1
|
Bildyug N. Inhibition of Integrin-Associated Kinases FAK and ILK on the In Vitro Model of Skin Wound Healing. Appl Biochem Biotechnol 2024; 196:5604-5615. [PMID: 38165590 DOI: 10.1007/s12010-023-04842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/04/2024]
Abstract
Dermal fibroblasts are essential cells of skin tissue responsible for its normal functioning. In skin wounds, the differentiation of resident fibroblasts into myofibroblasts occurs, which is accompanied by the rearrangement of actin cytoskeleton with the expression of alpha-smooth muscle actin. This transformation is a prerequisite for a successful wound healing. At the same time, different studies indicate that extracellular matrix may be involved in regulation of this process. Since the connection between cells and matrix is provided by transmembrane integrin receptors, this work was aimed at studying the dynamics of signaling pathways associated with integrins on an in vitro model of wound healing using human skin fibroblasts. It was shown that the healing of simulated wound was accompanied by a change in the level of integrins as well as integrin-associated kinases ILK (integrin-linked kinase) and FAK (focal adhesion kinase). Pharmacological inhibition of ILK and FAK caused the suppression of p38 and Akt which proteins are involved in regulation of the actin cytoskeleton. Moreover, it resulted in an inefficient wound closure in vitro. The results of this study support the involvement of integrin-associated kinases in regulation of fibroblast-to-myofibroblast transition during wound healing.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology Russian Academy of Sciences, Centre for Cell Technologies, Tikhoretsky ave. 4, 194064, Saint Petersburg, Russia.
| |
Collapse
|
2
|
Cao S, Buchholz KS, Tan P, Stowe JC, Wang A, Fowler A, Knaus KR, Khalilimeybodi A, Zambon AC, Omens JH, Saucerman JJ, McCulloch AD. Differential sensitivity to longitudinal and transverse stretch mediates transcriptional responses in mouse neonatal ventricular myocytes. Am J Physiol Heart Circ Physiol 2024; 326:H370-H384. [PMID: 38063811 PMCID: PMC11245882 DOI: 10.1152/ajpheart.00562.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
To identify how cardiomyocyte mechanosensitive signaling pathways are regulated by anisotropic stretch, micropatterned mouse neonatal cardiomyocytes were stretched primarily longitudinally or transversely to the myofiber axis. Four hours of static, longitudinal stretch induced differential expression of 557 genes, compared with 30 induced by transverse stretch, measured using RNA-seq. A logic-based ordinary differential equation model of the cardiac myocyte mechanosignaling network, extended to include the transcriptional regulation and expression of 784 genes, correctly predicted measured expression changes due to anisotropic stretch with 69% accuracy. The model also predicted published transcriptional responses to mechanical load in vitro or in vivo with 63-91% accuracy. The observed differences between transverse and longitudinal stretch responses were not explained by differential activation of specific pathways but rather by an approximately twofold greater sensitivity to longitudinal stretch than transverse stretch. In vitro experiments confirmed model predictions that stretch-induced gene expression is more sensitive to angiotensin II and endothelin-1, via RhoA and MAP kinases, than to the three membrane ion channels upstream of calcium signaling in the network. Quantitative cardiomyocyte gene expression differs substantially with the axis of maximum principal stretch relative to the myofilament axis, but this difference is due primarily to differences in stretch sensitivity rather than to selective activation of mechanosignaling pathways.NEW & NOTEWORTHY Anisotropic stretch applied to micropatterned neonatal mouse ventricular myocytes induced markedly greater acute transcriptional responses when the major axis of stretch was parallel to the myofilament axis than when it was transverse. Analysis with a novel quantitative network model of mechanoregulated cardiomyocyte gene expression suggests that this difference is explained by higher cell sensitivity to longitudinal loading than transverse loading than by the activation of differential signaling pathways.
Collapse
Affiliation(s)
- Shulin Cao
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Kyle S Buchholz
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Philip Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Jennifer C Stowe
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Ariel Wang
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Annabelle Fowler
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Katherine R Knaus
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California, United States
| | - Alexander C Zambon
- Department of Biopharmaceutical Sciences, Keck Graduate Institute, Claremont, California, United States
| | - Jeffrey H Omens
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
- Department of Medicine, University of California San Diego, La Jolla, California, United States
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States
- Department of Medicine, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
3
|
Claridge B, Drack A, Pinto AR, Greening DW. Defining cardiac fibrosis complexity and regulation towards therapeutic development. CLINICAL AND TRANSLATIONAL DISCOVERY 2023; 3. [DOI: 10.1002/ctd2.163] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2025]
Abstract
AbstractCardiac fibrosis is insidious, accelerating cardiovascular diseases, heart failure, and death. With a notable lack of effective therapies, advances in both understanding and targeted treatment of fibrosis are urgently needed. Remodelling of the extracellular matrix alters the biomechanical and biochemical cardiac structure and function, disrupting cell‐matrix interactions and exacerbating pathogenesis to ultimately impair cardiac function. Attempts at clinical fibrotic reduction have been fruitless, constrained by an understanding which severely underestimates its dynamic complexity and regulation. Integration of single‐cell sequencing and quantitative proteomics has provided new insights into cardiac fibrosis, including reparative or maladaptive processes, spatiotemporal changes and fibroblast heterogeneity. Further studies have revealed microenvironmental and intercellular signalling mechanisms (including soluble mediators and extracellular vesicles), and intracellular regulators including post‐translational/epigenetic modifications, RNA binding proteins, and non‐coding RNAs. This understanding of novel disease processes and molecular targets has supported the development of innovative therapeutic strategies. Indeed, targeted modulation of cellular heterogeneity, microenvironmental signalling, and intracellular regulation offer promising pre‐clinical therapeutic leads. Clinical development will require further advances in our mechanistic understanding of cardiac fibrosis and dissection of the molecular basis for fibrotic remodelling. This review provides an overview of the complexities of cardiac fibrosis, emerging regulatory mechanisms and therapeutic strategies, and highlights knowledge gaps and opportunities for further investigation towards therapeutic/clinical translation.
Collapse
Affiliation(s)
- Bethany Claridge
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment La Trobe University Melbourne Australia
| | - Auriane Drack
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment La Trobe University Melbourne Australia
| | - Alexander R. Pinto
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute Melbourne Australia
- Baker Department of Cardiovascular Research Translation and Implementation La Trobe University Melbourne Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment La Trobe University Melbourne Australia
- Baker Department of Cardiometabolic Health University of Melbourne Melbourne Australia
- Central Clinical School Monash University Melbourne Australia
| |
Collapse
|
4
|
Manso AM, Romaine A, Christensen G, Ross RS. Integrins in Cardiac Form, Function, and Disease. BIOLOGY OF EXTRACELLULAR MATRIX 2023:135-183. [DOI: 10.1007/978-3-031-23781-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Singh S, Winkelstein BA. Inhibiting the β1integrin subunit increases the strain threshold for neuronal dysfunction under tensile loading in collagen gels mimicking innervated ligaments. Biomech Model Mechanobiol 2022; 21:885-898. [DOI: 10.1007/s10237-022-01565-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 02/13/2022] [Indexed: 11/28/2022]
|
6
|
Regulation of collagen deposition in the trout heart during thermal acclimation. Curr Res Physiol 2022; 5:99-108. [PMID: 35243359 PMCID: PMC8857596 DOI: 10.1016/j.crphys.2022.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022] Open
Abstract
The passive mechanical properties of the vertebrate heart are controlled in part by the composition of the extracellular matrix (ECM). Changes in the ECM, caused by increased blood pressure, injury or disease can affect the capacity of the heart to fill with blood during diastole. In mammalian species, cardiac fibrosis caused by an increase in collagen in the ECM, leads to a loss of heart function and these changes in composition are considered to be permanent. Recent work has demonstrated that the cardiac ventricle of some fish species have the capacity to both increase and decrease collagen content in response to thermal acclimation. It is thought that these changes in collagen content help maintain ventricle function over seasonal changes in environmental temperatures. This current work reviews the cellular mechanisms responsible for regulating collagen deposition in the mammalian heart and proposes a cellular pathway by which a change in temperature can affect the collagen content of the fish ventricle through mechanotransduction. This work specifically focuses on the role of transforming growth factor β1, MAPK signaling pathways, and biomechanical stretch in regulating collagen content in the fish ventricle. It is hoped that this work increases the appreciation of the use of comparative models to gain insight into phenomenon with biomedical relevance.
Collapse
|
7
|
Bildyug N. Integrins in cardiac hypertrophy: lessons learned from culture systems. ESC Heart Fail 2021; 8:3634-3642. [PMID: 34232557 PMCID: PMC8497369 DOI: 10.1002/ehf2.13497] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/16/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Heart growth and pathological changes are accompanied by extracellular matrix‐dependent alterations in integrins and integrin‐associated proteins, suggesting their role in heart development and disease. Most of our knowledge on the involvement of integrins in heart pathology is provided by the in vivo experiments, including cardiac hypertrophy models. However, in vivo studies are limited by the complex organization of heart tissue and fail to discern cell types and particular integrins implicated in hypertrophic signalling. This problem is being addressed by isolated cardiomyocyte primary cultures, which have been successfully used in different in vitro disease models. This review aimed to analyse the general approaches to studying integrins and integrin‐associated signalling pathways in cardiac hypertrophy focusing on the in vitro systems. The lessons learned from culture experiments on the models of hypertrophy induced by stretch, stimulating factors, and/or extracellular matrix components are summarized, demonstrating the major involvement of integrin‐mediated signalling in cardiac hypertrophic response and its apparent crosstalk with signal pathways induced by stretch or hypertrophy stimulating factors. The benefits and perspectives of using cardiomyocyte primary culture as a hypertrophy model are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, 194064, Russia
| |
Collapse
|
8
|
ERK1/2: An Integrator of Signals That Alters Cardiac Homeostasis and Growth. BIOLOGY 2021; 10:biology10040346. [PMID: 33923899 PMCID: PMC8072600 DOI: 10.3390/biology10040346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/24/2022]
Abstract
Integration of cellular responses to extracellular cues is essential for cell survival and adaptation to stress. Extracellular signal-regulated kinase (ERK) 1 and 2 serve an evolutionarily conserved role for intracellular signal transduction that proved critical for cardiomyocyte homeostasis and cardiac stress responses. Considering the importance of ERK1/2 in the heart, understanding how these kinases operate in both normal and disease states is critical. Here, we review the complexity of upstream and downstream signals that govern ERK1/2-dependent regulation of cardiac structure and function. Particular emphasis is given to cardiomyocyte hypertrophy as an outcome of ERK1/2 activation regulation in the heart.
Collapse
|
9
|
Petersen AP, Cho N, Lyra-Leite DM, Santoso JW, Gupta D, Ariyasinghe NR, McCain ML. Regulation of calcium dynamics and propagation velocity by tissue microstructure in engineered strands of cardiac tissue. Integr Biol (Camb) 2021; 12:34-46. [PMID: 32118279 DOI: 10.1093/intbio/zyaa003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 01/27/2020] [Accepted: 01/30/2020] [Indexed: 01/13/2023]
Abstract
Disruptions to cardiac tissue microstructure are common in diseased or injured myocardium and are known substrates for arrhythmias. However, we have a relatively coarse understanding of the relationships between myocardial tissue microstructure, propagation velocity and calcium cycling, due largely to the limitations of conventional experimental tools. To address this, we used microcontact printing to engineer strands of cardiac tissue with eight different widths, quantified several structural and functional parameters and established correlation coefficients. As strand width increased, actin alignment, nuclei density, sarcomere index and cell aspect ratio decreased with unique trends. The propagation velocity of calcium waves decreased and the rise time of calcium transients increased with increasing strand width. The decay time constant of calcium transients decreased and then slightly increased with increasing strand width. Based on correlation coefficients, actin alignment was the strongest predictor of propagation velocity and calcium transient rise time. Sarcomere index and cell aspect ratio were also strongly correlated with propagation velocity. Actin alignment, sarcomere index and cell aspect ratio were all weak predictors of the calcium transient decay time constant. We also measured the expression of several genes relevant to propagation and calcium cycling and found higher expression of the genes that encode for connexin 43 (Cx43) and a subunit of L-type calcium channels in thin strands compared to isotropic tissues. Together, these results suggest that thinner strands have higher values of propagation velocity and calcium transient rise time due to a combination of favorable tissue microstructure and enhanced expression of genes for Cx43 and L-type calcium channels. These data are important for defining how microstructural features regulate intercellular and intracellular calcium handling, which is needed to understand mechanisms of propagation in physiological situations and arrhythmogenesis in pathological situations.
Collapse
Affiliation(s)
- Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey W Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Ho WJ, Jaffee EM, Zheng L. The tumour microenvironment in pancreatic cancer - clinical challenges and opportunities. Nat Rev Clin Oncol 2020; 17:527-540. [PMID: 32398706 PMCID: PMC7442729 DOI: 10.1038/s41571-020-0363-5] [Citation(s) in RCA: 665] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
Metastatic pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumours despite the use of multi-agent conventional chemotherapy regimens. Such poor outcomes have fuelled ongoing efforts to exploit the tumour microenvironment (TME) for therapy, but strategies aimed at deconstructing the surrounding desmoplastic stroma and targeting the immunosuppressive pathways have largely failed. In fact, evidence has now shown that the stroma is multi-faceted, which illustrates the complexity of exploring features of the TME as isolated targets. In this Review, we describe ways in which the PDAC microenvironment has been targeted and note the current understanding of the clinical outcomes that have unexpectedly contradicted preclinical observations. We also consider the more sophisticated therapeutic strategies under active investigation - multi-modal treatment approaches and exploitation of biologically integrated targets - which aim to remodel the TME against PDAC.
Collapse
Affiliation(s)
- Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
11
|
Johnston EF, Gillis TE. Short-term cyclical stretch phosphorylates p38 and ERK1/2 MAPKs in cultured fibroblasts from the hearts of rainbow trout, Oncorhynchus mykiss. Biol Open 2020; 9:bio.049296. [PMID: 31862862 PMCID: PMC6994941 DOI: 10.1242/bio.049296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The form and function of the rainbow trout heart can remodel in response to various stressors including changes in environmental temperature and anemia. Previous studies have hypothesized that changes in biomechanical forces experienced by the trout myocardium as result of such physiological stressors could play a role in triggering the remodeling response. However, there has been no work examining the influence of biomechanical forces on the trout myocardium or of the cellular signals that would translate such a stimuli into a biological response. In this study, we test the hypothesis that the application of biomechanical forces to trout cardiac fibroblasts activate the cell signaling pathways associated with cardiac remodeling. This was done by cyclically stretching cardiac fibroblasts to 10% equibiaxial deformation at 0.33 Hz and quantifying the activation of the p38-JNK-ERK mitogen activated protein kinase (MAPK) pathway. After 20 min, p38 MAPK phosphorylation was elevated by 4.2-fold compared to control cells (P<0.05) and after 24 h of stretch, p38 MAPK phosphorylation remained elevated and extracellular-regulated kinase 1/2 was phosphorylated by 2.4-fold compared to control (P<0.05). Together, these results indicate that mechanotransductive pathways are active in cardiac fibroblasts, and lead to the activation of cell signaling pathways involved in cardiac remodeling.
Collapse
Affiliation(s)
- Elizabeth F Johnston
- Department of Integrative Biology, University of Guelph, Ontario, Canada, N1G 2W1
| | - Todd E Gillis
- Department of Integrative Biology, University of Guelph, Ontario, Canada, N1G 2W1
| |
Collapse
|
12
|
Bildyug N. Extracellular Matrix in Regulation of Contractile System in Cardiomyocytes. Int J Mol Sci 2019; 20:E5054. [PMID: 31614676 PMCID: PMC6834325 DOI: 10.3390/ijms20205054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The contractile apparatus of cardiomyocytes is considered to be a stable system. However, it undergoes strong rearrangements during heart development as cells progress from their non-muscle precursors. Long-term culturing of mature cardiomyocytes is also accompanied by the reorganization of their contractile apparatus with the conversion of typical myofibrils into structures of non-muscle type. Processes of heart development as well as cell adaptation to culture conditions in cardiomyocytes both involve extracellular matrix changes, which appear to be crucial for the maturation of contractile apparatus. The aim of this review is to analyze the role of extracellular matrix in the regulation of contractile system dynamics in cardiomyocytes. Here, the remodeling of actin contractile structures and the expression of actin isoforms in cardiomyocytes during differentiation and adaptation to the culture system are described along with the extracellular matrix alterations. The data supporting the regulation of actin dynamics by extracellular matrix are highlighted and the possible mechanisms of such regulation are discussed.
Collapse
Affiliation(s)
- Natalya Bildyug
- Institute of Cytology, Russian Academy of Sciences, St-Petersburg 194064, Russia.
| |
Collapse
|
13
|
Zhang Y, Wang F, Bao L, Li J, Shi Z, Wang J. Cyclic hydrostatic compress force regulates apoptosis of meniscus fibrochondrocytes via integrin α5β1. Physiol Res 2019; 68:639-649. [DOI: 10.33549/physiolres.934088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Meniscus is a semilunar fibrocartilaginous tissue, serving important roles in load buffering, stability, lubrication, proprioception, and nutrition of the knee joint. The degeneration and damage of meniscus has been proved to be a risk factor of knee osteoarthritis. Mechanical stimulus is a critical factor of the development, maintenance and repair of the meniscus fibrochondrocytes. However, the mechanism of the mechano-transduction process remains elusive. Here we reported that cyclic hydrostatic compress force (CHCF) treatment promotes proliferation and inhibits apoptosis of the isolated primary meniscus fibrochondrocytes (PMFs), via upregulating the expression level of integrin α5β1. Consequently, increased phosphorylated-ERK1/2 and phosphorylated-PI3K, and decreased caspase-3 were detected. These effects of CHCF treatment can be abolished by integrin α5β1 inhibitor or specific siRNA transfection. These data indicate that CHCF regulates apoptosis of PMFs via integrin α5β1-FAK-PI3K/ERK pathway, which may be an important candidate approach during meniscus degeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - J. Wang
- Department of orthopedic surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Xie S, Jiang X, Wang R, Xie S, Hua Y, Zhou S, Yang Y, Zhang J. Low-intensity pulsed ultrasound promotes the proliferation of human bone mesenchymal stem cells by activating PI3K/AKt signaling pathways. J Cell Biochem 2019; 120:15823-15833. [PMID: 31090943 DOI: 10.1002/jcb.28853] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 12/27/2022]
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a promising therapy that is widely used in clinical applications and fundamental research. Previous research has shown that LIPUS exposure has a positive effect on stem cell proliferation. However, the impact of LIPUS exposure on human bone marrow mesenchymal stem cells (hBMSCs) remains unknown. In our study, the effect and mechanism of LIPUS exposure on the proliferation of hBMSCs were investigated, and the optimal parameters of LIPUS were determined. hBMSCs were obtained and identified by flow cytometry, and the proliferation of hBMSCs was measured using the Cell Counting Kit-8 assay to determine cell cycle and cell count. Expression levels of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKt) pathway proteins and cyclin D1 were determined by western blot analysis. Next, hBMSCs were successfully cultured and identified as multipotent mesenchymal stem cells. We found that LIPUS could promote the proliferation of hBMSCs when the exposure time was 5 or 10 minutes per day. Furthermore, 50 or 60 mW/cm2 LIPUS had a more significant effect on cell proliferation, but if cells were irradiated by LIPUS for 20 minutes once a day, an intensity of at least 50 mW/cm2 could markedly inhibit cell growth. Cell cycle analysis demonstrated that LIPUS treatment drives cells to enter S and G2/M phases from the G0/G1 phase. LIPUS exposure increased phosphorylation of PI3K/AKt and significantly upregulated expression of cyclin D1. However, these effects were inhibited when cells were treated with PI3K inhibitor (LY294002), which in turn reduced LIPUS-mediated proliferation of hBMSCs. These results suggest that LIPUS exposure may be involved in the proliferation of hBMSCs via activation of the PI3K/AKt signaling pathway and high expression of cyclin D1, and the intensity of 50 or 60 mW/cm2 and exposure time of 5 minutes were determined to be the optimal parameters for LIPUS exposure.
Collapse
Affiliation(s)
- Shucai Xie
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Xili Jiang
- Department of Radiology, The Second People's Hospital of Hunan Province/Brain Hospital of Hunan Province, Changsha, Hunan, People's Republic of China
| | - Rui Wang
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Shaowei Xie
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Yongyong Hua
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Shuai Zhou
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Yijun Yang
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Jianquan Zhang
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
15
|
Dowrick JM, Tran K, Loiselle DS, Nielsen PMF, Taberner AJ, Han J, Ward M. The slow force response to stretch: Controversy and contradictions. Acta Physiol (Oxf) 2019; 226:e13250. [PMID: 30614655 DOI: 10.1111/apha.13250] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/20/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
When exposed to an abrupt stretch, cardiac muscle exhibits biphasic active force enhancement. The initial, instantaneous, force enhancement is well explained by the Frank-Starling mechanism. However, the cellular mechanisms associated with the second, slower phase remain contentious. This review explores hypotheses regarding this "slow force response" with the intention of clarifying some apparent contradictions in the literature. The review is partitioned into three sections. The first section considers pathways that modify the intracellular calcium handling to address the role of the sarcoplasmic reticulum in the mechanism underlying the slow force response. The second section focuses on extracellular calcium fluxes and explores the identity and contribution of the stretch-activated, non-specific, cation channels as well as signalling cascades associated with G-protein coupled receptors. The final section introduces promising candidates for the mechanosensor(s) responsible for detecting the stretch perturbation.
Collapse
Affiliation(s)
- Jarrah M. Dowrick
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Kenneth Tran
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Denis S. Loiselle
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Physiology University of Auckland Auckland New Zealand
| | - Poul M. F. Nielsen
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - Andrew J. Taberner
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - June‐Chiew Han
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Marie‐Louise Ward
- Department of Physiology University of Auckland Auckland New Zealand
| |
Collapse
|
16
|
Fujita M, Sato-Shigeta M, Mori H, Iwasa A, Kawai N, Hassan AH, Tanaka E. Protective Effects of Low-Intensity Pulsed Ultrasound on Mandibular Condylar Cartilage Exposed to Mechanical Overloading. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:944-953. [PMID: 30732913 DOI: 10.1016/j.ultrasmedbio.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 06/09/2023]
Abstract
The aim of this study was to assess the effect of low-intensity pulsed ultrasound (LIPUS) application on rat temporomandibular joints (TMJs) with early-stage of osteoarthritis-like conditions induced by mechanical overloading. Fifteen-week-old male Wistar rats were divided into two experimental groups and a control group (n = 10 each). Both TMJs of all rats in one experimental group were subjected to mechanical overloading for 5 d, and those in the other experimental group were exposed to LIPUS for 20 min/d after overloading. Condyles were assessed using micro-computed tomography, histology and histomorphometry. LIPUS treatment attenuated cartilage degeneration, decreased the number of osteoclastic cells and restored the expression of aggrecan after an initial decrease induced by mechanical overloading. These results indicate that LIPUS may have a protective effect on the early progression of TMJ osteoarthritis.
Collapse
Affiliation(s)
- Mutsumi Fujita
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Oral Sciences, Tokushima, Japan.
| | - Minami Sato-Shigeta
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Hospital, Tokushima, Japan
| | - Hiroki Mori
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Hospital, Tokushima, Japan
| | - Akihiko Iwasa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Nobuhiko Kawai
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Ali H Hassan
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
17
|
Franchi MV, Ruoss S, Valdivieso P, Mitchell KW, Smith K, Atherton PJ, Narici MV, Flück M. Regional regulation of focal adhesion kinase after concentric and eccentric loading is related to remodelling of human skeletal muscle. Acta Physiol (Oxf) 2018; 223:e13056. [PMID: 29438584 DOI: 10.1111/apha.13056] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/31/2018] [Accepted: 02/07/2018] [Indexed: 12/31/2022]
Abstract
AIMS We assessed focal adhesion kinase (FAK) response to concentric (CON) vs eccentric (ECC) resistance training (RT) at two vastus lateralis (VL) sites, and the relationships between FAK, muscle protein synthesis (MPS) and morphological remodelling. METHODS Six young males trained both legs unilaterally 3 times/week for 8 weeks; one leg performed CON RT, the contralateral performed ECC RT. Muscle biopsies were collected after training from VL mid-belly (MID) and distal (distal) sites at 0, 4, 8 weeks. Focal adhesion kinase content and activation were evaluated by immunoblotting. MPS was assessed by deuterium oxide tracer; morphological adaptations were evaluated by ultrasound and DXA. RESULTS pY397-FAK 8 weeks levels were ~4-fold greater after ECC at the distal site compared to CON (P < .05); pY397FAK to total FAK ratio was greater in ECC vs CON at 4 (~2.2-fold, P < .05) and 8 weeks (~9-fold, P < .001) at the distal site. Meta-vinculin was found transiently increased at 4 weeks at the distal site only after ECC RT. ECC presented greater fascicle length (Lf) increases (10.5% vs 4%), whereas CON showed greater in pennation angle (PA) changes (12.3% vs 2.1%). MPS did not differ between exercise types or muscle sites at all time points. distal pY397-FAK and pY397-FAK/FAK values correlated to changes in Lf at 8 weeks (r = .76, P < .01 and r = .66, P < .05 respectively). CONCLUSION Focal adhesion kinase phosphorylation was greater at 8 weeks after ECC RT and was muscle region-specific. FAK activity correlated to contraction-dependent architectural remodelling, suggesting a potential role of FAK in orienting muscle structural changes in response to distinct mechanical stimuli.
Collapse
Affiliation(s)
- M V Franchi
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - S Ruoss
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - P Valdivieso
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - K W Mitchell
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - K Smith
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - P J Atherton
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
| | - M V Narici
- MRC-ARUK Centre for Musculoskeletal Ageing, Royal Derby Hospital, University of Nottingham, Derby, UK
- Department of Biomedical Sciences, Institute of Physiology, University of Padua, Padua, Italy
| | - M Flück
- Laboratory for Muscle Plasticity, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Biological Aspect of Pathophysiology for Frozen Shoulder. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7274517. [PMID: 29992159 PMCID: PMC5994312 DOI: 10.1155/2018/7274517] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/02/2018] [Accepted: 04/22/2018] [Indexed: 12/27/2022]
Abstract
It is fairly well understood that frozen shoulder involves several stages, which reflect the series of process from capsular inflammation and fibrosis to spontaneous resolution of this fibrosis. However, the underlying pathophysiologic process remains poorly determined. For this reason, management of frozen shoulder remains controversial. Determining the pathophysiological processes of frozen shoulder is a pivotal milestone in the development of novel treatment for patients with frozen shoulder. This article reviews what is known to date about the biological pathophysiology of frozen shoulder. Although articles for the pathophysiology of frozen shoulder provide inconsistent and inconclusive results, they have suggested both inflammation and fibrosis mediated by cytokines, growth factors, matrix metalloproteinases, and immune cells. Proinflammatory cytokines and growth factors released from immune cells control the action of fibroblast and matrix remodeling is regulated by the matrix metalloproteinases and their inhibitors. To improve our understanding of the disease continuum, better characterizing the biology of these processes at clearly defined stages will be needed. Further basic studies that use standardized protocols are required to more narrowly identify the role of cytokines, growth factors, matrix metalloproteinases, and immune cells. The results of these studies will provide needed clarity into the control mechanism of the pathogenesis of frozen shoulder and help identify new therapeutic targets for its treatment.
Collapse
|
19
|
Cardiomyocyte-specific deletion of GSK-3β leads to cardiac dysfunction in a diet induced obesity model. Int J Cardiol 2018; 259:145-152. [PMID: 29398139 DOI: 10.1016/j.ijcard.2018.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND RATIONALE Obesity, an independent risk factor for the development of myocardial diseases is a growing healthcare problem worldwide. It's well established that GSK-3β is critical to cardiac pathophysiology. However, the role cardiomyocyte (CM) GSK-3β in diet-induced cardiac dysfunction is unknown. METHODS CM-specific GSK-3β knockout (CM-GSK-3β-KO) and littermate controls (WT) mice were fed either a control diet (CD) or high-fat diet (HFD) for 55weeks. Cardiac function was assessed by transthoracic echocardiography. RESULTS At baseline, body weights and cardiac function were comparable between the WT and CM-GSK-3β-KOs. However, HFD-fed CM-GSK-3β-KO mice developed severe cardiac dysfunction. Consistently, both heart weight/tibia length and lung weight/tibia length were significantly elevated in the HFD-fed CM-GSK-3β-KO mice. The impaired cardiac function and adverse ventricular remodeling in the CM-GSK-3β-KOs were independent of body weight or the lean/fat mass composition as HFD-fed CM-GSK-3β-KO and controls demonstrated comparable body weight and body masses. At the molecular level, on a CD, CM-GSK-3α compensated for the loss of CM-GSK-3β, as evident by significantly reduced GSK-3αs21 phosphorylation (activation) resulting in a preserved canonical β-catenin ubiquitination pathway and cardiac function. However, this protective compensatory mechanism is lost with HFD, leading to excessive accumulation of β-catenin in HFD-fed CM-GSK-3β-KO hearts, resulting in adverse ventricular remodeling and cardiac dysfunction. CONCLUSION In summary, these results suggest that cardiac GSK-3β is crucial to protect against obesity-induced adverse ventricular remodeling and cardiac dysfunction.
Collapse
|
20
|
Enhanced expression and phosphorylation of Sirt7 activates smad2 and ERK signaling and promotes the cardiac fibrosis differentiation upon angiotensin-II stimulation. PLoS One 2017; 12:e0178530. [PMID: 28582407 PMCID: PMC5459426 DOI: 10.1371/journal.pone.0178530] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/15/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibroblasts (CFs) phenotypic conversion to myofibroblasts (MFs) represents a crucial event in cardiac fibrosis that leads to impaired cardiac function. However, regulation of this phenotypic transformation remains unclear. Here, we showed that sirtuin-7 (Sirt7) plays an important role in the regulation of MFs differentiation. Sirt7 expression and phosphorylation were upregulated in CFs upon angiotensin-II (Ang-II) stimulation. Sirt7 depletion by siRNA in CFs resulted in decreased cell proliferation and extracellular matrix (ECM) deposition. Further, examination of Sirt7-depleted CFs demonstrated significantly lower expression of α-smooth muscle actin (α-SMA), the classical marker of MFs differentiation, and decreased formation of focal adhesions. Moreover, overexpression of Sirt7 increased α-SMA expression in Ang-II treated CFs and exacerbated Ang-II-induced MFs differentiation. Moreover, Sirt7 depletion could largely reverse Ang-II induced increase of nuclear translocalization and activity of smad2 and extracellular regulated kinases (ERK) in CFs. Importantly, the increased differentiation of CFs to MFs was also abolished by smad2 siRNA or U0126. Our findings reveal a novel role of Sirt7 and its phosphorylation in the phenotypic conversion of CFs to MFs and might lead to the development of new therapeutic and prognostic tools for cardiac fibrosis.
Collapse
|
21
|
Park SW, Shin KC, Park HJ, Yoou SK, Park JY, Kang YS, Sung DJ, Kim JG, Park SH, Kim B, Cho H, Bae YM. Caveolar remodeling is a critical mechanotransduction mechanism of the stretch-induced L-type Ca 2+ channel activation in vascular myocytes. Pflugers Arch 2017; 469:829-842. [PMID: 28303387 DOI: 10.1007/s00424-017-1957-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 01/26/2017] [Accepted: 02/13/2017] [Indexed: 12/11/2022]
Abstract
Activation of L-type voltage-dependent Ca2+ channels (VDCCL) by membrane stretch contributes to many biological responses such as myogenic contraction of arteries. However, mechanism for the stretch-induced VDCCL activation is unclear. In this study, we examined the hypothesis that caveolar remodeling and its related signaling cascade contribute to the stretch-induced activation of VDCCL in rat mesenteric arterial smooth muscle cells. The VDCCL currents were recorded with nystatin-perforated or with conventional whole-cell patch-clamp technique. Hypotonic (~230 mOsm) swelling-induced membrane stretch reversibly increased the VDCCL currents. Electron microscope and confocal imaging analysis revealed that both hypotonic swelling and cholesterol depletion by methyl-β-cychlodextrin (MβCD) similarly disrupted the caveolae structure and translocated caveolin-1 (Cav-1) from membrane to cytosolic space. Accordingly, MβCD also increased VDCCL currents. Moreover, subsequent hypotonic swelling after MβCD treatment failed to increase the VDCCL currents further. Western blotting experiments revealed that hypotonic swelling phosphorylated Cav-1 and JNK. Inhibitors of tyrosine kinases (genistein) and JNK (SP00125) prevented the swelling-induced facilitation of VDCCL currents. Knockdown of Cav-1 by small interfering RNA blocked both the VDCCL current facilitation by stretch and the related phosphorylation of JNK. Taken together, the results suggest that membrane stretch is transduced to the facilitation of VDCCL currents via caveolar structure-dependent tyrosine phosphorylation of Cav-1 and subsequent activation of JNK in rat mesenteric arterial myocytes.
Collapse
Affiliation(s)
- Sang Woong Park
- Department of Emergency Medical Services, Eulji University, Seongnam, Gyeonggi-do, 461-713, South Korea
| | - Kyung Chul Shin
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Hyun Ji Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Soon-Kyu Yoou
- Department of Emergency Medical Services, Eulji University, Seongnam, Gyeonggi-do, 461-713, South Korea
| | - Jin-Yeon Park
- Colleage of Veterinary Medicine, Department of Biomedical Science & Technology (DBST), Konkuk University, Seoul, 143-701, South Korea
| | - Young-Sun Kang
- Colleage of Veterinary Medicine, Department of Biomedical Science & Technology (DBST), Konkuk University, Seoul, 143-701, South Korea
| | - Dong Jun Sung
- Division of Sport Science, College of Science and Technology, Konkuk University, Chungju, Chungbuk, 380-701, South Korea
| | - Jae Gon Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Seung Hwa Park
- Department of Anatomy, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - BoKyung Kim
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea
| | - Hana Cho
- Department of Physiology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, Chungbuk, 380-701, South Korea.
| |
Collapse
|
22
|
Wanjare M, Huang NF. Regulation of the microenvironment for cardiac tissue engineering. Regen Med 2017; 12:187-201. [PMID: 28244821 DOI: 10.2217/rme-2016-0132] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The microenvironment of myocardium plays an important role in the fate and function of cardiomyocytes (CMs). Cardiovascular tissue engineering strategies commonly utilize stem cell sources in conjunction with microenvironmental cues that often include biochemical, electrical, spatial and biomechanical factors. Microenvironmental stimulation of CMs, in addition to the incorporation of intercellular interactions from non-CMs, results in the generation of engineered cardiac constructs. Current studies suggest that use of these factors when engineering cardiac constructs improve cardiac function when implanted in vivo. In this review, we summarize the approaches to modulate biochemical, electrical, biomechanical and spatial factors to induce CM differentiation and their subsequent organization for cardiac tissue engineering application.
Collapse
Affiliation(s)
- Maureen Wanjare
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ngan F Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.,Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
23
|
Abstract
In this chapter, we discuss the manner through which the immune system regulates the cardiovascular system in health and disease. We define the cardiovascular system and elements of atherosclerotic disease, the main focus in this chapter. Herein we elaborate on the disease process that can result in myocardial infarction (heart attack), ischaemic stroke and peripheral arterial disease. We have discussed broadly the homeostatic mechanisms in place that help autoregulate the cardiovascular system including the vital role of cholesterol and lipid clearance as well as the role lipid homeostasis plays in cardiovascular disease in the context of atherosclerosis. We then elaborate on the role played by the immune system in this setting, namely, major players from the innate and adaptive immune system, as well as discussing in greater detail specifically the role played by monocytes and macrophages.This chapter should represent an overview of the role played by the immune system in cardiovascular homeostasis; however further reading of the references cited can expand the reader's knowledge of the detail, and we point readers to many excellent reviews which summarise individual immune systems and their role in cardiovascular disease.
Collapse
Affiliation(s)
- Mohammed Shamim Rahman
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, UK
| | - Kevin Woollard
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
24
|
Lü D, Li Z, Gao Y, Luo C, Zhang F, Zheng L, Wang J, Sun S, Long M. β1 integrin signaling in asymmetric migration of keratinocytes under mechanical stretch in a co-cultured wound repair model. Biomed Eng Online 2016; 15:130. [PMID: 28155694 PMCID: PMC5259898 DOI: 10.1186/s12938-016-0263-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Keratinocyte (KC) migration in re-epithelization is crucial in repairing injured skin. But the mechanisms of how mechanical stimuli regulate the migration of keratinocytes have been poorly understood. Methods Human immortalized keratinocyte HaCaT cells were co-cultured with skin fibroblasts on PDMS membranes and transferred to the static stretch device developed in-house for additional 6 day culture under mechanical stretch to mimic surface tension in skin. To detect the expression of proteins on different position at different time points and the effect of β1 integrin mechanotransduction on HaCaT migration, Immunofluorescence, Reverse transcription-polymerase chain reaction, Flow cytometry, Western blotting assays were applied. Results Mechanical receptor of β1 integrin that recognizes its ligand of collagen I was found to be strongly associated with migration of HaCaT cells since the knockdown of β1 integrin via RNA silence eliminated the key protein expression dynamically. Here the expression of vinculin was lower but that of Cdc42 was higher for the cells at outward edge than those at inward edge, respectively, supporting that the migration capability of keratinocytes is inversely correlated with the formation of focal adhesion complexes but positively related to the lamellipodia formation. This asymmetric expression feature was further confirmed by high or low expression of PI3K for outward- or inward-migrating cells. And ERK1/2 phosphorylation was up-regulated by mechanical stretch. Conclusion We reported here, a novel mechanotransduction signaling pathways were β1 integrin-dependent pattern of keratinocytes migration under static stretch in an in vitro co-culture model. These results provided an insight into underlying molecular mechanisms of keratinocyte migration under mechanical stimuli.
Collapse
Affiliation(s)
- Dongyuan Lü
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhan Li
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuxin Gao
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chunhua Luo
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Fan Zhang
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Lu Zheng
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiawen Wang
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Shujin Sun
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Mian Long
- Center of Biomechanics and Bioengineering, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China. .,Key Laboratory of Microgravity (National Microgravity Laboratory), Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China. .,Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
25
|
Ma D, Kou X, Jin J, Xu T, Wu M, Deng L, Fu L, Liu Y, Wu G, Lu H. Hydrostatic Compress Force Enhances the Viability and Decreases the Apoptosis of Condylar Chondrocytes through Integrin-FAK-ERK/PI3K Pathway. Int J Mol Sci 2016; 17:ijms17111847. [PMID: 27827993 PMCID: PMC5133847 DOI: 10.3390/ijms17111847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/27/2016] [Accepted: 10/31/2016] [Indexed: 01/23/2023] Open
Abstract
Reduced mechanical stimuli in many pathological cases, such as hemimastication and limited masticatory movements, can significantly affect the metabolic activity of mandibular condylar chondrocytes and the growth of mandibles. However, the molecular mechanisms for these phenomena remain unclear. In this study, we hypothesized that integrin-focal adhesion kinase (FAK)-ERK (extracellular signal-regulated kinase)/PI3K (phosphatidylinositol-3-kinase) signaling pathway mediated the cellular response of condylar chondrocytes to mechanical loading. Primary condylar chondrocytes were exposed to hydrostatic compressive forces (HCFs) of different magnitudes (0, 50, 100, 150, 200, and 250 kPa) for 2 h. We measured the viability, morphology, and apoptosis of the chondrocytes with different treatments as well as the gene, protein expression, and phosphorylation of mechanosensitivity-related molecules, such as integrin α2, integrin α5, integrin β1, FAK, ERK, and PI3K. HCFs could significantly increase the viability and surface area of condylar chondrocytes and decrease their apoptosis in a dose-dependent manner. HCF of 250 kPa resulted in a 1.51 ± 0.02-fold increase of cell viability and reduced the ratio of apoptotic cells from 18.10% ± 0.56% to 7.30% ± 1.43%. HCFs could significantly enhance the mRNA and protein expression of integrin α2, integrin α5, and integrin β1 in a dose-dependent manner, but not ERK1, ERK2, or PI3K. Instead, HCF could significantly increase phosphorylation levels of FAK, ERK1/2, and PI3K in a dose-dependent manner. Cilengitide, the potent integrin inhibitor, could dose-dependently block such effects of HCFs. HCFs enhances the viability and decreases the apoptosis of condylar chondrocytes through the integrin-FAK-ERK/PI3K pathway.
Collapse
Affiliation(s)
- Dandan Ma
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China.
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), VU University Amsterdam and University of Amsterdam, MOVE Research Institute, Gustav Mahlerlaan 3004, Amsterdam 1081LA, Nord-Holland, The Netherlands.
| | - Xiaoxing Kou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Jing Jin
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Taotao Xu
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China.
| | - Mengjie Wu
- Department of Orthodontics, Stomatology Hospital Affiliated to Zhejiang University, Hangzhou 310053, China.
| | - Liquan Deng
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Lusi Fu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yi Liu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), VU University Amsterdam and University of Amsterdam, MOVE Research Institute, Gustav Mahlerlaan 3004, Amsterdam 1081LA, Nord-Holland, The Netherlands.
| | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), VU University Amsterdam and University of Amsterdam, MOVE Research Institute, Gustav Mahlerlaan 3004, Amsterdam 1081LA, Nord-Holland, The Netherlands.
| | - Haiping Lu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
26
|
Chacar S, Farès N, Bois P, Faivre JF. Basic Signaling in Cardiac Fibroblasts. J Cell Physiol 2016; 232:725-730. [DOI: 10.1002/jcp.25624] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Stéphanie Chacar
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
- Laboratoire de recherche en Physiologie et Physiopathologie (LRPP); pôle technologie santé; Faculté de Médecine; Université Saint Joseph; Beyrouth Liban
| | - Nassim Farès
- Laboratoire de recherche en Physiologie et Physiopathologie (LRPP); pôle technologie santé; Faculté de Médecine; Université Saint Joseph; Beyrouth Liban
| | - Patrick Bois
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
| | - Jean-François Faivre
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM); Université de Poitiers; CNRS; Poitiers France
| |
Collapse
|
27
|
Yan Y, Sun H, Gong Y, Yan Z, Zhang X, Guo Y, Wang Y. Mechanical strain promotes osteoblastic differentiation through integrin-β1-mediated β-catenin signaling. Int J Mol Med 2016; 38:594-600. [DOI: 10.3892/ijmm.2016.2636] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 06/03/2016] [Indexed: 11/05/2022] Open
|
28
|
Abstract
Cells respond to their environment by relaying mechanical force into biochemical stimuli that activate intracellular signal transduction pathways. Subjecting cells to in vitro mechanical stretch can mimic cellular responses to changes in the rigidity of the extracellular matrix. Here we describe an in vitro model system that mimics stretch overload in vivo. In this stretch-mediated hypertrophy model, adult rat cardiomyocytes attached to laminin-coated flexible membranes are subjected to cyclic mechanical stretch at an extension level of 10 % at 30 cycles/min. At various time points VEGF secretion into the media is collected and quantitated.
Collapse
|
29
|
Liang Y, Sheikh F. Scaffold Proteins Regulating Extracellular Regulated Kinase Function in Cardiac Hypertrophy and Disease. Front Pharmacol 2016; 7:37. [PMID: 26973524 PMCID: PMC4770026 DOI: 10.3389/fphar.2016.00037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 02/11/2016] [Indexed: 01/07/2023] Open
Abstract
The mitogen activated protein kinase (MAPK)-extracellular regulated kinase 1/2 (ERK1/2) pathway is a central downstream signaling pathway that is activated in cardiac muscle cells during mechanical and agonist-mediated hypertrophy. Studies in genetic mouse models deficient in ERK-associated MAPK components pathway have further reinforced a direct role for this pathway in stress-induced cardiac hypertrophy and disease. However, more recent studies have highlighted that these signaling pathways may exert their regulatory functions in a more compartmentalized manner in cardiac muscle. Emerging data has uncovered specific MAPK scaffolding proteins that tether MAPK/ERK signaling specifically at the sarcomere and plasma membrane in cardiac muscle and show that deficiencies in these scaffolding proteins alter ERK activity and phosphorylation, which are then critical in altering the cardiac myocyte response to stress-induced hypertrophy and disease progression. In this review, we provide insights on ERK-associated scaffolding proteins regulating cardiac myofilament function and their impact on cardiac hypertrophy and disease.
Collapse
Affiliation(s)
- Yan Liang
- Department of Medicine, University of California-San Diego, La Jolla CA, USA
| | - Farah Sheikh
- Department of Medicine, University of California-San Diego, La Jolla CA, USA
| |
Collapse
|
30
|
Extracellular matrix-mediated cellular communication in the heart. J Mol Cell Cardiol 2016; 91:228-37. [PMID: 26778458 DOI: 10.1016/j.yjmcc.2016.01.011] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/10/2016] [Accepted: 01/11/2016] [Indexed: 01/13/2023]
Abstract
The extracellular matrix (ECM) is a complex and dynamic scaffold that maintains tissue structure and dynamics. However, the view of the ECM as an inert architectural support has been increasingly challenged. The ECM is a vibrant meshwork, a crucial organizer of cellular microenvironments. It plays a direct role in cellular interactions regulating cell growth, survival, spreading, proliferation, differentiation and migration through the intricate relationship among cellular and acellular tissue components. This complex interrelationship preserves cardiac function during homeostasis; however it is also responsible for pathologic remodeling following myocardial injury. Therefore, enhancing our understanding of this cross-talk may provide mechanistic insights into the pathogenesis of heart failure and suggest new approaches to novel, targeted pharmacologic therapies. This review explores the implications of ECM-cell interactions in myocardial cell behavior and cardiac function at baseline and following myocardial injury.
Collapse
|
31
|
Feng H, Gerilechaogetu F, Golden HB, Nizamutdinov D, Foster DM, Glaser SS, Dostal DE. p38α MAPK inhibits stretch-induced JNK activation in cardiac myocytes through MKP-1. Int J Cardiol 2016; 203:145-55. [DOI: 10.1016/j.ijcard.2015.10.109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/20/2015] [Accepted: 10/12/2015] [Indexed: 01/18/2023]
|
32
|
The role of endothelial mechanosensitive genes in atherosclerosis and omics approaches. Arch Biochem Biophys 2015; 591:111-31. [PMID: 26686737 DOI: 10.1016/j.abb.2015.11.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is the leading cause of morbidity and mortality in the U.S., and is a multifactorial disease that preferentially occurs in regions of the arterial tree exposed to disturbed blood flow. The detailed mechanisms by which d-flow induces atherosclerosis involve changes in the expression of genes, epigenetic patterns, and metabolites of multiple vascular cells, especially endothelial cells. This review presents an overview of endothelial mechanobiology and its relation to the pathogenesis of atherosclerosis with special reference to the anatomy of the artery and the underlying fluid mechanics, followed by a discussion of a variety of experimental models to study the role of fluid mechanics and atherosclerosis. Various in vitro and in vivo models to study the role of flow in endothelial biology and pathobiology are discussed in this review. Furthermore, strategies used for the global profiling of the genome, transcriptome, miR-nome, DNA methylome, and metabolome, as they are important to define the biological and pathophysiological mechanisms of atherosclerosis. These "omics" approaches, especially those which derive data based on a single animal model, provide unprecedented opportunities to not only better understand the pathophysiology of atherosclerosis development in a holistic and integrative manner, but also to identify novel molecular and diagnostic targets.
Collapse
|
33
|
Sato M, Kuroda S, Mansjur KQ, Khaliunaa G, Nagata K, Horiuchi S, Inubushi T, Yamamura Y, Azuma M, Tanaka E. Low-intensity pulsed ultrasound rescues insufficient salivary secretion in autoimmune sialadenitis. Arthritis Res Ther 2015; 17:278. [PMID: 26445930 PMCID: PMC4596462 DOI: 10.1186/s13075-015-0798-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/24/2015] [Indexed: 11/17/2022] Open
Abstract
Introduction Low-intensity pulsed ultrasound (LIPUS) has been known to promote bone healing by nonthermal effects. In recent studies, LIPUS has been shown to reduce inflammation in injured soft tissues. Xerostomia is one of the most common symptoms in Sjögren syndrome (SS). It is caused by a decrease in the quantity or quality of saliva. The successful treatment of xerostomia is still difficult to achieve and often unsatisfactory. The aim of this study is to clarify the therapeutic effects of LIPUS on xerostomia in SS. Methods Human salivary gland acinar (NS-SV-AC) and ductal (NS-SV-DC) cells were cultured with or without tumor necrosis factor-α (TNF-α; 10 ng/ml) before LIPUS or sham exposure. The pulsed ultrasound signal was transmitted at a frequency of 1.5 MHz or 3 MHz with a spatial average intensity of 30 mW/cm2 and a pulse rate of 20 %. Cell number, net fluid secretion rate, and expression of aquaporin 5 (AQP5) and TNF-α were subsequently analyzed. Inhibitory effects of LIPUS on the nuclear factor κB (NF-κB) pathway were determined by Western blot analysis. The effectiveness of LIPUS in recovering salivary secretion was also examined in a MRL/MpJ/lpr/lpr (MRL/lpr) mouse model of SS with autoimmune sialadenitis. Results TNF-α stimulation of NS-SV-AC and NS-SV-DC cells resulted in a significant decrease in cell number and net fluid secretion rate (p < 0.01), whereas LIPUS treatment abolished them (p < 0.05). The expression changes of AQP5 and TNF-α were also inhibited in LIPUS treatment by blocking the NF-κB pathway. Furthermore, we found that mRNA expression of A20, a negative feedback regulator, was significantly increased by LIPUS treatment after TNF-α or interleukin 1β stimulation (NS-SV-AC, p < 0.01; NS-SV-DC, p < 0.05). In vivo LIPUS exposure to MRL/lpr mice exhibited a significant increase in both salivary flow and AQP5 expression by reducing inflammation in salivary glands (p < 0.01). Conclusions These results suggest that LIPUS upregulates expression of AQP5 and inhibits TNF-α production. Thus, LIPUS may restore secretion by inflamed salivary glands. It may synergistically activate negative feedback of NF-κB signaling in response to inflammatory stimulation. Collectively, LIPUS might be a new strategic therapy for xerostomia in autoimmune sialadenitis with SS. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0798-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Minami Sato
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Shingo Kuroda
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Karima Qurnia Mansjur
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Ganzorig Khaliunaa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Kumiko Nagata
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Shinya Horiuchi
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Toshihiro Inubushi
- Genetic Disease Program, Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| | - Yoshiko Yamamura
- Department of Oral Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Masayuki Azuma
- Department of Oral Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan. .,Department of Orthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
34
|
Freedman BR, Bade ND, Riggin CN, Zhang S, Haines PG, Ong KL, Janmey PA. The (dys)functional extracellular matrix. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3153-64. [PMID: 25930943 DOI: 10.1016/j.bbamcr.2015.04.015] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022]
Abstract
The extracellular matrix (ECM) is a major component of the biomechanical environment with which cells interact, and it plays important roles in both normal development and disease progression. Mechanical and biochemical factors alter the biomechanical properties of tissues by driving cellular remodeling of the ECM. This review provides an overview of the structural, compositional, and mechanical properties of the ECM that instruct cell behaviors. Case studies are reviewed that highlight mechanotransduction in the context of two distinct tissues: tendons and the heart. Although these two tissues demonstrate differences in relative cell-ECM composition and mechanical environment, they share similar mechanisms underlying ECM dysfunction and cell mechanotransduction. Together, these topics provide a framework for a fundamental understanding of the ECM and how it may vary across normal and diseased tissues in response to mechanical and biochemical cues. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Benjamin R Freedman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan D Bade
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Corinne N Riggin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sijia Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Philip G Haines
- Division of Cardiovascular Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katy L Ong
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Janmey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Dostal D, Glaser S, Baudino TA. Cardiac Fibroblast Physiology and Pathology. Compr Physiol 2015; 5:887-909. [DOI: 10.1002/cphy.c140053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Low-intensity pulsed ultrasound in dentofacial tissue engineering. Ann Biomed Eng 2015; 43:871-86. [PMID: 25672801 DOI: 10.1007/s10439-015-1274-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/04/2015] [Indexed: 02/04/2023]
Abstract
Oral and maxillofacial diseases affect millions of people worldwide and hence tissue engineering can be considered an interesting and clinically relevant approach to regenerate orofacial tissues after being affected by different diseases. Among several innovations for tissue regeneration, low-intensity pulsed ultrasound (LIPUS) has been used extensively in medicine as a therapeutic, operative, and diagnostic tool. LIPUS is accepted to promote bone fracture repair and regeneration. Furthermore, the effect of LIPUS on soft tissues regeneration has been paid much attention, and many studies have performed to evaluate the potential use of LIPUS to tissue engineering soft tissues. The present article provides an overview about the status of LIPUS stimulation as a tool to be used to enhance regeneration/tissue engineering. This review consists of five parts. Part 1 is a brief introduction of the acoustic description of LIPUS and mechanical action. In Part 2, biological problems in dentofacial tissue engineering are proposed. Part 3 explores biologic mechanisms of LIPUS to cells and tissues in living body. In Part 4, the effectiveness of LIPUS on cell metabolism and tissue regeneration in dentistry are summarized. Finally, Part 5 relates the possibility of clinical application of LIPUS in orthodontics. The present review brings out better understanding of the bioeffect of LIPUS therapy on orofacial tissues which is essential to the successful integration of management remedies for tissue regeneration/engineering. To develop an evidence-based approach to clinical management and treatment of orofacial degenerative diseases using LIPUS, we would like to be in full pursuit of LIPUS biotherapy. Still, there are many challenges for this relatively new strategy, but the up to date achievements using it promises to go far beyond the present possibilities.
Collapse
|
37
|
Graham ZA, Touchberry CD, Gupte AA, Bomhoff GL, Geiger PC, Gallagher PM. Changes in α7β1 integrin signaling after eccentric exercise in heat-shocked rat soleus. Muscle Nerve 2015; 51:562-8. [PMID: 24956997 DOI: 10.1002/mus.24324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2014] [Indexed: 12/16/2022]
Abstract
INTRODUCTION α7β1 integrin links the extracellular matrix to the focal adhesion (FA) in skeletal muscle and serves as a stabilizing and signal relayer. Heat shock (HS) induces expression of proteins that interact with the FA. METHODS Male Wistar rats were assigned to 1 of 3 groups: control (CON); eccentric exercise (EE); or EE+HS (HS). Soleus muscle was analyzed at 2 h and 48 h post-exercise. RESULTS The 120-kDa α7 integrin decreased in the EE and HS groups, and the 70-kDa peptide decreased in the EE group at 2 h post-exercise. Total expression of focal adhesion kinase (FAK) and RhoA were decreased in EE and HS at 2 h post-exercise. Expression of phosphorylated FAK(397) decreased in the EE group but not the HS group at 2 h post-exercise. CONCLUSIONS Long-duration EE may cause alterations in the FA in rat soleus muscle through the α7 integrin subunit and FAK.
Collapse
Affiliation(s)
- Zachary A Graham
- Applied Physiology Laboratory, Department of Health, Sport, and Exercise Sciences, University of Kansas, 101DJ Robinson Center, 1301 Sunnyside Avenue, Lawrence, Kansas, 66045, USA
| | | | | | | | | | | |
Collapse
|
38
|
Hamzeh MT, Sridhara R, Alexander LD. Cyclic stretch-induced TGF-β1 and fibronectin expression is mediated by β1-integrin through c-Src- and STAT3-dependent pathways in renal epithelial cells. Am J Physiol Renal Physiol 2014; 308:F425-36. [PMID: 25477471 DOI: 10.1152/ajprenal.00589.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Extracellular matrix (ECM) proteins, including fibronectin, may contribute to the early development and progression of renal interstitial fibrosis associated with chronic renal disease. Recent studies showed that β1-integrin is associated with the development of renal fibrosis in a murine model of unilateral ureteral obstruction (UUO). However, the molecular events responsible for β1-integrin-mediated signaling, following UUO, have yet to be determined. In this study, we investigated the mechanism by which mechanical stretch, an in vitro model for chronic obstructive nephropathy, regulates fibronectin and transforming growth factor-β1 (TGF-β1) expression in cultured human proximal tubular epithelium (HK-2) cells. Mechanical stretch upregulated fibronectin and TGF-β1 expression and activated signal transducer and transcription factor 3 (STAT3) in a time-dependent manner. Stretch-induced fibronectin and TGF-β1 were suppressed by a STAT3 inhibitor, S3I-201, and by small interfering RNA (siRNA) targeting human STAT3 (STAT3 siRNA). Similarly, fibronectin and TGF-β1 expression and STAT3 activation induced by mechanical stretch were suppressed by the Src family kinase inhibitor PP2 and by transfection of HK-2 cells with a dominant-negative mutant of c-Src (DN-Src), whereas PP3, an inactive analog of PP2, had no significant effect. Furthermore, mechanical stretch resulted in increased β1-integrin mRNA and protein levels in HK-2 cells. Furthermore, neutralizing antibody against β1-integrin and silencing of β1-integrin expression with siRNAs resulted in decreased c-Src and STAT3 activation and TGF-β1 and fibronectin expression evoked by mechanical stretch. This work demonstrates, for the first time, a role for β1-integrin in stretch-induced renal fibrosis through the activation of c-Src and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Mona T Hamzeh
- Department of Biology, Division of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan
| | - Rashmi Sridhara
- Midwestern University, Arizona College of Osteopathic Medicine, Department of Physiology, Glendale, Arizona; and
| | - Larry D Alexander
- Midwestern University, Arizona College of Osteopathic Medicine, Department of Physiology, Glendale, Arizona; and
| |
Collapse
|
39
|
Israeli-Rosenberg S, Chen C, Li R, Deussen DN, Niesman IR, Okada H, Patel HH, Roth DM, Ross RS. Caveolin modulates integrin function and mechanical activation in the cardiomyocyte. FASEB J 2014; 29:374-84. [PMID: 25366344 DOI: 10.1096/fj.13-243139] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
β1 integrins (β1) transduce mechanical signals in many cells, including cardiac myocytes (CM). Given their close localization, as well as their role in mechanotransduction and signaling, we hypothesized that caveolin (Cav) proteins might regulate integrins in the CM. β1 localization, complex formation, activation state, and signaling were analyzed using wild-type, Cav3 knockout, and Cav3 CM-specific transgenic heart and myocyte samples. Studies were performed under basal and mechanically loaded conditions. We found that: (1) β1 and Cav3 colocalize in CM and coimmunoprecipitate from CM protein lysates; (2) β1 is detected in a subset of caveolae; (3) loss of Cav3 caused reduction of β1D integrin isoform and active β1 integrin from the buoyant domains in the heart; (4) increased expression of myocyte Cav3 correlates with increased active β1 integrin in adult CM; (5) in vivo pressure overload of the wild-type heart results in increased activated integrin in buoyant membrane domains along with increased association between active integrin and Cav3; and (6) Cav3-deficient myocytes have perturbed basal and stretch mediated signaling responses. Thus, Cav3 protein can modify integrin function and mechanotransduction in the CM and intact heart.
Collapse
Affiliation(s)
- Sharon Israeli-Rosenberg
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Chao Chen
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Ruixia Li
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Daniel N Deussen
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Ingrid R Niesman
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Hideshi Okada
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Hemal H Patel
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - David M Roth
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| | - Robert S Ross
- *Department of Medicine and Department of Anesthesiology, University of California at San Diego, School of Medicine, San Diego, California, USA; U.S. Veterans Administration, San Diego Healthcare System, San Diego, California, USA; and Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
40
|
Youtz DJ, Isfort MC, Eichenseer CM, Nelin TD, Wold LE. In vitro effects of exercise on the heart. Life Sci 2014; 116:67-73. [PMID: 25218762 DOI: 10.1016/j.lfs.2014.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 08/21/2014] [Accepted: 08/23/2014] [Indexed: 12/28/2022]
Abstract
Pathologic and physiologic factors acting on the heart can produce consistent pressure changes, volume overload, or increased cardiac output. These changes may then lead to cardiac remodeling, ultimately resulting in cardiac hypertrophy. Exercise can also induce hypertrophy, primarily physiologic in nature. To determine the mechanisms responsible for each type of remodeling, it is important to examine the heart at the functional unit, the cardiomyocyte. Tests of individual cardiomyocyte function in vitro provide a deeper understanding of the changes occurring within the heart during hypertrophy. Examination of cardiomyocyte function during exercise primarily follows one of two pathways: the addition of hypertrophic inducing agents in vitro to normal cardiomyocytes, or the use of trained animal models and isolating cells following the development of hypertrophy in vivo. Due to the short lifespan of adult cardiomyocytes, a proportionately scant amount of research exists involving the direct stimulation of cells in vitro to induce hypertrophy. These attempts provide the only current evidence, as it is difficult to gather extensive data demonstrating cell growth as a result of in vitro physical stimulation. Researchers have created ways to combine skeletal myocytes with cardiomyocytes to produce functional muscle cells used to repair pathologic heart tissue, but continue to struggle with the short lifespan of these cells. While there have been promising findings regarding the mechanisms that surround cardiac hypertrophy in vitro, the translation of in vitro findings to in vivo function is not consistent. Therefore, the focus of this review is to highlight recent studies that have investigated the effect of exercise on the heart, both in vitro and in vivo.
Collapse
Affiliation(s)
- Dane J Youtz
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Michael C Isfort
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Timothy D Nelin
- College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Loren E Wold
- College of Nursing, The Ohio State University, Columbus, OH, USA; College of Medicine, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA; Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
41
|
Sato M, Nagata K, Kuroda S, Horiuchi S, Nakamura T, Karima M, Inubushi T, Tanaka E. Low-intensity pulsed ultrasound activates integrin-mediated mechanotransduction pathway in synovial cells. Ann Biomed Eng 2014; 42:2156-63. [PMID: 25096496 DOI: 10.1007/s10439-014-1081-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/30/2014] [Indexed: 01/15/2023]
Abstract
Low-intensity pulsed ultrasound (LIPUS) suppresses synovial hyperplasia and synovial cell proliferation characterized for rheumatoid arthritis, but the molecular mechanisms remain unknown. The purpose of this study was to examine the mechanotransduction pathway via the integrin/mitogen-activated protein kinase (MAPK) pathway in LIPUS exposure on the synovial membrane cells. Rabbit knee synovial membrane cell line, HIG-82, was cultured with or without FAK phosphorylation inhibitor, PF-573228. One hour after stimulation with PF-573228, the cells exposed to LIPUS for 20 min or sham exposure. A possible integrin/MAPK pathway was examined by immunofluorescence and Western blotting analysis with antibodies targeting specific phosphorylation sites on intracellular signaling proteins. LIPUS exposure increased phosphorylation of FAK, JNK, ERK, and p38, but the phosphorylation was inhibited by PF-573228. In conclusion, LIPUS exposure might be involved in cell apoptosis and survival of synovial membrane cells via integrin/FAK/MAPK pathway.
Collapse
Affiliation(s)
- Minami Sato
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Bioscience, Tokushima University Graduate School, 3-18-15 Kuramoto-Cho, Tokushima, 770-8504, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang W, Liu Q, Zhang Y, Zhao L. Involvement of ILK/ERK1/2 and ILK/p38 pathways in mediating the enhanced osteoblast differentiation by micro/nanotopography. Acta Biomater 2014; 10:3705-15. [PMID: 24769109 DOI: 10.1016/j.actbio.2014.04.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/20/2014] [Accepted: 04/16/2014] [Indexed: 12/31/2022]
Abstract
The hierarchical micro/nanotextured topography (MNT) on titanium (Ti) implant surface significantly enhances osteoblast differentiation. We have demonstrated that integrin-linked kinase (ILK) is a key underlying signal molecule and β-catenin is one of its downstream mediators in MNT-regulated osteoblast behavior. Here we propose that mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase 1/2 (ERK1/2), p38 and c-Jun NH2-terminal kinase (JNK), are other mediators downstream of ILK, and this study aims to confirm this. Firstly, the levels of ILK and MAPK activity in MG63 cells on MNT are examined by Western blot analysis. The ILK, ERK1/2 and p38 signals are significantly up-regulated by MNT, whereas the JNK activity is undetectable by Western blot. The MG63 cell morphology, proliferation and differentiation are studied in the absence and presence of the MAPK subgroup inhibitors to confirm their roles in cell functions on the Ti surface. The MAPK subgroup inhibitors obviously change the cell shape and depress cell proliferation. Blocking the ERK1/2 or p38 signaling, but not the JNK signaling, significantly down-regulates the cell osteogenesis-related gene expression, ALP production, collagen secretion and matrix mineralization. Afterwards, the ILK expression is down-regulated using ILK-specific siRNA (ILKsi) and then the MAPK activity is determined. ILKsi significantly attenuates the phosphorylated ERK1/2 and p38 levels on MNT, explicitly demonstrating that the ERK1/2 and p38 signalings are downstream effectors of ILK. In conclusion, these data demonstrate that both ILK/ERK1/2 and ILK/p38 pathways are involved in the mechanisms mediating the enhanced osteoblast differentiation by biomaterial surface topography, hopefully directing the biomaterial modification and biofunctionalization.
Collapse
|
43
|
Dhein S, Schreiber A, Steinbach S, Apel D, Salameh A, Schlegel F, Kostelka M, Dohmen PM, Mohr FW. Mechanical control of cell biology. Effects of cyclic mechanical stretch on cardiomyocyte cellular organization. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 115:93-102. [PMID: 24983489 DOI: 10.1016/j.pbiomolbio.2014.06.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 01/04/2023]
Abstract
OBJECTIVES The aim of our study was to elucidate how cyclic mechanical stretch is sensed by cardiomyocytes and in which way it affects cytoskeletal organization. METHODS Neonatal rat cardiomyocytes, cultured on flexible membranes, were subjected to cyclic mechanical stretch (1 Hz, 10% elongation) for 24 h using either round or rectangular loading posts for equibi-axial or uni-axial stretch, respectively, using the FlexCell stretch system. Cells were treated either with vehicle, the focal adhesion kinase (FAK) inhibitor PF-573,228 (200 nM), or the stretch-activated ion channel blocker gadolinium (Gd(3+); 100 μM). RESULTS Cyclic mechanical stretch (36 mm diameter silicone membrane, equibi-axial stretch, 10% elongation, 1 Hz) induced elongation of the cardiomyocytes together with accentuation of Cx43 at the cell poles, and with an orientation of the cell axis between the radial axis and the circumferential axis (mean deviation: 11° from the circumference). Moreover, stretch resulted in ca. 1.4 fold increased Cx43 expression. FAK was found to be phosphorylated at the edges of the cells. In order to find out, how cardiomyocytes might sense stretch, we investigated possible effects of Gd(3+)and PF-573,228. Gd(3+) had no effect on elongation or polarization and did not affect stretch-induced Cx43 expression. Interestingly, the FAK inhibitor completely antagonized the stretch-induced elongation, orientation and Cx43-polarization. However, the stretch-induced Cx43 expression was insensitive to this treatment. In order to clarify our result that the cells in equibi-axial stretch did not exactly organize to the circumference or to the radial axis, we decided to use a uni-axial stretch protocol. In uni-axially stretched cells, we found that the cardiomyocytes also showed elongation, Cx43 polarization, and orientation near to the stretch axis, but not exactly in the stretch axis but ca. 25° oblique to it. Furthermore, we investigated the tubular system, the Golgi apparatus, the SR and the nucleus. After 24 h stretch the microtubules were localized nearly (but not completely) parallel to the stretch axis (i.e. in longitudinal cell axis). Moreover, the localization of nucleus and the Golgi was also changed: while under static conditions, the Golgi was distributed more or less around the nucleus, after stretch the Golgi was accentuated at one site of the nucleus facing a cell pole with the nucleus facing the opposite cell pole. The plus motor protein kinesin accentuated at the cell poles and at the cell periphery, while the minus motor protein dynein was found near to the Golgi apparatus. CONCLUSIONS The stretch signal sensing is mediated via FAK and leads to intracellular re-organization and orientation. The oblique orientation of the cell with regard to the direction of stretch may define a directed force vector which could allow the cell to orientate.
Collapse
Affiliation(s)
- Stefan Dhein
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany.
| | - Anna Schreiber
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Sabine Steinbach
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Daniel Apel
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Aida Salameh
- Clinic for Pediatric Cardiology, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Franziska Schlegel
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Martin Kostelka
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Pascal M Dohmen
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| | - Friedrich Wilhelm Mohr
- Clinic for Cardiac Surgery, Heart Center Leipzig, University Leipzig, Struempellstr. 39, 04289 Leipzig, Germany
| |
Collapse
|
44
|
Dostal DE, Feng H, Nizamutdinov D, Golden HB, Afroze SH, Dostal JD, Jacob JC, Foster DM, Tong C, Glaser S, Gerilechaogetu F. Mechanosensing and Regulation of Cardiac Function. ACTA ACUST UNITED AC 2014; 5:314. [PMID: 25485172 PMCID: PMC4255974 DOI: 10.4172/2155-9880.1000314] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The role of mechanical force as an important regulator of structure and function of mammalian cells, tissues, and organs has recently been recognized. However, mechanical overload is a pathogenesis or comorbidity existing in a variety of heart diseases, such as hypertension, aortic regurgitation and myocardial infarction. Physical stimuli sensed by cells are transmitted through intracellular signal transduction pathways resulting in altered physiological responses or pathological conditions. Emerging evidence from experimental studies indicate that β1-integrin and the angiotensin II type I (AT1) receptor play critical roles as mechanosensors in the regulation of heart contraction, growth and leading to heart failure. Integrin link the extracellular matrix and the intracellular cytoskeleton to initiate the mechanical signalling, whereas, the AT1 receptor could be activated by mechanical stress through an angiotensin-II-independent mechanism. Recent studies show that both Integrin and AT1 receptor and their downstream signalling factors including MAPKs, AKT, FAK, ILK and GTPase regulate heart function in cardiac myocytes. In this review we describe the role of mechanical sensors residing within the plasma membrane, mechanical sensor induced downstream signalling factors and its potential roles in cardiac contraction and growth.
Collapse
Affiliation(s)
- David E Dostal
- Central Texas Veterans Health Care System, Temple, Texas, USA ; Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Hao Feng
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Damir Nizamutdinov
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Honey B Golden
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Syeda H Afroze
- Scott & White Healthcare - Digestive Disease Research Centre, Temple, Texas, USA
| | - Joseph D Dostal
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - John C Jacob
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Donald M Foster
- Central Texas Veterans Health Care System, Temple, Texas, USA
| | - Carl Tong
- Systems Biology and Translational Medicine, the Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Shannon Glaser
- Central Texas Veterans Health Care System, Temple, Texas, USA ; Scott & White Healthcare - Digestive Disease Research Centre, Temple, Texas, USA
| | - Fnu Gerilechaogetu
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| |
Collapse
|
45
|
|
46
|
Li R, Narici MV, Erskine RM, Seynnes OR, Rittweger J, Pišot R, Šimunič B, Flück M. Costamere remodeling with muscle loading and unloading in healthy young men. J Anat 2013; 223:525-36. [PMID: 24010829 PMCID: PMC3916893 DOI: 10.1111/joa.12101] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2013] [Indexed: 11/28/2022] Open
Abstract
Costameres are mechano-sensory sites of focal adhesion in the sarcolemma that provide a structural anchor for myofibrils. Their turnover is regulated by integrin-associated focal adhesion kinase (FAK). We hypothesized that changes in content of costamere components (beta 1 integrin, FAK, meta-vinculin, gamma-vinculin) with increased and reduced loading of human anti-gravity muscle would: (i) relate to changes in muscle size and molecular parameters of muscle size regulation [p70S6K, myosin heavy chain (MHC)1 and MHCIIA]; (ii) correspond to adjustments in activity and expression of FAK, and its negative regulator, FRNK; and (iii) reflect the temporal response to reduced and increased loading. Unloading induced a progressive decline in thickness of human vastus lateralis muscle after 8 and 34 days of bedrest (−4% and −14%, respectively; n = 9), contrasting the increase in muscle thickness after 10 and 27 days of resistance training (+5% and +13%; n = 6). Changes in muscle thickness were correlated with changes in cross-sectional area of type I muscle fibers (r = 0.66) and beta 1 integrin content (r = 0.76) at the mid-point of altered loading. Changes in meta-vinculin and FAK-pY397 content were correlated (r = 0.85) and differed, together with the changes of beta 1 integrin, MHCI, MHCII and p70S6K, between the mid- and end-point of resistance training. By contrast, costamere protein level changes did not differ between time points of bedrest. The findings emphasize the role of FAK-regulated costamere turnover in the load-dependent addition and removal of myofibrils, and argue for two phases of muscle remodeling with resistance training, which do not manifest at the macroscopic level.
Collapse
Affiliation(s)
- Ruowei Li
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, Manchester, UK
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Jacques P, Lambrecht S, Verheugen E, Pauwels E, Kollias G, Armaka M, Verhoye M, Van der Linden A, Achten R, Lories RJ, Elewaut D. Proof of concept: enthesitis and new bone formation in spondyloarthritis are driven by mechanical strain and stromal cells. Ann Rheum Dis 2013; 73:437-45. [PMID: 23921997 DOI: 10.1136/annrheumdis-2013-203643] [Citation(s) in RCA: 299] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Spondyloarthritides (SpA) are characterised by both peripheral and axial arthritis. The hallmarks of peripheral SpA are the development of enthesitis, most typically of the Achilles tendon and plantar fascia, and new bone formation. This study was undertaken to unravel the mechanisms leading towards enthesitis and new bone formation in preclinical models of SpA. RESULTS First, we demonstrated that TNF(ΔARE) mice show typical inflammatory features highly reminiscent of SpA. The first signs of inflammation were found at the entheses. Importantly, enthesitis occurred equally in the presence or absence of mature T and B cells, underscoring the importance of stromal cells. Hind limb unloading in TNF(ΔARE) mice significantly suppressed inflammation of the Achilles tendon compared with weight bearing controls. Erk1/2 signalling plays a crucial role in mechanotransduction-associated inflammation. Furthermore, new bone formation is strongly promoted at entheseal sites by biomechanical stress and correlates with the degree of inflammation. CONCLUSIONS These findings provide a formal proof of the concept that mechanical strain drives both entheseal inflammation and new bone formation in SpA.
Collapse
Affiliation(s)
- Peggy Jacques
- Laboratory for Molecular Immunology and Inflammation, Department of Rheumatology, University Hospital, , Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Klossner S, Li R, Ruoss S, Durieux AC, Flück M. Quantitative changes in focal adhesion kinase and its inhibitor, FRNK, drive load-dependent expression of costamere components. Am J Physiol Regul Integr Comp Physiol 2013; 305:R647-57. [PMID: 23904105 DOI: 10.1152/ajpregu.00007.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Costameres are mechanosensory sites of focal adhesion in the sarcolemma that reinforce the muscle-fiber composite and provide an anchor for myofibrillogenesis. We hypothesized that elevated content of the integrin-associated regulator of costamere turnover in culture, focal adhesion kinase (FAK), drives changes in costamere component content in antigravity muscle in a load-dependent way in correspondence with altered muscle weight. The content of FAK in soleus muscle being phosphorylated at autoregulatory tyrosine 397 (FAK-pY397) was increased after 20 s of stretch. FAK-pY397 content remained elevated after 24 h of stretch-overload due to upregulated FAK content. Overexpression of FAK in soleus muscle fibers by means of gene electrotransfer increased the β1-integrin (+56%) and meta-vinculin (+88%) content. α7-Integrin (P = 0.46) and γ-vinculin (P = 0.18) content was not altered after FAK overexpression. Co-overexpression of the FAK inhibitor FAK-related nonkinase (FRNK) reduced FAK-pY397 content by 33% and increased the percentage of fast-type fibers that arose in connection with hybrid fibers with gene transfer. Transplantation experiments confirmed the association of FRNK expression with slow-to-fast fiber transformation. Seven days of unloading blunted the elevation of FAK-pY397, β1-integrin, and meta-vinculin content with FAK overexpression, and this was reversed by 1 day of reloading. The results highlight that the expression of components for costameric attachment sites of myofibrils is under load- and fiber type-related control via FAK and its inhibitor FRNK.
Collapse
|
49
|
Anthrax lethal toxin induces acute diastolic dysfunction in rats through disruption of the phospholamban signaling network. Int J Cardiol 2013; 168:3884-95. [PMID: 23907041 DOI: 10.1016/j.ijcard.2013.06.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/09/2013] [Accepted: 06/28/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND Anthrax lethal toxin (LT), secreted by Bacillus anthracis, causes severe cardiac dysfunction by unknown mechanisms. LT specifically cleaves the docking domains of MAPKK (MEKs); thus, we hypothesized that LT directly impairs cardiac function through dysregulation of MAPK signaling mechanisms. METHODS AND RESULTS In a time-course study of LT toxicity, echocardiography revealed acute diastolic heart failure accompanied by pulmonary regurgitation and left atrial dilation in adult Sprague-Dawley rats at time points corresponding to dysregulated JNK, phospholamban (PLB) and protein phosphatase 2A (PP2A) myocardial signaling. Using isolated rat ventricular myocytes, we identified the MEK7-JNK1-PP2A-PLB signaling axis to be important for regulation of intracellular calcium (Ca(2+)(i)) handling, PP2A activation and targeting of PP2A-B56α to Ca(2+)(i) handling proteins, such as PLB. Through a combination of gain-of-function and loss-of-function studies, we demonstrated that over-expression of MEK7 protects against LT-induced PP2A activation and Ca(2+)(i) dysregulation through activation of JNK1. Moreover, targeted phosphorylation of PLB-Thr(17) by Akt improved sarcoplasmic reticulum Ca(2+)(i) release and reuptake during LT toxicity. Co-immunoprecipitation experiments further revealed the pivotal role of MEK7-JNK-Akt complex formation for phosphorylation of PLB-Thr(17) during acute LT toxicity. CONCLUSIONS Our findings support a cardiogenic mechanism of LT-induced diastolic dysfunction, by which LT disrupts JNK1 signaling and results in Ca(2+)(i) dysregulation through diminished phosphorylation of PLB by Akt and increased dephosphorylation of PLB by PP2A. Integration of the MEK7-JNK1 signaling module with Akt represents an important stress-activated signalosome that may confer protection to sustain cardiac contractility and maintain normal levels of Ca(2+)(i) through PLB-T(17) phosphorylation.
Collapse
|
50
|
Cha KJ, Hong JM, Cho DW, Kim DS. Enhanced osteogenic fate and function of MC3T3-E1 cells on nanoengineered polystyrene surfaces with nanopillar and nanopore arrays. Biofabrication 2013; 5:025007. [DOI: 10.1088/1758-5082/5/2/025007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|