discrete polymeric biomaterial microstructures for applications in imaging and three-dimensional manipulation. ACS APPLIED MATERIALS & INTERFACES 2014; 6:14477-14485. [PMID: 25068888 PMCID: PMC4149329 DOI: 10.1021/am503778t] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/14/2014] [Indexed: 05/30/2023]
Abstract
Adapting ways to functionalize polymer materials is becoming increasingly important to their implementation in translational biomedical sciences. By tuning the mechanical, chemical, and biological qualities of these materials, their applications can be broadened, opening the door for more advanced integration into modern medical techniques. Here, we report on a method to integrate chemical functionalizations into discrete, microscale polymer structures, which are used for tissue engineering applications, for in vivo localization, and three-dimensional manipulation. Iron oxide nanoparticles were incorporated into the polymer matrix using common photolithographic techniques to create stably functional microstructures with magnetic potential. Using magnetic resonance imaging (MRI), we can promote visualization of microstructures contained in small collections, as well as facilitate the manipulation and alignment of microtopographical cues in a realistic tissue environment. Using similar polymer functionalization techniques, fluorine-containing compounds were also embedded in the polymer matrix of photolithographically fabricated microstructures. The incorporation of fluorine-containing compounds enabled highly sensitive and specific detection of microstructures in physiologic settings using fluorine MRI techniques ((19)F MRI). These functionalization strategies will facilitate more reliable noninvasive tracking and characterization of microstructured polymer implants as well as have implications for remote microstructural scaffolding alignment for three-dimensional tissue engineering applications.
Collapse
Affiliation(s)
- James R. Pinney
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 Fourth Street, Byers Hall Room 203, San Francisco, California 94158, United States
- UC Berkeley-UCSF Graduate Program in Bioengineering, 1700 Fourth Street, Byers Hall
Room 216, San Francisco, California 94158, United States
| | - Gerd Melkus
- Department
of Radiology, UCSF Imaging Center at China Basin, University of California, San Francisco, 185 Berry Street, Suite 190, Lobby 6, San Francisco, California 94107, United States
- Department
of Medical Imaging, Ottawa Hospital, 1053 Carling Avenue, Ottawa K1Y 4E9, Ontario, Canada
| | - Alec Cerchiari
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 Fourth Street, Byers Hall Room 203, San Francisco, California 94158, United States
- UC Berkeley-UCSF Graduate Program in Bioengineering, 1700 Fourth Street, Byers Hall
Room 216, San Francisco, California 94158, United States
| | - James Hawkins
- Department
of Radiology, UCSF Imaging Center at China Basin, University of California, San Francisco, 185 Berry Street, Suite 190, Lobby 6, San Francisco, California 94107, United States
| | - Tejal A. Desai
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 Fourth Street, Byers Hall Room 203, San Francisco, California 94158, United States
| |
Collapse
|
24
|
Pinney JR, Du KT, Ayala P, Fang Q, Sievers RE, Chew P, Delrosario L, Lee RJ, Desai TA. Discrete microstructural cues for the attenuation of fibrosis following myocardial infarction. Biomaterials 2014; 35:8820-8828. [PMID: 25047625 DOI: 10.1016/j.biomaterials.2014.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/02/2014] [Indexed: 01/14/2023]
Abstract
Chronic fibrosis caused by acute myocardial infarction (MI) leads to increased morbidity and mortality due to cardiac dysfunction. We have developed a therapeutic materials strategy that aims to mitigate myocardial fibrosis by utilizing injectable polymeric microstructures to mechanically alter the microenvironment. Polymeric microstructures were fabricated using photolithographic techniques and studied in a three-dimensional culture model of the fibrotic environment and by direct injection into the infarct zone of adult rats. Here, we show dose-dependent down-regulation of expression of genes associated with the mechanical fibrotic response in the presence of microstructures. Injection of this microstructured material into the infarct zone decreased levels of collagen and TGF-β, increased elastin deposition and vascularization in the infarcted region, and improved functional outcomes after six weeks. Our results demonstrate the efficacy of these discrete anti-fibrotic microstructures and suggest a potential therapeutic materials approach for combatting pathologic fibrosis.
Collapse
Affiliation(s)
- James R Pinney
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; UCSF Medical Scientist Training Program, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA
| | - Kim T Du
- UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Perla Ayala
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; Beth Israel Deaconess Medical Center, Department of Surgery, Center for Life Science Surgery/BIDMC, 11th Floor, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Qizhi Fang
- UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Richard E Sievers
- UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Patrick Chew
- UCSF Bioengineering and Therapeutic Sciences, 1700 4th Street, Byers Hall Room 203, San Francisco, CA 94158, USA
| | - Lawrence Delrosario
- UCSF School of Medicine, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Randall J Lee
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Tejal A Desai
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; UCSF Bioengineering and Therapeutic Sciences, 1700 4th Street, Byers Hall Room 203, San Francisco, CA 94158, USA.
| |
Collapse
|
25
|
Li J, Russell B. Phosphatidylinositol 4,5-bisphosphate regulates CapZβ1 and actin dynamics in response to mechanical strain. Am J Physiol Heart Circ Physiol 2013; 305:H1614-23. [PMID: 24043251 DOI: 10.1152/ajpheart.00477.2013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanical stress causes filament remodeling leading to myocyte hypertrophy and heart failure. The actin capping protein Z (CapZ) tightly binds to the barbed end of actin filaments, thus regulating actin assembly. The hypothesis is that the binding between CapZ and the actin filament is modulated through phosphatidylinositol 4,5-bisphosphate (PIP2) and how the COOH-terminus of CapZβ1 regulates this binding. Primary neonatal rat ventricular myocytes (NRVMs) were strained at 10% amplitude and 1-Hz frequency. Dot blotting measured the PIP2 amount, and affinity precipitation assay assessed the direct interaction between PIP2 and CapZβ1. Fluorescence recovery after photobleaching of green fluorescent protein-CapZβ1 and actin-green fluorescent protein after 1 h of strain shows the dynamics significantly increased above the unstrained group. The increases in CapZ and actin dynamics were blunted by neomycin, suggesting PIP2 signaling is involved. The amount of PIP2 dramatically increased in NRVMs strained for 1 h. With a ROCK or RhoA inhibitor, changes were markedly reduced. Subcellular fractionation and antibody localization showed PIP2 distributed to the sarcomeres. More PIP2-bound CapZβ1 was found in strained NRVMs. Less PIP2 bound to the CapZβ1 with its COOH-terminus intact than in the COOH-terminal mutant of CapZβ1, suggesting some inhibitory role for the COOH-terminus. Myocyte hypertrophy normally induced by 48 h of cyclic strain was blunted by dominant negative RhoA or neomycin. This suggests that after many hours of cyclic strain, a possible mechanism for cell hypertrophy is the accumulation of thin filament assembly triggered partially by the increased PIP2 level and its binding to CapZ.
Collapse
Affiliation(s)
- Jieli Li
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois
| | | |
Collapse
|
26
|
Wang L, Liu L, Magome N, Agladze K, Chen Y. Influence of patterned topographic features on the formation of cardiac cell clusters and their rhythmic activities. Biofabrication 2013; 5:035013. [DOI: 10.1088/1758-5082/5/3/035013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
27
|
Yang N, Yang M, Bi S, Chen L, Zhu Z, Gao Y, Du Z. Cells behaviors and genotoxicity on topological surface. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2013; 33:3465-73. [PMID: 23706235 DOI: 10.1016/j.msec.2013.04.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/20/2013] [Accepted: 04/16/2013] [Indexed: 12/11/2022]
|
28
|
Yang CY, Sung CY, Shuai HH, Cheng CM, Yeh JA. Probing cellular behaviors through nanopatterned chitosan membranes. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2013; 14:044406. [PMID: 27877591 PMCID: PMC5090317 DOI: 10.1088/1468-6996/14/4/044406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/18/2013] [Indexed: 05/28/2023]
Abstract
This paper describes a high-throughput method for developing physically modified chitosan membranes to probe the cellular behavior of MDCK epithelial cells and HIG-82 fibroblasts adhered onto these modified membranes. To prepare chitosan membranes with micro/nanoscaled features, we have demonstrated an easy-to-handle, facile approach that could be easily integrated with IC-based manufacturing processes with mass production potential. These physically modified chitosan membranes were observed by scanning electron microscopy to gain a better understanding of chitosan membrane surface morphology. After MDCK cells and HIG-82 fibroblasts were cultured on these modified chitosan membranes for various culture durations (i.e. 1, 2, 4, 12 and 24 h), they were investigated to decipher cellular behavior. We found that both cells preferred to adhere onto a flat surface rather than on a nanopatterned surface. However, most (> 80%) of the MDCK cells showed rounded morphology and would suspend in the cultured medium instead of adhering onto the planar surface of negatively nanopatterned chitosan membranes. This means different cell types (e.g. fibroblasts versus epithelia) showed distinct capabilities/preferences of adherence for materials of varying surface roughness. We also showed that chitosan membranes could be re-used at least nine times without significant contamination and would provide us consistency for probing cell-material interactions by permitting reuse of the same substrate. We believe these results would provide us better insight into cellular behavior, specifically, microscopic properties and characteristics of cells grown under unique, nanopatterned cell-interface conditions.
Collapse
|
29
|
Radisic M, Christman KL. Materials science and tissue engineering: repairing the heart. Mayo Clin Proc 2013; 88:884-98. [PMID: 23910415 PMCID: PMC3786696 DOI: 10.1016/j.mayocp.2013.05.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/01/2013] [Accepted: 05/06/2013] [Indexed: 01/12/2023]
Abstract
Heart failure after a myocardial infarction continues to be a leading killer in the Western world. Currently, there are no therapies that effectively prevent or reverse the cardiac damage and negative left ventricular remodeling process that follows a myocardial infarction. Because the heart has limited regenerative capacity, there has been considerable effort to develop new therapies that could repair and regenerate the myocardium. Although cell transplantation alone was initially studied, more recently, tissue engineering strategies using biomaterial scaffolds have been explored. In this review, we cover the different approaches to engineering the myocardium, including cardiac patches, which are in vitro-engineered constructs of functional myocardium, and injectable scaffolds, which can either encourage endogenous repair and regeneration or act as vehicles to support the delivery of cells and other therapeutics.
Collapse
Affiliation(s)
- Milica Radisic
- Institute of Biomaterials and Biomedical Engineering and the Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| | | |
Collapse
|
30
|
Kharaziha M, Nikkhah M, Shin SR, Annabi N, Masoumi N, Gaharwar AK, Camci-Unal G, Khademhosseini A. PGS:Gelatin nanofibrous scaffolds with tunable mechanical and structural properties for engineering cardiac tissues. Biomaterials 2013; 34:6355-66. [PMID: 23747008 DOI: 10.1016/j.biomaterials.2013.04.045] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 04/23/2013] [Indexed: 12/27/2022]
Abstract
A significant challenge in cardiac tissue engineering is the development of biomimetic grafts that can potentially promote myocardial repair and regeneration. A number of approaches have used engineered scaffolds to mimic the architecture of the native myocardium tissue and precisely regulate cardiac cell functions. However, previous attempts have not been able to simultaneously recapitulate chemical, mechanical, and structural properties of the myocardial extracellular matrix (ECM). In this study, we utilized an electrospinning approach to fabricate elastomeric biodegradable poly(glycerol sebacate) (PGS):gelatin nanofibrous scaffolds with a wide range of chemical composition, stiffness and anisotropy. Our findings demonstrated that through incorporation of PGS, it is possible to create nanofibrous scaffolds with well-defined anisotropy that mimic the left ventricular myocardium architecture. Furthermore, we studied attachment, proliferation, differentiation and alignment of neonatal rat cardiac fibroblast cells (CFs) as well as protein expression, alignment, and contractile function of cardiomyocyte (CMs) on PGS:gelatin scaffolds with variable amount of PGS. Notably, aligned nanofibrous scaffold, consisting of 33 wt. % PGS, induced optimal synchronous contractions of CMs while significantly enhanced cellular alignment. Overall, our study suggests that the aligned nanofibrous PGS:gelatin scaffold support cardiac cell organization, phenotype and contraction and could potentially be used to develop clinically relevant constructs for cardiac tissue engineering.
Collapse
Affiliation(s)
- Mahshid Kharaziha
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Lin YH, Li J, Swanson ER, Russell B. CapZ and actin capping dynamics increase in myocytes after a bout of exercise and abates in hours after stimulation ends. J Appl Physiol (1985) 2013; 114:1603-9. [PMID: 23493359 DOI: 10.1152/japplphysiol.01283.2012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The time course of the response and recovery after acute activity seen in exercise is not well understood. The goal of this work is to address how proteins of the thin filament (actin and its capping protein CapZ) are changed by 1 h of mechanical stimulation and return to baseline over time. Neonatal rat ventricular myocytes in culture were subjected to cyclic 10% strain at 1 Hz for 1 h to mimic increased mechanical loading during exercise. CapZ and actin dynamics were analyzed by fluorescence recovery after photobleaching (FRAP) using CapZβ1-GFP, actin-GFP, or actin-RFP. After cyclic strain, CapZ dynamics increased above resting controls and abated 2-3 h after cessation of the cyclic strain. Similarly, actin dynamics initially increased and abated 1.5-2 h after the end of stimulation. Neurohormonal hypertrophic stimulation by phenylephrine or norepinephrine treatments also elevated actin dynamics but required a much longer time of treatment (24-48 h) to be detectable. The actin capping mechanism was explored by use of expression of CapZβ1 with a COOH-terminal deletion (CapZβ1ΔC). Increased dynamics of actin seen with CapZβ1ΔC was similar to the response to cyclic strain. Thus it is possible that mechanical stimulation alters the dynamics for CapZ capping of the actin filament through the CapZβ1 COOH terminus, known as the β tentacle, thereby remodeling sarcomeres in cardiac myocytes. This adaptive mechanism, which is probably regulating thin-filament addition, declines a few hours after the end of a bout of exercise.
Collapse
Affiliation(s)
- Ying-Hsi Lin
- Department of Physiology and Biophysics, University of Illinois at Chicago, College of Medicine, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
32
|
Doroudian G, Curtis MW, Gang A, Russell B. Cyclic strain dominates over microtopography in regulating cytoskeletal and focal adhesion remodeling of human mesenchymal stem cells. Biochem Biophys Res Commun 2013; 430:1040-6. [PMID: 23257161 PMCID: PMC3612286 DOI: 10.1016/j.bbrc.2012.11.120] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 11/21/2012] [Indexed: 11/25/2022]
Abstract
Human bone marrow-derived mesenchymal stem cell (hMSCs) function depends on chemical factors and also on the physical cues of the microenvironmental niche. Here, this physical microenvironment is recapitulated with controlled modes of mechanical strain applied to substrata containing three-dimensional features in order to analyze the effects on cell morphology, focal adhesion distribution, and gene expression. Ten percentage of strain at 1 Hz is delivered for 48 h to hMSCs cultured on flat surfaces, or on substrata with 15 μm-high microtopographic posts spaced 75 μm apart. Adding strain to microtopography produced stable semicircular focal adhesions, and actin spanning from post to post. Strain dominated over microtopography for expression of genes for the cytoskeleton (caldesmon-1 and calponin 3), cell adhesion (integrin-α2, vinculin, and paxillin), and extracellular matrix remodeling (MMP13) (p<0.05). Overall, attention to external mechanical stimuli is necessary for optimizing the stem cell niche for regenerative medicine.
Collapse
Affiliation(s)
- Golnar Doroudian
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Matthew W. Curtis
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Anjulie Gang
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
33
|
Kim DH, Kim P, Suh K, Kyu Choi S, Ho Lee S, Kim B. Modulation of adhesion and growth of cardiac myocytes by surface nanotopography. CONFERENCE PROCEEDINGS : ... ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL CONFERENCE 2012; 2005:4091-4. [PMID: 17281132 DOI: 10.1109/iembs.2005.1615362] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We have introduced well-defined nanopillar arrays of polyethylene glycol (PEG) as a platform for studying the adhesion and growth of cultured cardiomyocytes. The nanopillar arrays were fabricated by using a simple molding technique involving the placement of a patterned polyurethane acrylate mold on top of a drop-dispensed ultraviolet (UV) curable PEG polymer followed by UV exposure and mold removal. The adhesion and growth of cardiomyocytes turned out be guided by an external nanotopography, which has been characterized in terms of cell morphology and cytoskeletal arrangement. In particular, the nanopillars provided guiding posts to both elongating filopodia and expanding lamellipodia. Interestingly, the 3D growth of cardiomyocytes was mediated by the increased hydrophobicity of the nanostructured PEG substrate, indicating that the cell adhesion and growth is very sensitive to the nanotopography. The precise nanostructures of PEG-based polymer with controlled geometrical features presented in this study not only open opportunities for understanding and tailoring cell adhesion and growth, but could serve as a template for better tissue engineering by controlling cellular activities at the molecular level.
Collapse
Affiliation(s)
- Deok-Ho Kim
- Member, IEEE, Microsystem Research Center, Korea Institute of Science and Technology, Seoul, 138-791, Korea
| | | | | | | | | | | |
Collapse
|
34
|
Łopacińska JM, Grădinaru C, Wierzbicki R, Købler C, Schmidt MS, Madsen MT, Skolimowski M, Dufva M, Flyvbjerg H, Mølhave K. Cell motility, morphology, viability and proliferation in response to nanotopography on silicon black. NANOSCALE 2012; 4:3739-3745. [PMID: 22614757 DOI: 10.1039/c2nr11455k] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Knowledge of cells' interactions with nanostructured materials is fundamental for bio-nanotechnology. We present results for how individual mouse fibroblasts from cell line NIH3T3 respond to highly spiked surfaces of silicon black that were fabricated by maskless reactive ion etching (RIE). We did standard measurements of cell viability, proliferation, and morphology on various surfaces. We also analyzed the motility of cells on the same surfaces, as recorded in time lapse movies of sparsely populated cell cultures. We find that motility and morphology vary strongly with nano-patterns, while viability and proliferation show little dependence on substrate type. We conclude that motility analysis can show a wide range of cell responses e.g. over a factor of two in cell speed to different nano-topographies, where standard assays, such as viability or proliferation, in the tested cases show much less variation of the order 10-20%.
Collapse
Affiliation(s)
- Joanna M Łopacińska
- Department of Micro- and Nanotechnology, Technical University of Denmark, Ørsteds Plads 345a, 2800 Kongens Lyngby, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hörning M, Takagi S, Yoshikawa K. Controlling activation site density by low-energy far-field stimulation in cardiac tissue. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2012; 85:061906. [PMID: 23005126 DOI: 10.1103/physreve.85.061906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 03/19/2012] [Indexed: 06/01/2023]
Abstract
Tachycardia and fibrillation are potentially fatal arrhythmias associated with the formation of rotating spiral waves in the heart. Presently, the termination of these types of arrhythmia is achieved by use of antitachycardia pacing or cardioversion. However, these techniques have serious drawbacks, in that they either have limited application or produce undesirable side effects. Low-energy far-field stimulation has recently been proposed as a superior therapy. This proposed therapeutic method would exploit the phenomenon in which the application of low-energy far-field shocks induces a large number of activation sites ("virtual electrodes") in tissue. It has been found that the formation of such sites can lead to the termination of undesired states in the heart and the restoration of normal beating. In this study we investigate a particular aspect of this method. Here we seek to determine how the activation site density depends on the applied electric field through in vitro experiments carried out on neonatal rat cardiac tissue cultures. The results indicate that the activation site density increases exponentially as a function of the intracellular conductivity and the level of cell isotropy. Additionally, we report numerical results obtained from bidomain simulations of the Beeler-Reuter model that are quantitatively consistent with our experimental results. Also, we derive an intuitive analytical framework that describes the activation site density and provides useful information for determining the ratio of longitudinal to transverse conductivity in a cardiac tissue culture. The results obtained here should be useful in the development of an actual therapeutic method based on low-energy far-field pacing. In addition, they provide a deeper understanding of the intrinsic properties of cardiac cells.
Collapse
Affiliation(s)
- Marcel Hörning
- Department of Physics, Graduate School of Science, Kyoto University, Japan.
| | | | | |
Collapse
|
36
|
He XL, Nie PP, Chen BZ, Li XX, Chen L, Guo G, Zhang R. A novel method to fabricate thermoresponsive microstructures with improved cell attachment/detachment properties. J Biomed Mater Res A 2012; 100:1946-53. [PMID: 22492628 DOI: 10.1002/jbm.a.34138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 02/09/2012] [Accepted: 02/21/2012] [Indexed: 11/07/2022]
Abstract
A novel, simple, and rapid method to fabricate thermoresponsive micropatterned substrate for cell adhesion, growth, and thermally induced detachment was developed. Thermoresponsive polymer, poly(N-isopropylacrylamide) (PNIPAAm), was grafted onto the surface of polystyrene (PS) film with microstructure by plasma-induced graft polymerization technique. The thermoresponsive micropatterned films were characterized by attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy, hydrogen nuclear magnetic resonance ((1) H NMR) spectroscopy, X-ray photoelectron spectroscopy (XPS) and scanning electron microscope (SEM). These results indicated that the grafting ratio of PNIPAAm increased with increasing roughness of PS film. However, the microstructures on the substrate were not affected by grafted PNIPAAm. The optimal grafting conditions, such as plasma treatment time, monomer concentration, graft polymerization time, and graft medium were investigated. The thermoresponsive micropatterned films were investigated with the fibroblast cell (L929) adhesion, proliferation, and thermally induced detachment assay. The microstructure on the thermoresponsive micropatterned substrate facilitated cell adhesion above the lower critical solution temperature (LCST) of PNIPAAm and cell detachment below the LCST. Moreover, it can be used to regulate cell organization and tissue growth.
Collapse
Affiliation(s)
- Xiao-Ling He
- Department of Chemistry, School of Environment and Chemical Engineering, Tianjin Polytechnic University, Tianjin 300160, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Hörning M, Kidoaki S, Kawano T, Yoshikawa K. Rigidity matching between cells and the extracellular matrix leads to the stabilization of cardiac conduction. Biophys J 2012; 102:379-87. [PMID: 22325259 PMCID: PMC3274804 DOI: 10.1016/j.bpj.2011.12.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 12/04/2011] [Accepted: 12/12/2011] [Indexed: 11/15/2022] Open
Abstract
Biomechanical dynamic interactions between cells and the extracellular environment dynamically regulate physiological tissue behavior in living organisms, such as that seen in tissue maintenance and remodeling. In this study, the substrate-induced modulation of synchronized beating in cultured cardiomyocyte tissue was systematically characterized on elasticity-tunable substrates to elucidate the effect of biomechanical coupling. We found that myocardial conduction is significantly promoted when the rigidity of the cell culture environment matches that of the cardiac cells (4 kiloPascals). The stability of spontaneous target wave activity and calcium transient alternans in high frequency-paced tissue were both enhanced when the cell substrate and cell tissue showed the same rigidity. By adapting a simple theoretical model, we reproduced the experimental trend on the rigidity matching for the synchronized excitation. We conclude that rigidity matching in cell-to-substrate interactions critically improves cardiomyocyte-tissue synchronization, suggesting that mechanical coupling plays an essential role in the dynamic activity of the beating heart.
Collapse
Affiliation(s)
- Marcel Hörning
- Department of Physics, Graduate School of Science, Kyoto University, Japan
| | - Satoru Kidoaki
- Division of Biomolecular Chemistry, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| | - Takahito Kawano
- Division of Biomolecular Chemistry, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| | - Kenichi Yoshikawa
- Department of Physics, Graduate School of Science, Kyoto University, Japan
| |
Collapse
|
38
|
Zhang B, Xiao Y, Hsieh A, Thavandiran N, Radisic M. Micro- and nanotechnology in cardiovascular tissue engineering. NANOTECHNOLOGY 2011; 22:494003. [PMID: 22101261 DOI: 10.1088/0957-4484/22/49/494003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
While in nature the formation of complex tissues is gradually shaped by the long journey of development, in tissue engineering constructing complex tissues relies heavily on our ability to directly manipulate and control the micro-cellular environment in vitro. Not surprisingly, advancements in both microfabrication and nanofabrication have powered the field of tissue engineering in many aspects. Focusing on cardiac tissue engineering, this paper highlights the applications of fabrication techniques in various aspects of tissue engineering research: (1) cell responses to micro- and nanopatterned topographical cues, (2) cell responses to patterned biochemical cues, (3) controlled 3D scaffolds, (4) patterned tissue vascularization and (5) electromechanical regulation of tissue assembly and function.
Collapse
Affiliation(s)
- Boyang Zhang
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 164 College Street, Rm 407, Toronto, ON M5S 3G9, Canada
| | | | | | | | | |
Collapse
|
39
|
Bacakova L, Filova E, Parizek M, Ruml T, Svorcik V. Modulation of cell adhesion, proliferation and differentiation on materials designed for body implants. Biotechnol Adv 2011; 29:739-67. [PMID: 21821113 DOI: 10.1016/j.biotechadv.2011.06.004] [Citation(s) in RCA: 577] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 05/30/2011] [Accepted: 06/09/2011] [Indexed: 12/12/2022]
Affiliation(s)
- Lucie Bacakova
- Department of Growth and Differentiation of Cell Populations, Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1082, 14220 Prague 4-Krc, Czech Republic.
| | | | | | | | | |
Collapse
|
40
|
Abstract
One of the central challenges in cardiac tissue engineering is the control of the assembly and organization of functional cardiac tissue. Maintenance of a three-dimensional tissue architecture is key to myocardial function in vivo, and a variety of studies hint that provision of topological cues within scaffolds can facilitate the engineering of functional myocardial tissue by promoting this architecture. To explore this possibility in an isolated and well-defined fashion, we have designed scaffolds of polydimethylsiloxane (PDMS) with microtopographic pillars ("micropegs") to provide cells with defined structures with which to interact in three dimensions. We show that these surfaces permit HL-1 cardiomyocytes to grow, form myofibrillar structures and cell-cell adhesions, and beat spontaneously. Additionally, the cells and their nuclei interact with the full length of the micropegs, indicating that the micropegs promote a three-dimensional cytoarchitecture in the context of a two-dimensional scaffold. We also show that the number of cells interacting with a micropeg can be controlled by manipulating incubation time, micropeg spatial arrangement, or micropeg diameter. Western blots reveal that the expression of the junctional markers N-cadherin and connexin 43 is upregulated in the presence of specific arrangements of micropegs, suggesting that micropegs can enhance cardiomyocyte function. Together, these data show that microtopography can be used to provide three-dimensional adhesion and control the assembly of functional cardiac tissue on a two-dimensional surface.
Collapse
Affiliation(s)
- Anuj A Patel
- The UC Berkeley - UCSF Graduate Program in Bioengineering, USA
| | | | | |
Collapse
|
41
|
Ayala P, Desai TA. Integrin α3 blockade enhances microtopographical down-regulation of α-smooth muscle actin: role of microtopography in ECM regulation. Integr Biol (Camb) 2011; 3:733-41. [PMID: 21666923 DOI: 10.1039/c1ib00012h] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Development of functional engineered matrices for regenerative therapies can benefit from an understanding of how physical cues at the microscale affect cell behavior. In this work, we use microfabricated systems to study how stiffness and microscale topographical cues in the form of "micropegs" affect extracellular matrix synthesis. Previous work from our lab has shown that microtopographical cues in 2D and 3D systems decrease cellular proliferation and regulate matrix synthesis. In this work, the combined role of stiffness and topography on ECM synthesis is investigated in a 2D micropeg system. These studies show that fibroblasts cultured on polydimethylsiloxane (PDMS) substrates with micropegs have reduced expression of collagen type I (Col I) and collagen type VI (Col VI) compared to fibroblasts cultured on flat substrates. In addition, cells on micropegged substrates exhibit down-regulation of other important regulators of ECM synthesis such as α-smooth muscle actin (α-SMA), and integrin α3 (Int α3). Interestingly, this effect is dependent on the contractility and adhesion of the cells. When cultured in the presence of RhoA kinase (ROCK) and myosin light chain kinase (MLCK) inhibitors, no significant differences in the expression of collagen, α-SMA, Int α3, and TGFB1 are observed. Additionally, disruptions in cell adhesion prevent microtopographical regulation of ECM synthesis. When using an antibody to block the extracellular domain of Int α3, no differences in the expression of collagen are observed and blocking Int α3 results in enhanced down-regulation of α-SMA on the stiffer micropegged substrates. These findings demonstrate that regulation of extracellular matrix production by cells on a synthetic substrate can be guided via physical cues at the microscale, and add to the body of knowledge on the role of integrin-mediated mechanotransduction.
Collapse
Affiliation(s)
- Perla Ayala
- UCSF/UC Berkeley Joint Graduate Group in Bioengineering, USA
| | | |
Collapse
|
42
|
Ayala P, Lopez JI, Desai TA. Microtopographical cues in 3D attenuate fibrotic phenotype and extracellular matrix deposition: implications for tissue regeneration. Tissue Eng Part A 2011; 16:2519-27. [PMID: 20235832 DOI: 10.1089/ten.tea.2009.0815] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent studies have highlighted the role of external biophysical cues on cell morphology and function. In particular, substrate geometry and rigidity in two dimensions has been shown to impact cell growth, death, differentiation, and motility. Knowledge of how these physical cues affect cell function in three dimensions is critical for successful development of novel regenerative therapies. In this work, the effect of discrete micromechanical cues in three-dimensional (3D) system on cell proliferation, gene expression, and extracellular matrix synthesis was investigated. Poly(ethylene glycol) dimethacrylate hydrogel microrods were fabricated using photolithography and suspended in gel to create a 3D culture with microscale cues of defined mechanical properties in the physiological range (2-50 kPa). These microrods significantly affected fibroblast proliferation, matrix production, and gene expression. Cultures with stiff microrods reduced fibroblast proliferation and downregulated expression of key extracellular matrix proteins involved in scar tissue formation. In addition, the contractility marker alpha smooth muscle actin and adhesion molecule integrin alpha3 were also significantly downregulated. Cultures with soft microrods had no significant difference on fibroblast proliferation and expression of Cyclin D1, alpha smooth muscle actin, and integrin alpha3 compared to cultures with no microrods. Here, we present a new platform of potentially injectable microrods with tunable elasticity; in addition, we show that cell proliferation and gene expression are influenced by discrete physical cues in 3D.
Collapse
Affiliation(s)
- Perla Ayala
- Joint Graduate Group in Bioengineering, University of California San Francisco-University of California Berkeley, San Francisco, California, USA
| | | | | |
Collapse
|
43
|
Hayes JS, Czekanska EM, Richards RG. The Cell–Surface Interaction. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2011; 126:1-31. [DOI: 10.1007/10_2011_110] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Abstract
Biophysical cues encoded in the extracellular matrix (ECM) are increasingly being explored to control cell behavior in tissue engineering applications. Recently, we showed that cell adhesion to microtopographical structures (“micropegs”) can suppress proliferation in a manner that may be blunted by inhibiting cellular contractility, suggesting that this effect is related to altered cell-scaffold mechanotransduction. We now directly investigate this possibility at the microscale through a combination of live-cell imaging, single-cell mechanics methods, and analysis of gene expression. Using time-lapse imaging, we show that when cells break adhesive contacts with micropegs, they form F-actin-filled tethers that extend and then rupture at a maximum, critical length that is greater than trailing-edge tethers observed on topographically flat substrates. This critical tether length depends on myosin activation, with inhibition of Rho-associated kinase abolishing topography-dependent differences in tether length. Using cellular de-adhesion and atomic force microscopy indentation measurements, we show that the micropegs enhance cell-scaffold adhesive interactions without changing whole-cell elasticity. Moreover, micropeg adhesion increases expression of specific mechanotransductive genes, including RhoA GTPase and myosin heavy chain II, and, in myoblasts, the functional marker connexin 43. Together, our data support a model in which microtopographical cues alter the local mechanical microenvironment of cells by modulating adhesion and adhesion-dependent mechanotransductive signaling.
Collapse
|
45
|
Kumar S. Cell-matrix mechanobiology: applications to brain tumors and design of tissue engineering scaffolds. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2010; 2009:3350-2. [PMID: 19964076 DOI: 10.1109/iembs.2009.5333188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It is becoming increasingly clear that mechanical and other biophysical signals from the extracellular matrix (ECM) can powerfully influence a wide variety of fundamental cell behaviors, including proliferation, differentiation, death, and motility. This concept has significant implications both for understanding the pathophysiology of disease and the design of biointerfaces found in cellular microdevices and tissue engineering platforms. Here we briefly review recent progress from our laboratory in investigating the role of ECM-derived mechanical signals in the specific context of two systems: The growth and spread of malignant brain tumors and the design of microscale cardiac tissue engineering systems. In both cases, mechanical signals encoded in the ECM govern motility, mechanics, and/or proliferation in profound and unexpected ways and rely upon the cell's reciprocal ability to generate contractile force through myosin and its molecular regulators.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
46
|
Bhana B, Iyer RK, Chen WLK, Zhao R, Sider KL, Likhitpanichkul M, Simmons CA, Radisic M. Influence of substrate stiffness on the phenotype of heart cells. Biotechnol Bioeng 2010; 105:1148-60. [PMID: 20014437 DOI: 10.1002/bit.22647] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adult cardiomyocytes (CM) retain little capacity to regenerate, which motivates efforts to engineer heart tissues that can emulate the functional and mechanical properties of native myocardium. Although the effects of matrix stiffness on individual CM have been explored, less attention was devoted to studies at the monolayer and the tissue level. The purpose of this study was to characterize the influence of substrate mechanical stiffness on the heart cell phenotype and functional properties. Neonatal rat heart cells were seeded onto collagen-coated polyacrylamide (PA) substrates with Young's moduli of 3, 22, 50, and 144 kPa. Collagen-coated glass coverslips without PA represented surfaces with effectively "infinite" stiffness. The local elastic modulus of native neonatal rat heart tissue was measured to range from 4.0 to 11.4 kPa (mean value of 6.8 kPa) and for native adult rat heart tissue from 11.9 to 46.2 kPa (mean value of 25.6 kPa), motivating our choice of the above PA gel stiffness. Overall, by 120 h of cultivation, the lowest stiffness PA substrates (3 kPa) exhibited the lowest excitation threshold (ET; 3.5 +/- 0.3 V/cm), increased troponin I staining (52% positively stained area) but reduced cell density, force of contraction (0.18 +/- 0.1 mN/mm(2)), and cell elongation (aspect ratio = 1.3-1.4). Higher stiffness (144 kPa) PA substrates exhibited reduced troponin I staining (30% positively stained area), increased fibroblast density (70% positively stained area), and poor electrical excitability. Intermediate stiffness PA substrates of stiffness comparable to the native adult rat myocardium (22-50 kPa) were found to be optimal for heart cell morphology and function, with superior elongation (aspect ratio > 4.3), reasonable ET (ranging from 3.95 +/- 0.8 to 4.4 +/- 0.7 V/cm), high contractile force development (ranging from 0.52 +/- 0.2 to 1.60 +/- 0.6 mN/mm(2)), and well-developed striations, all consistent with a differentiated phenotype.
Collapse
Affiliation(s)
- Bashir Bhana
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 164 College Street, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Mechanical stress-induced sarcomere assembly for cardiac muscle growth in length and width. J Mol Cell Cardiol 2010; 48:817-23. [PMID: 20188736 DOI: 10.1016/j.yjmcc.2010.02.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 02/15/2010] [Accepted: 02/17/2010] [Indexed: 12/27/2022]
Abstract
A ventricular myocyte experiences changes in length and load during every beat of the heart and has the ability to remodel cell shape to maintain cardiac performance. Specifically, myocytes elongate in response to increased diastolic strain by adding sarcomeres in series, and they thicken in response to continued systolic stress by adding filaments in parallel. Myocytes do this while still keeping the resting sarcomere length close to its optimal value at the peak of the length-tension curve. This review focuses on the little understood mechanisms by which direction of growth is matched in a physiologically appropriate direction. We propose that the direction of strain is detected by differential phosphorylation of proteins in the costamere, which then transmit signaling to the Z-disc for parallel or series addition of thin filaments regulated via the actin capping processes. In this review, we link mechanotransduction to the molecular mechanisms for regulation of myocyte length and width.
Collapse
|
48
|
Scharnagl N, Lee S, Hiebl B, Sisson A, Lendlein A. Design principles for polymers as substratum for adherent cells. ACTA ACUST UNITED AC 2010. [DOI: 10.1039/c0jm00997k] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
49
|
Cell culture on MEMS platforms: a review. Int J Mol Sci 2009; 10:5411-5441. [PMID: 20054478 PMCID: PMC2802002 DOI: 10.3390/ijms10125411] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 12/13/2009] [Accepted: 12/16/2009] [Indexed: 01/09/2023] Open
Abstract
Microfabricated systems provide an excellent platform for the culture of cells, and are an extremely useful tool for the investigation of cellular responses to various stimuli. Advantages offered over traditional methods include cost-effectiveness, controllability, low volume, high resolution, and sensitivity. Both biocompatible and bio-incompatible materials have been developed for use in these applications. Biocompatible materials such as PMMA or PLGA can be used directly for cell culture. However, for bio-incompatible materials such as silicon or PDMS, additional steps need to be taken to render these materials more suitable for cell adhesion and maintenance. This review describes multiple surface modification strategies to improve the biocompatibility of MEMS materials. Basic concepts of cell-biomaterial interactions, such as protein adsorption and cell adhesion are covered. Finally, the applications of these MEMS materials in Tissue Engineering are presented.
Collapse
|
50
|
Biehl JK, Yamanaka S, Desai TA, Boheler KR, Russell B. Proliferation of mouse embryonic stem cell progeny and the spontaneous contractile activity of cardiomyocytes are affected by microtopography. Dev Dyn 2009; 238:1964-73. [PMID: 19618471 DOI: 10.1002/dvdy.22030] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The niche in which stem cells reside and differentiate is a complex physicochemical microenvironment that regulates cell function. The role played by three-dimensional physical contours was studied on cell progeny derived from mouse embryonic stem cells using microtopographies created on PDMS (poly-dimethyl-siloxane) membranes. While markers of differentiation were not affected, the proliferation of heterogeneous mouse embryonic stem cell-derived progeny was attenuated by 15 microm-, but not 5 microm-high microprojections. This reduction was reversed by Rho kinase and myosin light chain kinase inhibition, which diminishes the tension generating ability of stress fibers. Purified cardiomyocytes derived from embryonic stem cells also showed significant blunting of proliferation and increased beating rates compared with cells grown on flat substrates. Thus, proliferation of stem cell-derived progeny appears to be regulated by microtopography through tension-generation of contractility in the third-dimension. These results emphasize the importance of topographic cues in the modulation of stem cell progeny behavior.
Collapse
Affiliation(s)
- Jesse K Biehl
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60612-7342, USA
| | | | | | | | | |
Collapse
|