1
|
Hartley A, Williams PM, Mata A. A Comparison of the Mechanical Properties of ECM Components and Synthetic Self-Assembling Peptides. Adv Healthc Mater 2025; 14:e2402385. [PMID: 39972630 PMCID: PMC12023840 DOI: 10.1002/adhm.202402385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 01/20/2025] [Indexed: 02/21/2025]
Abstract
The field of tissue engineering is increasingly moving away from a one-size-fits-all approach of simple synthetic homogeneous gels, and embracing more tailored designs to optimize cell function and differentiation for the organ of interest. Extracellular matrix (ECM) proteins are still the optimal route for controlling cell function, while a field of great promise is that of synthetic self-assembling peptides (SSAPs), which are fully biocompatible, biodegradable, and offer both the hierarchical structure and dynamic properties displayed by protein networks found in natural tissue. However, the mechanical properties of neither group have been comprehensively reviewed. In this review, rheological data and the Young's modulus of the most prevalent proteins involved in the ECM (collagen I, elastin, and fibronectin) are collated for the first time, and compared against the most widely researched SSAPs: peptide amphiphiles (PAs), β-sheets, β-hairpin peptides, and Fmoc-based gels (with a focus on PA-E3, RADA16, MAX1, and FmocFF, respectively).
Collapse
Affiliation(s)
- Alex Hartley
- School of Pharmacy, University of NottinghamUniversity Park CampusNottinghamNG7 2RDUK
- Biodiscovery Institute, University of NottinghamUniversity Park CampusNottinghamNG7 2RDUK
| | | | - Alvaro Mata
- School of Pharmacy, University of NottinghamUniversity Park CampusNottinghamNG7 2RDUK
- Biodiscovery Institute, University of NottinghamUniversity Park CampusNottinghamNG7 2RDUK
- Department of Chemical and Environmental Engineering, University of NottinghamUniversity Park CampusNottinghamNG7 2RDUK
| |
Collapse
|
2
|
Kalaitzidou C, Grekas G, Zilian A, Makridakis C, Rosakis P. Compressive instabilities enable cell-induced extreme densification patterns in the fibrous extracellular matrix: Discrete model predictions. PLoS Comput Biol 2024; 20:e1012238. [PMID: 38950077 PMCID: PMC11244807 DOI: 10.1371/journal.pcbi.1012238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/12/2024] [Accepted: 06/08/2024] [Indexed: 07/03/2024] Open
Abstract
We present a new model and extensive computations that explain the dramatic remodelling undergone by a fibrous collagen extracellular matrix (ECM), when subjected to contractile mechanical forces from embedded cells or cell clusters. This remodelling creates complex patterns, comprising multiple narrow localised bands of severe densification and fiber alignment, extending far into the ECM, often joining distant cells or cell clusters (such as tumours). Most previous models cannot capture this behaviour, as they assume stable mechanical fiber response with stress an increasing function of fiber stretch, and a restriction to small displacements. Our fully nonlinear network model distinguishes between two types of single-fiber nonlinearity: fibers that undergo stable (supercritical) buckling (as in previous work) versus fibers that suffer unstable (subcritical) buckling collapse. The model allows unrestricted, arbitrarily large displacements (geometric nonlinearity). Our assumptions on single-fiber instability are supported by recent simulations and experiments on buckling of individual beams with a hierarchical microstructure, such as collagen fibers. We use simple scenarios to illustrate, for the first time, two distinct compressive-instability mechanisms at work in our model: unstable buckling collapse of single fibers, and snap-through of multiple-fiber groups. The latter is possible even when single fibers are stable. Through simulations of large fiber networks, we show how these instabilities lead to spatially extended patterns of densification, fiber alignment and ECM remodelling induced by cell contraction. Our model is simple, but describes a very complex, multi-stable energy landscape, using sophisticated numerical optimisation methods that overcome the difficulties caused by instabilities in large systems. Our work opens up new ways of understanding the unique biomechanics of fibrous-network ECM, by fully accounting for nonlinearity and associated loss of stability in fiber networks. Our results provide new insights on tumour invasion and metastasis.
Collapse
Affiliation(s)
- Chrysovalantou Kalaitzidou
- Department of Engineering, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch sur Alzette, Luxembourg
| | - Georgios Grekas
- Aerospace Engineering and Mechanics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Andreas Zilian
- Department of Engineering, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch sur Alzette, Luxembourg
| | - Charalambos Makridakis
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Mathematics and Applied Mathematics, University of Crete, Heraklion, Greece
- Department of Mathematics, MPS, University of Sussex, Brighton, United Kingdom
| | - Phoebus Rosakis
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Mathematics and Applied Mathematics, University of Crete, Heraklion, Greece
| |
Collapse
|
3
|
Liu H, Zhou Y, Guo P, Zheng X, Chen W, Zhang S, Fu Y, Zhou X, Wan Z, Zhao B, Zhao Y. Hemodialysis bilayer bionic blood vessels developed by the mechanical stimulation of hepatitis B viral X( HBX) gene- transfected hepatic stellate cells. J Zhejiang Univ Sci B 2024; 25:499-512. [PMID: 38910495 PMCID: PMC11199092 DOI: 10.1631/jzus.b2300479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/23/2023] [Indexed: 06/25/2024]
Abstract
Artificial vascular graft (AVG) fistula is widely used for hemodialysis treatment in patients with renal failure. However, it has poor elasticity and compliance, leading to stenosis and thrombosis. The ideal artificial blood vessel for dialysis should replicate the structure and components of a real artery, which is primarily maintained by collagen in the extracellular matrix (ECM) of arterial cells. Studies have revealed that in hepatitis B virus (HBV)-induced liver fibrosis, hepatic stellate cells (HSCs) become hyperactive and produce excessive ECM fibers. Furthermore, mechanical stimulation can encourage ECM secretion and remodeling of a fiber structure. Based on the above factors, we transfected HSCs with the hepatitis B viral X (HBX) gene for simulating the process of HBV infection. Subsequently, these HBX-HSCs were implanted into a polycaprolactone-polyurethane (PCL-PU) bilayer scaffold in which the inner layer is dense and the outer layer consists of pores, which was mechanically stimulated to promote the secretion of collagen nanofiber from the HBX-HSCs and to facilitate crosslinking with the scaffold. We obtained an ECM-PCL-PU composite bionic blood vessel that could act as access for dialysis after decellularization. Then, the vessel scaffold was implanted into a rabbit's neck arteriovenous fistula model. It exhibited strong tensile strength and smooth blood flow and formed autologous blood vessels in the rabbit's body. Our study demonstrates the use of human cells to create biomimetic dialysis blood vessels, providing a novel approach for creating clinical vascular access for dialysis.
Collapse
Affiliation(s)
- Hongyi Liu
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yuanyuan Zhou
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China.
- School of Medicine, Xiamen University, Xiamen 361102, China.
| | - Peng Guo
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Xiongwei Zheng
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Weibin Chen
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Shichao Zhang
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yu Fu
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
- School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xu Zhou
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Zheng Wan
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Bin Zhao
- Xiamen Health and Medical Big Data Center, Xiamen 361008, China
| | - Yilin Zhao
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China.
- School of Medicine, Xiamen University, Xiamen 361102, China.
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Zhongshan Hospital Affiliated to Xiamen University), Xiamen 361004, China.
- Xiamen Key Laboratory of Cellular Intervention and Interventional Medical Materials, Xiamen 361004, China.
| |
Collapse
|
4
|
Azzari NA, Segars KL, Rapaka S, Kushimi L, Rich CB, Trinkaus-Randall V. Aberrations in Cell Signaling Quantified in Diabetic Murine Globes after Injury. Cells 2023; 13:26. [PMID: 38201230 PMCID: PMC10778404 DOI: 10.3390/cells13010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The corneal epithelium is an avascular structure that has a unique wound healing mechanism, which allows for rapid wound closure without compromising vision. This wound healing mechanism is attenuated in diabetic patients, resulting in poor clinical outcomes and recurrent non-healing erosion. We investigated changes in cellular calcium signaling activity during the wound response in murine diabetic tissue using live cell imaging from both ex vivo and in vitro models. The calcium signaling propagation in diabetic cells was significantly decreased and displayed altered patterns compared to non-diabetic controls. Diabetic cells and tissue display distinct expression of the purinergic receptor, P2X7, which mediates the wound healing response. We speculate that alterations in P2X7 expression, interactions with other proteins, and calcium signaling activity significantly impact the wound healing response. This may explain aberrations in the diabetic wound response.
Collapse
Affiliation(s)
- Nicholas A. Azzari
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA; (N.A.A.); (C.B.R.)
| | - Kristen L. Segars
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA;
| | - Srikar Rapaka
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA;
| | - Landon Kushimi
- Department of Computer Science, Center for Computing and Data Sciences, Boston University, 665 Commonwealth Ave, Boston, MA 02115, USA;
| | - Celeste B. Rich
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA; (N.A.A.); (C.B.R.)
| | - Vickery Trinkaus-Randall
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA; (N.A.A.); (C.B.R.)
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA;
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, 72 E. Concord St., Boston, MA 02118, USA
| |
Collapse
|
5
|
Krajnik A, Nimmer E, Brazzo JA, Biber JC, Drewes R, Tumenbayar BI, Sullivan A, Pham K, Krug A, Heo Y, Kolega J, Heo SJ, Lee K, Weil BR, Kim DH, Gupte SA, Bae Y. Survivin regulates intracellular stiffness and extracellular matrix production in vascular smooth muscle cells. APL Bioeng 2023; 7:046104. [PMID: 37868708 PMCID: PMC10590228 DOI: 10.1063/5.0157549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Vascular dysfunction is a common cause of cardiovascular diseases characterized by the narrowing and stiffening of arteries, such as atherosclerosis, restenosis, and hypertension. Arterial narrowing results from the aberrant proliferation of vascular smooth muscle cells (VSMCs) and their increased synthesis and deposition of extracellular matrix (ECM) proteins. These, in turn, are modulated by arterial stiffness, but the mechanism for this is not fully understood. We found that survivin is an important regulator of stiffness-mediated ECM synthesis and intracellular stiffness in VSMCs. Whole-transcriptome analysis and cell culture experiments showed that survivin expression is upregulated in injured femoral arteries in mice and in human VSMCs cultured on stiff fibronectin-coated hydrogels. Suppressed expression of survivin in human VSMCs significantly decreased the stiffness-mediated expression of ECM components related to arterial stiffening, such as collagen-I, fibronectin, and lysyl oxidase. By contrast, expression of these ECM proteins was rescued by ectopic expression of survivin in human VSMCs cultured on soft hydrogels. Interestingly, atomic force microscopy analysis showed that suppressed or ectopic expression of survivin decreases or increases intracellular stiffness, respectively. Furthermore, we observed that inhibiting Rac and Rho reduces survivin expression, elucidating a mechanical pathway connecting intracellular tension, mediated by Rac and Rho, to survivin induction. Finally, we found that survivin inhibition decreases FAK phosphorylation, indicating that survivin-dependent intracellular tension feeds back to maintain signaling through FAK. These findings suggest a novel mechanism by which survivin potentially modulates arterial stiffness.
Collapse
Affiliation(s)
- Amanda Krajnik
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Erik Nimmer
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - John C. Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Andra Sullivan
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | - Khanh Pham
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Alanna Krug
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York 14260, USA
| | | | - John Kolega
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Su-Jin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | - Brian R. Weil
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Sachin A. Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | - Yongho Bae
- Author to whom correspondence should be addressed:
| |
Collapse
|
6
|
Mielnicka A, Kołodziej T, Dziob D, Lasota S, Sroka J, Rajfur Z. Impact of elastic substrate on the dynamic heterogeneity of WC256 Walker carcinosarcoma cells. Sci Rep 2023; 13:15743. [PMID: 37735532 PMCID: PMC10514059 DOI: 10.1038/s41598-023-35313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 05/16/2023] [Indexed: 09/23/2023] Open
Abstract
Cellular heterogeneity is a phenomenon in which cell populations are composed of subpopulations that vary in their behavior. Heterogeneity is particularly pronounced in cancer cells and can affect the efficacy of oncological therapies. Previous studies have considered heterogeneity dynamics to be indicative of evolutionary changes within subpopulations; however, these studies do not consider the short-time morphological plasticity of cells. Physical properties of the microenvironment elasticity have also been poorly investigated within the context of cellular heterogeneity, despite its role in determining cellular behavior. This article demonstrates that cellular heterogeneity can be highly dynamic and dependent on the micromechanical properties of the substrate. During observation, migrating Walker carcinosarcoma WC256 cells were observed to belong to different subpopulations, in which their morphologies and migration strategies differed. Furthermore, the application of an elastic substrate (E = 40 kPa) modified three aspects of cellular heterogeneity: the occurrence of subpopulations, the occurrence of transitions between subpopulations, and cellular migration and morphology. These findings provide a new perspective in the analysis of cellular heterogeneity, whereby it may not be a static feature of cancer cell populations, instead varying over time. This helps further the understanding of cancer cell behavior, including their phenotype and migration strategy, which may help to improve cancer therapies by extending their suitability to investigate tumor heterogeneity.
Collapse
Affiliation(s)
- Aleksandra Mielnicka
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, ul. Lojasiewicza 11, 30-348, Kraków, Poland
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, PAS, ul. Ludwika Pasteura 3, 02-093, Warsaw, Poland
| | - Tomasz Kołodziej
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, ul. Lojasiewicza 11, 30-348, Kraków, Poland
- Department of Pharmaceutical Biophysics, Faculty of Pharmacy, Jagiellonian University Medical College, ul. Medyczna 9, 30-688, Kraków, Poland
| | - Daniel Dziob
- Department of Pharmaceutical Biophysics, Faculty of Pharmacy, Jagiellonian University Medical College, ul. Medyczna 9, 30-688, Kraków, Poland
| | - Sławomir Lasota
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387, Kraków, Poland
| | - Jolanta Sroka
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387, Kraków, Poland
| | - Zenon Rajfur
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, ul. Lojasiewicza 11, 30-348, Kraków, Poland.
- Jagiellonian Center of Biomedical Imaging, Jagiellonian University, 30-348, Kraków, Poland.
| |
Collapse
|
7
|
Sun CC, Lee SY, Chen LH, Lai CH, Shen ZQ, Chen NN, Lai YS, Tung CY, Tzeng TY, Chiu WT, Tsai TF. Targeting Ca 2+-dependent pathways to promote corneal epithelial wound healing induced by CISD2 deficiency. Cell Signal 2023:110755. [PMID: 37315750 DOI: 10.1016/j.cellsig.2023.110755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Chronic epithelial defects of the cornea, which are usually associated with severe dry eye disease, diabetes mellitus, chemical injuries or neurotrophic keratitis, as well as aging, are an unmet clinical need. CDGSH Iron Sulfur Domain 2 (CISD2) is the causative gene for Wolfram syndrome 2 (WFS2; MIM 604928). CISD2 protein is significantly decreased in the corneal epithelium of patients with various corneal epithelial diseases. Here we summarize the most updated publications and discuss the central role of CISD2 in corneal repair, as well as providing new results describing how targeting Ca2+-dependent pathways can improve corneal epithelial regeneration. This review mainly focuses on the following topics. Firstly, an overview of the cornea and of corneal epithelial wound healing. The key players involved in this process, such as Ca2+, various growth factors/cytokines, extracellular matrix remodeling, focal adhesions and proteinases, are briefly discussed. Secondly, it is well known that CISD2 plays an essential role in corneal epithelial regeneration via the maintenance of intracellular Ca2+ homeostasis. CISD2 deficiency dysregulates cytosolic Ca2+, impairs cell proliferation and migration, decreases mitochondrial function and increases oxidative stress. As a consequence, these abnormalities bring about poor epithelial wound healing and this, in turn, will lead to persistent corneal regeneration and limbal progenitor cell exhaustion. Thirdly, CISD2 deficiency induces three distinct Ca2+-dependent pathways, namely the calcineurin, CaMKII and PKCα signaling pathways. Intriguingly, inhibition of each of the Ca2+-dependent pathways seems to reverse cytosolic Ca2+ dysregulation and restore cell migration during corneal wound healing. Notably, cyclosporin, an inhibitor of calcineurin, appears to have a dual effect on both inflammatory and corneal epithelial cells. Finally, corneal transcriptomic analyses have revealed that there are six major functional groupings of differential expression genes when CISD2 deficiency is present: (1) inflammation and cell death; (2) cell proliferation, migration and differentiation; (3) cell adhesion, junction and interaction; (4) Ca2+ homeostasis; (5) wound healing and extracellular matrix; and (6) oxidative stress and aging. This review highlights the importance of CISD2 in corneal epithelial regeneration and identifies the potential of repurposing venerable FDA-approved drugs that target Ca2+-dependent pathways for new uses, namely treating chronic epithelial defects of the cornea.
Collapse
Affiliation(s)
- Chi-Chin Sun
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Shao-Yun Lee
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Li-Hsien Chen
- Department of Pharmacology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chia-Hui Lai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 333, Taiwan
| | - Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Nan-Ni Chen
- Department of Ophthalmology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Yi-Shyun Lai
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Chien-Yi Tung
- Genomics Center for Clinical and Biotechnological Applications, Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Tsai-Yu Tzeng
- Genomics Center for Clinical and Biotechnological Applications, Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan 701, Taiwan.
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 350, Taiwan; Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| |
Collapse
|
8
|
Segars KL, Azzari NA, Gomez S, Machen C, Rich CB, Trinkaus-Randall V. Age Dependent Changes in Corneal Epithelial Cell Signaling. Front Cell Dev Biol 2022; 10:886721. [PMID: 35602595 PMCID: PMC9117764 DOI: 10.3389/fcell.2022.886721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
The cornea is exposed daily to a number of mechanical stresses including shear stress from tear film and blinking. Over time, these stressors can lead to changes in the extracellular matrix that alter corneal stiffness, cell-substrate structures, and the integrity of cell-cell junctions. We hypothesized that changes in tissue stiffness of the cornea with age may alter calcium signaling between cells after injury, and the downstream effects of this signaling on cellular motility and wound healing. Nanoindentation studies revealed that there were significant differences in the stiffness of the corneal epithelium and stroma between corneas of 9- and 27-week mice. These changes corresponded to differences in the timeline of wound healing and in cell signaling. Corneas from 9-week mice were fully healed within 24 h. However, the wounds on corneas from 27-week mice remained incompletely healed. Furthermore, in the 27-week cohort there was no detectable calcium signaling at the wound in either apical or basal corneal epithelial cells. This is in contrast to the young cohort, where there was elevated basal cell activity relative to background levels. Cell culture experiments were performed to assess the roles of P2Y2, P2X7, and pannexin-1 in cellular motility during wound healing. Inhibition of P2Y2, P2X7, or pannexin-1 all significantly reduce wound closure. However, the inhibitors all have different effects on the trajectories of individual migrating cells. Together, these findings suggest that there are several significant differences in the stiffness and signaling that underlie the decreased wound healing efficacy of the cornea in older mice.
Collapse
Affiliation(s)
- Kristen L. Segars
- Department of Pharmacology, School of Medicine, Boston University, Boston, MA, United States
| | - Nicholas A. Azzari
- Department of Biochemistry, School of Medicine, Boston University, Boston, MA, United States
| | - Stephanie Gomez
- Department of Biochemistry, School of Medicine, Boston University, Boston, MA, United States
| | - Cody Machen
- Department of Biochemistry, School of Medicine, Boston University, Boston, MA, United States
| | - Celeste B. Rich
- Department of Ophthalmology, School of Medicine, School of Medicine, Boston, MA, United States
| | - Vickery Trinkaus-Randall
- Department of Biochemistry, School of Medicine, Boston University, Boston, MA, United States
- Department of Ophthalmology, School of Medicine, School of Medicine, Boston, MA, United States
- *Correspondence: Vickery Trinkaus-Randall,
| |
Collapse
|
9
|
Buglakov AI, Vasilevskaya VV. Fibrillar gel self-assembly via cononsolvency of amphiphilic polymer. J Colloid Interface Sci 2022; 614:181-193. [DOI: 10.1016/j.jcis.2022.01.095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 11/25/2022]
|
10
|
Mechanically Tunable Extracellular Matrix of Genipin Crosslinked Collagen and Its Effect on Endothelial Function. APPLIED SCIENCES-BASEL 2022; 12:2401. [PMID: 36713025 PMCID: PMC9881191 DOI: 10.3390/app12052401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mechanical rigidity of a matrix, to which cells adhere, plays a significant role in regulating phenotypic cellular behaviors such as spreading and junction formation because vascular cells sense and respond to changes in their mechanical environment. Controlling mechanical properties of extracellular matrix by using a crosslinker is important for cell and tissue mechanobiology. In this paper, we explored genipin, a natural plant extract, to crosslink collagen-I in order to enhance mechanical properties with low cytotoxicity. We characterized the effects of genipin concentration on the mechanical properties, color change, degradation, structure, cell viability, and endothelial function such as transendothelial electrical resistance (TEER). Through the analysis of both material properties and endothelial response, it was found that genipin-based glycation caused an increase in viscoelastic moduli in collagen hydrogels, as well as increased fiber density in their structural morphology. Endothelial cells were found to form better barriers, express higher levels of tight junction proteins, and exhibit better adhesion on stiffer matrices.
Collapse
|
11
|
Atat OE, Farzaneh Z, Pourhamzeh M, Taki F, Abi-Habib R, Vosough M, El-Sibai M. 3D modeling in cancer studies. Hum Cell 2021; 35:23-36. [PMID: 34761350 DOI: 10.1007/s13577-021-00642-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/31/2021] [Indexed: 01/01/2023]
Abstract
The tumor microenvironment contributes significantly to tumor initiation, progression, and resistance to chemotherapy. Much of our understanding of the tumor and its microenvironment is developed using various methods of cell culture. Throughout the last two decades, research has increasingly shown that 3D cell culture systems can remarkably recapitulate the complexity of tumor architecture and physiology compared to traditional 2D models. Unlike the flat culture system, these novel models enabled more cell-cell and cell-extracellular matrix interactions. By mimicking in vivo microenvironment, 3D culture systems promise to become accurate tools ready to be used in diagnosis, drug screening, and personalized medicine. In this review, we discussed the importance of 3D culture in simulating the tumor microenvironment and focused on the effects of cancer cell-microenvironment interactions on cancer behavior, resistance, proliferation, and metastasis. Finally, we assessed the role of 3D cell culture systems in the contexts of drug screening. 2D culture system is used to study cancer cell growth, progression, behavior, and drug response. It provides contact between cells and supports paracrine crosstalk between host cells and cancer cells. However, this system fails to simulate the architecture and the physiological aspects of in vivo tumor microenvironment due to the absence of cell-cell/ cell-ECM interactions as well as unlimited access to O2 and nutrients, and the absence of tumor heterogeneity. Recently advanced research has led researchers to generate 3D culture system that can better recapitulate the in vivo environment by providing hypoxic medium, facilitating cell-cell and cell-ECM, interactions, and recapitulating heterogeneity of the tumor. Several approaches are used to maintain and expand cancer cells in 3D culture systems such as tumor spheroids (cell aggregate that mimics the in vivo growth of tumor cells), scaffold-based approaches, bioreactors, microfluidic derives, and organoids. 3D systems are currently used for disease modeling and pre-clinical drug testing.
Collapse
Affiliation(s)
- Oula El Atat
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Zahra Farzaneh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatima Taki
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Ralph Abi-Habib
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mirvat El-Sibai
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
12
|
Buglakov AI, Vasilevskaya VV. Fibril Assembly and Gelation of Macromolecules with Amphiphilic Repeating Units. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:12377-12387. [PMID: 34637315 DOI: 10.1021/acs.langmuir.1c01953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This paper reports the self-assembly of the fibrillar network in a concentrated solution of macromolecules with an amphiphilic structure of repeating units. The investigation of amphiphilic homopolymers and alternating copolymers with the linear and cyclic topologies, the solution with different polymer concentrations and solvent qualities, allows us to conclude that the ability to form a fibrillar gel with branched fibrils and regular subchain thickness is inherent for macromolecules with the solvophobic backbone and solvophilic pendants. The elements of the gel structure, such as the mesh size and fibrillar thickness, the number of cross-links, and their functionality, can be tuned and customized according to the requirements of their application. The results could be helpful for the directed design of the synthetic analogue of the relevant extracellular matrix, in tissue engineering, for fibrotic disease treatment and cell encapsulation.
Collapse
Affiliation(s)
- Aleksandr I Buglakov
- A. N. Nesmeyanov Institute of Organoelement Compounds RAS, Vavilova ul., 28, Moscow 119991, Russia
- Faculty of Physics, M. V. Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia
| | - Valentina V Vasilevskaya
- A. N. Nesmeyanov Institute of Organoelement Compounds RAS, Vavilova ul., 28, Moscow 119991, Russia
| |
Collapse
|
13
|
Zhang XZ, Jiang YL, Hu JG, Zhao LM, Chen QZ, Liang Y, Zhang Y, Lei XX, Wang R, Lei Y, Zhang QY, Li-Ling J, Xie HQ. Procyanidins-crosslinked small intestine submucosa: A bladder patch promotes smooth muscle regeneration and bladder function restoration in a rabbit model. Bioact Mater 2021; 6:1827-1838. [PMID: 33336114 PMCID: PMC7721664 DOI: 10.1016/j.bioactmat.2020.11.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 02/05/2023] Open
Abstract
Currently the standard surgical treatment for bladder defects is augmentation cystoplasty with autologous tissues, which has many side effects. Biomaterials such as small intestine submucosa (SIS) can provide an alternative scaffold for the repair as bladder patches. Previous studies have shown that SIS could enhance the capacity and compliance of the bladder, but its application is hindered by issues like limited smooth muscle regeneration and stone formation since the fast degradation and poor mechanical properties of the SIS. Procyanidins (PC), a natural bio-crosslinking agent, has shown anti-calcification, anti-inflammatory and anti-oxidation properties. More importantly, PC and SIS can crosslink through hydrogen bonds, which may endow the material with enhanced mechanical property and stabilized functionalities. In this study, various concentrations of PC-crosslinked SIS (PC-SIS) were prepared to repair the full-thickness bladder defects, with an aim to reduce complications and enhance bladder functions. In vitro assays showed that the crosslinking has conferred the biomaterial with superior mechanical property and anti-calcification property, ability to promote smooth muscle cell adhesion and upregulate functional genes expression. Using a rabbit model with bladder defects, we demonstrated that the PC-SIS scaffold can rapidly promote in situ tissue regrowth and regeneration, in particular smooth muscle remodeling and improvement of urinary functions. The PC-SIS scaffold has therefore provided a promising material for the reconstruction of a functional bladder.
Collapse
Affiliation(s)
- Xiu-Zhen Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan-Lin Jiang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jun-Gen Hu
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Long-Mei Zhao
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Qiu-Zhu Chen
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yan Liang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Zhang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiong-Xin Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Rui Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Yi Lei
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Jesse Li-Ling
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Deville SS, Cordes N. The Extracellular, Cellular, and Nuclear Stiffness, a Trinity in the Cancer Resistome-A Review. Front Oncol 2019; 9:1376. [PMID: 31867279 PMCID: PMC6908495 DOI: 10.3389/fonc.2019.01376] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Alterations in mechano-physiological properties of a tissue instigate cancer burdens in parallel to common genetic and epigenetic alterations. The chronological and mechanistic interrelation between the various extra- and intracellular aspects remains largely elusive. Mechano-physiologically, integrins and other cell adhesion molecules present the main mediators for transferring and distributing forces between cells and the extracellular matrix (ECM). These cues are channeled via focal adhesion proteins, termed the focal adhesomes, to cytoskeleton and nucleus and vice versa thereby affecting the pathophysiology of multicellular cancer tissues. In combination with simultaneous activation of diverse downstream signaling pathways, the phenotypes of cancer cells are created and driven characterized by deregulated transcriptional and biochemical cues that elicit the hallmarks of cancer. It, however, remains unclear how elastostatic modifications, i.e., stiffness, in the extracellular, intracellular, and nuclear compartment contribute and control the resistance of cancer cells to therapy. In this review, we discuss how stiffness of unique tumor components dictates therapy response and what is known about the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
15
|
Kim J, Li B, Scheideler OJ, Kim Y, Sohn LL. Visco-Node-Pore Sensing: A Microfluidic Rheology Platform to Characterize Viscoelastic Properties of Epithelial Cells. iScience 2019; 13:214-228. [PMID: 30870780 PMCID: PMC6416673 DOI: 10.1016/j.isci.2019.02.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/26/2019] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Viscoelastic properties of cells provide valuable information regarding biological or clinically relevant cellular characteristics. Here, we introduce a new, electronic-based, microfluidic platform-visco-node-pore sensing (visco-NPS)-which quantifies cellular viscoelastic properties under periodic deformation. We measure the storage (G') and loss (G″) moduli (i.e., elasticity and viscosity, respectively) of cells. By applying a wide range of deformation frequencies, our platform quantifies the frequency dependence of viscoelastic properties. G' and G″ measurements show that the viscoelastic properties of malignant breast epithelial cells (MCF-7) are distinctly different from those of non-malignant breast epithelial cells (MCF-10A). With its sensitivity, visco-NPS can dissect the individual contributions of different cytoskeletal components to whole-cell mechanical properties. Moreover, visco-NPS can quantify the mechanical transitions of cells as they traverse the cell cycle or are initiated into an epithelial-mesenchymal transition. Visco-NPS identifies viscoelastic characteristics of cell populations, providing a biophysical understanding of cellular behavior and a potential for clinical applications.
Collapse
Affiliation(s)
- Junghyun Kim
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, CA, USA
| | - Brian Li
- Graduate Program in Bioengineering, University of California, Berkeley, University of California, San Francisco, Berkeley, CA, USA
| | - Olivia J Scheideler
- Graduate Program in Bioengineering, University of California, Berkeley, University of California, San Francisco, Berkeley, CA, USA
| | - Youngbin Kim
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA, USA
| | - Lydia L Sohn
- Department of Mechanical Engineering, University of California at Berkeley, Berkeley, CA, USA; Graduate Program in Bioengineering, University of California, Berkeley, University of California, San Francisco, Berkeley, CA, USA.
| |
Collapse
|
16
|
Onochie OE, Zollinger A, Rich CB, Smith M, Trinkaus-Randall V. Epithelial cells exert differential traction stress in response to substrate stiffness. Exp Eye Res 2019; 181:25-37. [PMID: 30653966 DOI: 10.1016/j.exer.2019.01.014] [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: 10/24/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 10/27/2022]
Abstract
Epithelial wound healing is essential for maintaining the function and clarity of the cornea. Successful repair after injury involves the coordinated movements of cell sheets over the wounded region. While collective migration has been the focus of studies, the effects that environmental changes have on this form of movement are poorly understood. To examine the role of substrate compliancy on multi-layered epithelial sheet migration, we performed traction force and confocal microscopy to determine differences in traction forces and to examine focal adhesions on synthetic and biological substrates. The leading edges of corneal epithelial sheets undergo retraction or contraction prior to migration, and alterations in the sheet's stiffness are affected by the amount of force exerted by cells at the leading edge. On substrates of 30 kPa, cells exhibited greater and more rapid movement than on substrates of 8 kPa, which are similar to that of the corneal basement membrane. Vinculin and its phosphorylated residue Y1065 were prominent along the basal surface of migrating cells, while Y822 was prominent between neighboring cells along the leading edge. Vinculin localization was diffuse on a substrate where the basement membrane was removed. Furthermore, when cells were cultured on fibronectin-coated acrylamide substrates of 8 and 50 kPa and then wounded, there was an injury-induced phosphorylation of Y1065 and substrate dependent changes in the number and size of vinculin containing focal adhesions. These results demonstrate that changes in substrate stiffness affected traction forces and vinculin dynamics, which potentially could contribute to the delayed healing response associated with certain corneal pathologies.
Collapse
Affiliation(s)
- Obianamma E Onochie
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Alicia Zollinger
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Celeste B Rich
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Michael Smith
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Vickery Trinkaus-Randall
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
17
|
Knox A. Arterial Aging, Metalloproteinase Regulation, and the Potential of Resistance Exercise. Curr Cardiol Rev 2018; 14:227-232. [PMID: 30068282 PMCID: PMC6300795 DOI: 10.2174/1573403x14666180801153801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/04/2018] [Accepted: 06/14/2018] [Indexed: 12/11/2022] Open
Abstract
Background Aging is a process that affects all living organisms. The transition through life elicits tissue specific alterations in the functional and structural capabilities of all physiological systems. In particular, the vasculature is vulnerable to aging specific adaptations which induces morphological changes and ultimately increases the risk of pathological states. Matrix metalloproteinases are a group of extremely active enzymes that regulate the age-associated structural changes of the vasculature which has been regarded as the hallmark of arterial aging. Although this process in unavoidable, the structural and functional changes to the vasculature that occur as a result of advancing age can be positively or negatively influenced by our lifestyle choices. Conclusion Exercise training has profound effects on the age-associated changes of the arteries which have been shown to be beneficial in offsetting the detrimental responses of aging. This review provides a brief synopsis of the matrix metalloproteinase induced alterations of the arteria during aging and highlights the potential of resistance exercise to influence such changes.
Collapse
Affiliation(s)
- Allan Knox
- Exercise Science Department, California Lutheran University, 60 Olsen Road, Thousand Oaks, CA, United States
| |
Collapse
|
18
|
Noninvasive Imaging: Brillouin Confocal Microscopy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1092:351-364. [DOI: 10.1007/978-3-319-95294-9_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
19
|
Nichols AEC, Settlage RE, Werre SR, Dahlgren LA. Novel roles for scleraxis in regulating adult tenocyte function. BMC Cell Biol 2018; 19:14. [PMID: 30086712 PMCID: PMC6081934 DOI: 10.1186/s12860-018-0166-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022] Open
Abstract
Background Tendinopathies are common and difficult to resolve due to the formation of scar tissue that reduces the mechanical integrity of the tissue, leading to frequent reinjury. Tenocytes respond to both excessive loading and unloading by producing pro-inflammatory mediators, suggesting that these cells are actively involved in the development of tendon degeneration. The transcription factor scleraxis (Scx) is required for the development of force-transmitting tendon during development and for mechanically stimulated tenogenesis of stem cells, but its function in adult tenocytes is less well-defined. The aim of this study was to further define the role of Scx in mediating the adult tenocyte mechanoresponse. Results Equine tenocytes exposed to siRNA targeting Scx or a control siRNA were maintained under cyclic mechanical strain before being submitted for RNA-seq analysis. Focal adhesions and extracellular matrix-receptor interaction were among the top gene networks downregulated in Scx knockdown tenocytes. Correspondingly, tenocytes exposed to Scx siRNA were significantly softer, with longer vinculin-containing focal adhesions, and an impaired ability to migrate on soft surfaces. Other pathways affected by Scx knockdown included increased oxidative phosphorylation and diseases caused by endoplasmic reticular stress, pointing to a larger role for Scx in maintaining tenocyte homeostasis. Conclusions Our study identifies several novel roles for Scx in adult tenocytes, which suggest that Scx facilitates mechanosensing by regulating the expression of several mechanosensitive focal adhesion proteins. Furthermore, we identified a number of other pathways and targets affected by Scx knockdown that have the potential to elucidate the role that tenocytes may play in the development of degenerative tendinopathy. Electronic supplementary material The online version of this article (10.1186/s12860-018-0166-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne E C Nichols
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA, 24061-0442, USA
| | - Robert E Settlage
- Advanced Research Computing, Virginia Biocomplexity Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Stephen R Werre
- Laboratory for Study Design and Statistical Analysis, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Linda A Dahlgren
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, 205 Duck Pond Drive, Blacksburg, VA, 24061-0442, USA.
| |
Collapse
|
20
|
McKenzie AJ, Hicks SR, Svec KV, Naughton H, Edmunds ZL, Howe AK. The mechanical microenvironment regulates ovarian cancer cell morphology, migration, and spheroid disaggregation. Sci Rep 2018; 8:7228. [PMID: 29740072 PMCID: PMC5940803 DOI: 10.1038/s41598-018-25589-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/24/2018] [Indexed: 01/13/2023] Open
Abstract
There is growing appreciation of the importance of the mechanical properties of the tumor microenvironment on disease progression. However, the role of extracellular matrix (ECM) stiffness and cellular mechanotransduction in epithelial ovarian cancer (EOC) is largely unknown. Here, we investigated the effect of substrate rigidity on various aspects of SKOV3 human EOC cell morphology and migration. Young’s modulus values of normal mouse peritoneum, a principal target tissue for EOC metastasis, were determined by atomic force microscopy (AFM) and hydrogels were fabricated to mimic these values. We find that cell spreading, focal adhesion formation, myosin light chain phosphorylation, and cellular traction forces all increase on stiffer matrices. Substrate rigidity also positively regulates random cell migration and, importantly, directional increases in matrix tension promote SKOV3 cell durotaxis. Matrix rigidity also promotes nuclear translocation of YAP1, an oncogenic transcription factor associated with aggressive metastatic EOC. Furthermore, disaggregation of multicellular EOC spheroids, a behavior associated with dissemination and metastasis, is enhanced by matrix stiffness through a mechanotransduction pathway involving ROCK, actomyosin contractility, and FAK. Finally, this pattern of mechanosensitivity is maintained in highly metastatic SKOV3ip.1 cells. These results establish that the mechanical properties of the tumor microenvironment may play a role in EOC metastasis.
Collapse
Affiliation(s)
- Andrew J McKenzie
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Stephanie R Hicks
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Kathryn V Svec
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Hannah Naughton
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Zöe L Edmunds
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Alan K Howe
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States.
| |
Collapse
|
21
|
Malandrino A, Mak M, Kamm RD, Moeendarbary E. Complex mechanics of the heterogeneous extracellular matrix in cancer. EXTREME MECHANICS LETTERS 2018; 21:25-34. [PMID: 30135864 PMCID: PMC6097546 DOI: 10.1016/j.eml.2018.02.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/07/2018] [Accepted: 02/20/2018] [Indexed: 05/14/2023]
Abstract
The extracellular matrix (ECM) performs many critical functions, one of which is to provide structural and mechanical integrity, and many of the constituent proteins have clear mechanical roles. The composition and structural characteristics of the ECM are widely variable among different tissues, suiting diverse functional needs. In diseased tissues, particularly solid tumors, the ECM is complex and influences disease progression. Cancer and stromal cells can significantly influence the matrix composition and structure and thus the mechanical properties of the tumor microenvironment (TME). In this review, we describe the interactions that give rise to the structural heterogeneity of the ECM and present the techniques that are widely employed to measure ECM properties and remodeling dynamics. Furthermore, we review the tools for measuring the distinct nature of cell-ECM interactions within the TME.
Collapse
Affiliation(s)
- Andrea Malandrino
- Institute for Bioengineering of Catalonia, Barcelona, Spain
- European Molecular Biology Laboratory, Barcelona, Spain
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Roger D. Kamm
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emad Moeendarbary
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, University College London, London, UK
| |
Collapse
|
22
|
Karki P, Birukova AA. Substrate stiffness-dependent exacerbation of endothelial permeability and inflammation: mechanisms and potential implications in ALI and PH (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018773044. [PMID: 29714090 PMCID: PMC5987909 DOI: 10.1177/2045894018773044] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The maintenance of endothelial barrier integrity is absolutely essential to prevent the vascular leak associated with pneumonia, pulmonary edema resulting from inhalation of toxins, acute elevation to high altitude, traumatic and septic lung injury, acute lung injury (ALI), and its life-threatening complication, acute respiratory distress syndrome (ARDS). In addition to the long-known edemagenic and inflammatory agonists, emerging evidences suggest that factors of endothelial cell (EC) mechanical microenvironment such as blood flow, mechanical strain of the vessel, or extracellular matrix stiffness also play an essential role in the control of endothelial permeability and inflammation. Recent studies from our group and others have demonstrated that substrate stiffening causes endothelial barrier disruption and renders EC more susceptible to agonist-induced cytoskeletal rearrangement and inflammation. Further in vivo studies have provided direct evidence that proinflammatory stimuli increase lung microvascular stiffness which in turn exacerbates endothelial permeability and inflammation and perpetuates a vicious circle of lung inflammation. Accumulating evidence suggests a key role for RhoA GTPases signaling in stiffness-dependent mechanotransduction mechanisms defining EC permeability and inflammatory responses. Vascular stiffening is also known to be a key contributor to other cardiovascular diseases such as arterial pulmonary hypertension (PH), although the precise role of stiffness in the development and progression of PH remains to be elucidated. This review summarizes the current understanding of stiffness-dependent regulation of pulmonary EC permeability and inflammation, and discusses potential implication of pulmonary vascular stiffness alterations at macro- and microscale in development and modulation of ALI and PH.
Collapse
Affiliation(s)
- Pratap Karki
- 12264 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland Baltimore, School of Medicine, Baltimore, MD, USA
| | - Anna A Birukova
- 12264 Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland Baltimore, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Liu CH, Nevozhay D, Schill A, Singh M, Das S, Nair A, Han Z, Aglyamov S, Larin KV, Sokolov KV. Nanobomb optical coherence elastography. OPTICS LETTERS 2018; 43:2006-2009. [PMID: 29714732 PMCID: PMC5973512 DOI: 10.1364/ol.43.002006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/17/2018] [Indexed: 05/18/2023]
Abstract
Wave-based optical elastography is rapidly emerging as a powerful technique for quantifying tissue biomechanical properties due to its noninvasive nature and high displacement sensitivity. However, current approaches are limited in their ability to produce high-frequency waves and highly localized mechanical stress. In this Letter, we demonstrate that the rapid liquid-to-gas phase transition of dye-loaded perfluorocarbon nanodroplets ("nanobombs") initiated by a pulsed laser can produce highly localized, high-frequency, and broadband elastic waves. The waves were detected by an ultra-fast line-field low-coherence holography system. For comparison, we also excited waves using a focused micro-air-pulse. Results from tissue-mimicking phantoms showed that the nanobombs produced elastic waves with frequencies up to ∼9 kHz, which was much greater than the ∼2 kHz waves excited by the air-pulse. Consequently, the nanobombs enabled more accurate quantification of sample viscoelasticity. Combined with their potential for functionalization, the nanobombs show promise for accurate and highly specific noncontact all-optical elastography.
Collapse
Affiliation(s)
- Chih-Hao Liu
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
| | - Dmitry Nevozhay
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- School of Biomedicine, Far Eastern Federal University, Vladivostok, 690090, Russian Federation
| | - Alexander Schill
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
| | - Manmohan Singh
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
| | - Susobhan Das
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
| | - Achuth Nair
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
| | - Zhaolong Han
- School of Naval Architecture, Ocean and Civil Engineering, Shanghai Jiao Tong University, P. R. China
| | - Salavat Aglyamov
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
| | - Kirill V. Larin
- Department of Biomedical Engineering, University of Houston, Texas, 77204, USA
- Interdisciplinary Laboratory of Biophotonics, Tomsk State University, Russian Federation
| | - Konstantin V. Sokolov
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
- Department of Bioengineering, Rice University, Texas, 77030, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
24
|
Chengappa P, Sao K, Jones TM, Petrie RJ. Intracellular Pressure: A Driver of Cell Morphology and Movement. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 337:185-211. [PMID: 29551161 DOI: 10.1016/bs.ircmb.2017.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intracellular pressure, generated by actomyosin contractility and the directional flow of water across the plasma membrane, can rapidly reprogram cell shape and behavior. Recent work demonstrates that cells can generate intracellular pressure with a range spanning at least two orders of magnitude; significantly, pressure is implicated as an important regulator of cell dynamics, such as cell division and migration. Changes to intracellular pressure can dictate the mechanisms by which single human cells move through three-dimensional environments. In this review, we chronicle the classic as well as recent evidence demonstrating how intracellular pressure is generated and maintained in metazoan cells. Furthermore, we highlight how this potentially ubiquitous physical characteristic is emerging as an important driver of cell morphology and behavior.
Collapse
Affiliation(s)
| | - Kimheak Sao
- Drexel University, Philadelphia, PA, United States
| | - Tia M Jones
- Drexel University, Philadelphia, PA, United States
| | | |
Collapse
|
25
|
Ahadian S, Civitarese R, Bannerman D, Mohammadi MH, Lu R, Wang E, Davenport-Huyer L, Lai B, Zhang B, Zhao Y, Mandla S, Korolj A, Radisic M. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv Healthc Mater 2018; 7. [PMID: 29034591 DOI: 10.1002/adhm.201700506] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/15/2017] [Indexed: 12/11/2022]
Abstract
Significant advances in biomaterials, stem cell biology, and microscale technologies have enabled the fabrication of biologically relevant tissues and organs. Such tissues and organs, referred to as organ-on-a-chip (OOC) platforms, have emerged as a powerful tool in tissue analysis and disease modeling for biological and pharmacological applications. A variety of biomaterials are used in tissue fabrication providing multiple biological, structural, and mechanical cues in the regulation of cell behavior and tissue morphogenesis. Cells derived from humans enable the fabrication of personalized OOC platforms. Microscale technologies are specifically helpful in providing physiological microenvironments for tissues and organs. In this review, biomaterials, cells, and microscale technologies are described as essential components to construct OOC platforms. The latest developments in OOC platforms (e.g., liver, skeletal muscle, cardiac, cancer, lung, skin, bone, and brain) are then discussed as functional tools in simulating human physiology and metabolism. Future perspectives and major challenges in the development of OOC platforms toward accelerating clinical studies of drug discovery are finally highlighted.
Collapse
Affiliation(s)
- Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Robert Civitarese
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Dawn Bannerman
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Rick Lu
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Erika Wang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Locke Davenport-Huyer
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Ben Lai
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Boyang Zhang
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Serena Mandla
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto M5S 3G9 Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto M5S 3G9 Ontario Canada
| |
Collapse
|
26
|
Coleman DT, Gray AL, Stephens CA, Scott ML, Cardelli JA. Repurposed drug screen identifies cardiac glycosides as inhibitors of TGF-β-induced cancer-associated fibroblast differentiation. Oncotarget 2017; 7:32200-9. [PMID: 27058757 PMCID: PMC5078007 DOI: 10.18632/oncotarget.8609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/14/2016] [Indexed: 12/22/2022] Open
Abstract
The tumor microenvironment, primarily composed of myofibroblasts, directly influences the progression of solid tumors. Through secretion of growth factors, extracellular matrix deposition, and contractile mechanotransduction, myofibroblasts, or cancer-associated fibroblasts (CAFs), support angiogenesis and cancer cell invasion and metastasis. The differentiation of fibroblasts to CAFs is primarily induced by TGF-β from cancer cells. To discover agents capable of blocking CAF differentiation, we developed a high content immunofluorescence-based assay to screen repurposed chemical libraries utilizing fibronectin expression as an initial CAF marker. Screening of the Prestwick chemical library and NIH Clinical Collection repurposed drug library, totaling over 1700 compounds, identified cardiac glycosides as particularly potent CAF blocking agents. Cardiac glycosides are traditionally used to regulate intracellular calcium by inhibiting the Na+/K+ ATPase to control cardiac contractility. Herein, we report that multiple cardiac glycoside compounds, including digoxin, are able to inhibit TGF-β-induced fibronectin expression at low nanomolar concentrations without undesirable cell toxicity. We found this inhibition to hold true for multiple fibroblast cell lines. Using real-time qPCR, we determined that digoxin prevented induction of multiple CAF markers. Furthermore, we report that digoxin is able to prevent TGF-β-induced fibroblast contraction of extracellular matrix, a major phenotypic consequence of CAF differentiation. Assessing the mechanism of inhibition, we found digoxin reduced SMAD promoter activity downstream of TGF-β, and we provide data that the effect is through inhibition of its known target, the Na+/K+ ATPase. These findings support a critical role for calcium signaling during CAF differentiation and highlight a novel, repurposable modality for cancer therapy.
Collapse
Affiliation(s)
- David T Coleman
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Alana L Gray
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Charles A Stephens
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Matthew L Scott
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - James A Cardelli
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| |
Collapse
|
27
|
Aoki FG, Moriya HT. Mechanical Evaluation of Tracheal Grafts on Different Scales. Artif Organs 2017; 42:476-483. [PMID: 29226358 DOI: 10.1111/aor.13063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/18/2017] [Accepted: 09/28/2017] [Indexed: 12/12/2022]
Abstract
Tissue engineered (or bioengineered) tracheas are alternative options under investigation when the resection with end-to-end anastomosis cannot be performed. One approach to develop bioengineered tracheas is a complex process that involves the use of decellularized tissue scaffolds, followed by recellularization in custom-made tracheal bioreactors. Tracheas withstand pressure variations and their biomechanics are of great importance so that they do not collapse during respiration, although there has been no preferred method of mechanical assay of tracheas among several laboratories over the years. These methods have been performed in segments or whole tracheas and in different species of mammals. This article aims to present some methods used by different research laboratories to evaluate the mechanics of tracheal grafts and presents the importance of the tracheal biomechanics in both macro and micro scales. If bioengineered tracheas become a reality in hospitals in the next few years, the standardization of biomechanical parameters will be necessary for greater consistency of results before transplantations.
Collapse
Affiliation(s)
- Fabio Gava Aoki
- Biomedical Engineering Laboratory, University of São Paulo, Escola Politécnica, São Paulo, Brazil
| | - Henrique Takachi Moriya
- Biomedical Engineering Laboratory, University of São Paulo, Escola Politécnica, São Paulo, Brazil
| |
Collapse
|
28
|
Carroll SF, Buckley CT, Kelly DJ. Cyclic Tensile Strain Can Play a Role in Directing both Intramembranous and Endochondral Ossification of Mesenchymal Stem Cells. Front Bioeng Biotechnol 2017; 5:73. [PMID: 29230389 PMCID: PMC5712005 DOI: 10.3389/fbioe.2017.00073] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/02/2017] [Indexed: 01/13/2023] Open
Abstract
Successfully regenerating damaged or diseased bone and other joint tissues will require a detailed understanding of how joint specific environmental cues regulate the fate of progenitor cells that are recruited or delivered to the site of injury. The goal of this study was to explore the role of cyclic tensile strain (CTS) in regulating the initiation of mesenchymal stem cell/multipotent stromal cell (MSC) differentiation, and specifically their progression along the endochondral pathway. To this end, we first explored the influence of CTS on the differentiation of MSCs in the absence of any specific growth factor, and secondly, we examined the influence of the long-term application of this mechanical stimulus on markers of endochondral ossification in MSCs maintained in chondrogenic culture conditions. A custom bioreactor was developed to apply uniaxial tensile deformation to bone marrow-derived MSCs encapsulated within physiological relevant 3D fibrin hydrogels. Mechanical loading, applied in the absence of soluble differentiation factors, was found to enhance the expression of both tenogenic (COL1A1) and osteogenic markers (BMP2, RUNX2, and ALPL), while suppressing markers of adipogenesis. No evidence of chondrogenesis was observed, suggesting that CTS can play a role in initiating direct intramembranous ossification. During long-term culture in the presence of a chondrogenic growth factor, CTS was shown to induce MSC re-organization and alignment, increase proteoglycan and collagen production, and to enhance the expression of markers associated with endochondral ossification (BMP2, RUNX2, ALPL, OPN, and COL10A1) in a strain magnitude-dependent manner. Taken together, these findings indicate that tensile loading may play a key role in promoting both intramembranous and endochondral ossification of MSCs in a context-dependent manner. In both cases, this loading-induced promotion of osteogenesis was correlated with an increase in the expression of the osteogenic growth factor BMP2. The results of this study demonstrate the potent role that extrinsic mechanical loading plays in guiding stem cell fate, which must be carefully considered when designing cell and tissue-engineering therapies if they are to realize their clinical potential.
Collapse
Affiliation(s)
- Simon F. Carroll
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Conor T. Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
29
|
Sala RL, Kwon MY, Kim M, Gullbrand SE, Henning EA, Mauck RL, Camargo ER, Burdick JA. * Thermosensitive Poly(N-vinylcaprolactam) Injectable Hydrogels for Cartilage Tissue Engineering. Tissue Eng Part A 2017; 23:935-945. [PMID: 28384053 PMCID: PMC5610396 DOI: 10.1089/ten.tea.2016.0464] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 02/16/2017] [Indexed: 11/13/2022] Open
Abstract
Injectable hydrogels have gained prominence in the field of tissue engineering for minimally invasive delivery of cells for tissue repair and in the filling of irregular defects. However, many injectable hydrogels exhibit long gelation times or are not stable for long periods after injection. To address these concerns, we used thermosensitive poly(N-vinylcaprolactam) (PNVCL) hydrogels due to their cytocompatibility and fast response to temperature stimuli. Changes in the PNVCL molecular weight and concentration enabled the development of hydrogels with tunable mechanical properties and fast gelation times (<60 s when the temperature was raised from room temperature to physiologic temperature). Chondrocytes (CHs) and mesenchymal stem cells were encapsulated in PNVCL hydrogels and exhibited high viability (∼90%), as monitored by Live/Dead staining and Alamar Blue assays. Three-dimensional constructs of CH-laden PNVCL hydrogels supported cartilage-specific extracellular matrix production both in vitro and after subcutaneous injection in nude rats for up to 8 weeks. Moreover, biochemical analyses of constructs demonstrated a time-dependent increase in glycosaminoglycans (GAGs) and collagen, which were significantly augmented in the implants cultured in vivo. Histological analyses also demonstrated regular distribution of synthesized cartilage components, including abundant GAGs and type II collagen. The findings from this study demonstrate thermosensitive PNVCL as a candidate injectable biomaterial to deliver cells for cartilage tissue engineering.
Collapse
Affiliation(s)
- Renata L. Sala
- Interdisciplinary Laboratory of Electrochemistry and Ceramics, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mi Y. Kwon
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Minwook Kim
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Translational Musculoskeletal Research Center, CMC VA Medical Center, Philadelphia, Pennsylvania
| | - Sarah E. Gullbrand
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Translational Musculoskeletal Research Center, CMC VA Medical Center, Philadelphia, Pennsylvania
| | - Elizabeth A. Henning
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Translational Musculoskeletal Research Center, CMC VA Medical Center, Philadelphia, Pennsylvania
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Translational Musculoskeletal Research Center, CMC VA Medical Center, Philadelphia, Pennsylvania
| | - Emerson R. Camargo
- Interdisciplinary Laboratory of Electrochemistry and Ceramics, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Stires JC, Latz MI. Contribution of the cytoskeleton to mechanosensitivity reported by dinoflagellate bioluminescence. Cytoskeleton (Hoboken) 2017; 75:12-21. [PMID: 28771965 DOI: 10.1002/cm.21392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/12/2017] [Accepted: 08/01/2017] [Indexed: 01/24/2023]
Abstract
The cytoskeleton is crucial to cell mechanics and sensing the extracellular physical environment. The objective of this study was to examine the role of the cortical cytoskeleton in mechanosensitivity in a unicellular protist, the marine dinoflagellate Lingulodinium polyedra, using its intrinsic bioluminescence as a rapid reporter of mechanotransduction. Pharmacological treatments resolved effects due to immediate cytoskeleton disruption from those due to cytoskeletal remodeling during the light to dark phase transition. The cytoskeleton was visualized by confocal laser scanning microscopy of immunohistochemically labeled microtubules and phalloidin labeled F-actin, and mechanosensitivity assessed based on the bioluminescence response to mechanical stimulation measured during the dark phase. Latrunculin B treatment after the transition from the light to dark phase resulted in some disruption of cortical F-actin, no observed effect on the cortical microtubules, and partial inhibition of the bioluminescence response. Treatment with oryzalin, which depolarizes microtubules, completely disrupted the microtubule network and cortical F-actin, and partially inhibited bioluminescence. These results demonstrate that cells retain some mechanosensitivity despite a disrupted cytoskeleton; link mechanosensitivity to intact F-actin; show a close connection between F-actin and microtubules comprising the cortical cytoskeleton; confirm a strong contribution of the actin cytoskeleton to the translocation of scintillons, vesicles containing the luminescent chemistry; and support the role of the actin cytoskeleton in the association of scintillons with the vacuole membrane.
Collapse
Affiliation(s)
- J C Stires
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, 92039
| | - M I Latz
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, 92039
| |
Collapse
|
31
|
Abhari RE, Martins JA, Morris HL, Mouthuy PA, Carr A. Synthetic sutures: Clinical evaluation and future developments. J Biomater Appl 2017; 32:410-421. [PMID: 28714329 DOI: 10.1177/0885328217720641] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Today's sutures are the result of a 4000-year innovation process with regard to their materials and manufacturing techniques, yet little has been done to enhance the therapeutic value of the suture itself. In this review, we explore the historical development, regulatory database and clinical literature of sutures to gain a fuller picture of suture advances to date. First, we examine historical shifts in suture manufacturing companies and review suture regulatory databases to understand the forces driving suture development. Second, we gather the existing clinical evidence of suture efficacy from reviewing the clinical literature and the Food and Drug Administration database in order to identify to what extent sutures have been clinically evaluated and the key clinical areas that would benefit from improved suture materials. Finally, we apply tissue engineering and regenerative medicine design hypotheses to suture materials to identify routes by which bioactive sutures can be designed and passed through regulatory hurdles, to improve surgical outcomes. Our review of the clinical literature revealed that many of the sutures currently in use have been available for decades, yet have never been clinically evaluated. Since suture design and development is industry driven, incremental modifications have allowed for a steady outflow of products while maintaining a safe regulatory position and limiting costs. Until recently, there has been little academic interest in suture development, however the rise of regenerative medicine strategies is shifting the suture paradigm from an inert material, which mechanically approximates tissue, to a bioactive material, which also actively promotes cell-directed repair and a positive healing response. These materials hold significant therapeutic potential, but could be associated with an increased regulatory burden, cost, and clinical evaluation compared with current devices.
Collapse
Affiliation(s)
- Roxanna E Abhari
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Old Road, Oxford, UK
| | - Joana A Martins
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Old Road, Oxford, UK
| | - Hayley L Morris
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Old Road, Oxford, UK
| | - Pierre-Alexis Mouthuy
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Old Road, Oxford, UK
| | - Andrew Carr
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Old Road, Oxford, UK
| |
Collapse
|
32
|
Ma K, Fan Y, Dong X, Dong D, Guo Y, Wei X, Ning J, Geng Q, Wang C, Hu Y, Li M, Niu W, Li E, Wu Y. MTA1 promotes epithelial to mesenchymal transition and metastasis in non-small-cell lung cancer. Oncotarget 2017; 8:38825-38840. [PMID: 28418915 PMCID: PMC5503575 DOI: 10.18632/oncotarget.16404] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/27/2017] [Indexed: 12/18/2022] Open
Abstract
The present study assessed the role of metastasis-associated protein 1 (MTA1) in epithelial to mesenchymal transition (EMT) and metastasis in non-small-cell lung cancer (NSCLC) cells using a normal lung epithelium cell line, three NSCLC cell lines, a mouse NSCLC model, and 56 clinical NSCLC samples. We observed that MTA1 overexpression decreased cellular adhesion, promoted migration and invasion, and changed cytoskeletal polarity. MTA1 knockdown had the opposite effects. MTA1 overexpression decreased E-cadherin, Claudin-1, and ZO-1 levels and increased Vimentin expression in vitro and in vivo, through activation of AKT/GSK3β/β-catenin signaling. However, treatment with the AKT inhibitor MK2206 did not completely rescue effects associated with MTA1 expression changes, indicating that pathways other than the AKT/GSK3β/β-catenin pathway could be involved in MTA1-induced EMT. Compared with normal lung tissues, MTA1 expression was elevated in NSCLC patient tissues and was correlated with American Joint Committee on Cancer stage, T stage, lymphatic metastasis, and patient overall survival. Additionally, MTA1 expression was positively associated with p-AKT and cytoplasmic β-catenin levels. These findings indicate MTA1 promotes NSCLC cell EMT and metastasis via AKT/GSK3β/β-catenin signaling, which suggests MTA1 may be an effective anti-NSCLC therapeutic target.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Antigens, CD
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/secondary
- Cell Adhesion
- Cell Movement
- Cell Proliferation
- Epithelial-Mesenchymal Transition
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Glycogen Synthase Kinase 3 beta/genetics
- Glycogen Synthase Kinase 3 beta/metabolism
- Histone Deacetylases/genetics
- Histone Deacetylases/metabolism
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lymphatic Metastasis
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Prognosis
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Survival Rate
- Trans-Activators
- Tumor Cells, Cultured
- Vimentin/genetics
- Vimentin/metabolism
- Xenograft Model Antitumor Assays
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Ke Ma
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yangwei Fan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xuyuan Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Danfeng Dong
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuyan Guo
- Department of Medical Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Wei
- Department of Medical Oncology, Shaanxi Province People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Jing Ning
- Department of Obstetrics and Gynecology, Xi'an Third Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Qianqian Geng
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Chuying Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuan Hu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Mengya Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenxia Niu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yinying Wu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
33
|
Neyazi B, Tanrikulu L, Wilkens L, Hartmann C, Stein KP, Dumitru CA, Sandalcioglu IE. Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase 2 Expression in Brain Arteriovenous Malformations and its Association with Brain Arteriovenous Malformation Size. World Neurosurg 2017; 102:79-84. [DOI: 10.1016/j.wneu.2017.02.116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 10/20/2022]
|
34
|
Kräter M, Jacobi A, Otto O, Tietze S, Müller K, Poitz DM, Palm S, Zinna VM, Biehain U, Wobus M, Chavakis T, Werner C, Guck J, Bornhauser M. Bone marrow niche-mimetics modulate HSPC function via integrin signaling. Sci Rep 2017; 7:2549. [PMID: 28566689 PMCID: PMC5451425 DOI: 10.1038/s41598-017-02352-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/10/2017] [Indexed: 12/25/2022] Open
Abstract
The bone marrow (BM) microenvironment provides critical physical cues for hematopoietic stem and progenitor cell (HSPC) maintenance and fate decision mediated by cell-matrix interactions. However, the mechanisms underlying matrix communication and signal transduction are less well understood. Contrary, stem cell culture is mainly facilitated in suspension cultures. Here, we used bone marrow-mimetic decellularized extracellular matrix (ECM) scaffolds derived from mesenchymal stromal cells (MSCs) to study HSPC-ECM interaction. Seeding freshly isolated HSPCs adherent (AT) and non-adherent (SN) cells were found. We detected enhanced expansion and active migration of AT-cells mediated by ECM incorporated stromal derived factor one. Probing cell mechanics, AT-cells displayed naïve cell deformation compared to SN-cells indicating physical recognition of ECM material properties by focal adhesion. Integrin αIIb (CD41), αV (CD51) and β3 (CD61) were found to be induced. Signaling focal contacts via ITGβ3 were identified to facilitate cell adhesion, migration and mediate ECM-physical cues to modulate HSPC function.
Collapse
Affiliation(s)
- Martin Kräter
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
| | - Angela Jacobi
- Biotechnology Center, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Oliver Otto
- Centre for Innovation Competence - Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, Greifswald, Mecklenburg-Western Pomerania, 17489, Germany
| | - Stefanie Tietze
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Katrin Müller
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
| | - David M Poitz
- Department of Internal Medicine and Cardiology, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Sandra Palm
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
| | - Valentina M Zinna
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
| | - Ulrike Biehain
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
| | - Manja Wobus
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials, Dresden, Saxony, 01307, Germany
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, Dresden, Saxony, 01307, Germany
| | - Martin Bornhauser
- Medical Clinic I, University Hospital Carl Gustav Carus, Dresden, Saxony, 01307, Germany.
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Saxony, 01307, Germany.
| |
Collapse
|
35
|
Teixeira SC, Lopes DS, Gimenes SNC, Teixeira TL, da Silva MS, Brígido RTES, da Luz FAC, da Silva AA, Silva MA, Florentino PV, Tavares PCB, dos Santos MA, Ávila VDMR, Silva MJB, Elias MC, Mortara RA, da Silva CV. Mechanistic Insights into the Anti-angiogenic Activity of Trypanosoma cruzi Protein 21 and its Potential Impact on the Onset of Chagasic Cardiomyopathy. Sci Rep 2017; 7:44978. [PMID: 28322302 PMCID: PMC5359584 DOI: 10.1038/srep44978] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/17/2017] [Indexed: 12/31/2022] Open
Abstract
Chronic chagasic cardiomyopathy (CCC) is arguably the most important form of the Chagas Disease, caused by the intracellular protozoan Trypanosoma cruzi; it is estimated that 10-30% of chronic patients develop this clinical manifestation. The most common and severe form of CCC can be related to ventricular abnormalities, such as heart failure, arrhythmias, heart blocks, thromboembolic events and sudden death. Therefore, in this study, we proposed to evaluate the anti-angiogenic activity of a recombinant protein from T. cruzi named P21 (rP21) and the potential impact of the native protein on CCC. Our data suggest that the anti-angiogenic activity of rP21 depends on the protein's direct interaction with the CXCR4 receptor. This capacity is likely related to the modulation of the expression of actin and angiogenesis-associated genes. Thus, our results indicate that T. cruzi P21 is an attractive target for the development of innovative therapeutic agents against CCC.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Daiana Silva Lopes
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, MG, Brasil
| | - Sarah Natalie Cirilo Gimenes
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, MG, Brasil
| | - Thaise Lara Teixeira
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Marcelo Santos da Silva
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, São Paulo, Brasil
| | - Rebecca Tavares e Silva Brígido
- Laboratório de Patologia Molecular e Biotecnologia do Centro de Referência Nacional em Dermatologia Sanitária/Hanseníase, Faculdade de Medicina, Universidade Federal de Uberlândia, MG, Brasil
| | - Felipe Andrés Cordero da Luz
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Aline Alves da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Makswell Almeida Silva
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, MG, Brasil
| | - Pilar Veras Florentino
- Departamento de Microbiologia Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brasil
| | - Paula Cristina Brígido Tavares
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Marlus Alves dos Santos
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | | | - Marcelo José Barbosa Silva
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| | - Maria Carolina Elias
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, São Paulo, Brasil
| | - Renato Arruda Mortara
- Departamento de Microbiologia Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brasil
| | - Claudio Vieira da Silva
- Laboratório de Tripanosomatídeos, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, MG, Brasil
| |
Collapse
|
36
|
Vuong AT, Rauch AD, Wall WA. A biochemo-mechano coupled, computational model combining membrane transport and pericellular proteolysis in tissue mechanics. Proc Math Phys Eng Sci 2017; 473:20160812. [PMID: 28413347 DOI: 10.1098/rspa.2016.0812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/03/2017] [Indexed: 11/12/2022] Open
Abstract
We present a computational model for the interaction of surface- and volume-bound scalar transport and reaction processes with a deformable porous medium. The application in mind is pericellular proteolysis, i.e. the dissolution of the solid phase of the extracellular matrix (ECM) as a response to the activation of certain chemical species at the cell membrane and in the vicinity of the cell. A poroelastic medium model represents the extra cellular scaffold and the interstitial fluid flow, while a surface-bound transport model accounts for the diffusion and reaction of membrane-bound chemical species. By further modelling the volume-bound transport, we consider the advection, diffusion and reaction of sequestered chemical species within the extracellular scaffold. The chemo-mechanical coupling is established by introducing a continuum formulation for the interplay of reaction rates and the mechanical state of the ECM. It is based on known experimental insights and theoretical work on the thermodynamics of porous media and degradation kinetics of collagen fibres on the one hand and a damage-like effect of the fibre dissolution on the mechanical integrity of the ECM on the other hand. The resulting system of partial differential equations is solved via the finite-element method. To the best of our knowledge, it is the first computational model including contemporaneously the coupling between (i) advection-diffusion-reaction processes, (ii) interstitial flow and deformation of a porous medium, and (iii) the chemo-mechanical interaction impelled by the dissolution of the ECM. Our numerical examples show good agreement with experimental data. Furthermore, we outline the capability of the methodology to extend existing numerical approaches towards a more comprehensive model for cellular biochemo-mechanics.
Collapse
Affiliation(s)
- A-T Vuong
- Institute for Computational Mechanics, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching bei München, Germany
| | - A D Rauch
- Institute for Computational Mechanics, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching bei München, Germany
| | - W A Wall
- Institute for Computational Mechanics, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching bei München, Germany
| |
Collapse
|
37
|
Humphries DL, Grogan JA, Gaffney EA. Mechanical Cell-Cell Communication in Fibrous Networks: The Importance of Network Geometry. Bull Math Biol 2017; 79:498-524. [PMID: 28130739 PMCID: PMC5331102 DOI: 10.1007/s11538-016-0242-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/14/2016] [Indexed: 01/24/2023]
Abstract
Cells contracting in extracellular matrix (ECM) can transmit stress over long distances, communicating their position and orientation to cells many tens of micrometres away. Such phenomena are not observed when cells are seeded on substrates with linear elastic properties, such as polyacrylamide (PA) gel. The ability for fibrous substrates to support far reaching stress and strain fields has implications for many physiological processes, while the mechanical properties of ECM are central to several pathological processes, including tumour invasion and fibrosis. Theoretical models have investigated the properties of ECM in a variety of network geometries. However, the effects of network architecture on mechanical cell-cell communication have received little attention. This work investigates the effects of geometry on network mechanics, and thus the ability for cells to communicate mechanically through different networks. Cell-derived displacement fields are quantified for various network geometries while controlling for network topology, cross-link density and micromechanical properties. We find that the heterogeneity of response, fibre alignment, and substrate displacement fields are sensitive to network choice. Further, we show that certain geometries support mechanical communication over longer distances than others. As such, we predict that the choice of network geometry is important in fundamental modelling of cell-cell interactions in fibrous substrates, as well as in experimental settings, where mechanical signalling at the cellular scale plays an important role. This work thus informs the construction of theoretical models for substrate mechanics and experimental explorations of mechanical cell-cell communication.
Collapse
Affiliation(s)
- D L Humphries
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK.
| | - J A Grogan
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| | - E A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, UK
| |
Collapse
|
38
|
Li H, Xu B, Zhou EH, Sunyer R, Zhang Y. Multiscale Measurements of the Mechanical Properties of Collagen Matrix. ACS Biomater Sci Eng 2017; 3:2815-2824. [DOI: 10.1021/acsbiomaterials.6b00634] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | | | - Enhua H. Zhou
- Ophthalmology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Raimon Sunyer
- Institute for Bioengineering of Catalonia, Baldiri-Reixac 15-21, 08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Av. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain
| | | |
Collapse
|
39
|
Dexter AF, Fletcher N, Creasey RG, Filardo F, Boehm MW, Jack KS. Fabrication and characterization of hydrogels formed from designer coiled-coil fibril-forming peptides. RSC Adv 2017; 7:27260-27271. [DOI: 10.1039/c7ra02811c] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
A peptide sequence was designed to form α-helical fibrils and hydrogels at physiological pH, utilising transient buffering by carbonic acid.
Collapse
Affiliation(s)
- A. F. Dexter
- The University of Queensland
- Australian Institute for Bioengineering and Biotechnology
- Australia
| | - N. L. Fletcher
- The University of Queensland
- Australian Institute for Bioengineering and Biotechnology
- Australia
| | - R. G. Creasey
- The University of Queensland
- School of Chemical Engineering
- Australia
| | - F. Filardo
- The University of Queensland
- Australian Institute for Bioengineering and Biotechnology
- Australia
| | - M. W. Boehm
- The University of Queensland
- School of Chemical Engineering
- Australia
| | - K. S. Jack
- The University of Queensland
- Centre for Microscopy and Microanalysis
- Australia
| |
Collapse
|
40
|
Domeradzka NE, Werten MWT, de Wolf FA, de Vries R. Cross-Linking and Bundling of Self-Assembled Protein-Based Polymer Fibrils via Heterodimeric Coiled Coils. Biomacromolecules 2016; 17:3893-3901. [DOI: 10.1021/acs.biomac.6b01242] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Natalia E. Domeradzka
- Wageningen UR
Food and Biobased Research, 6708 WG Wageningen, The Netherlands
- Physical
Chemistry and Soft Matter, Wageningen University Stippeneng 4, 6708 WE Wageningen, The Netherlands
| | - Marc W. T. Werten
- Wageningen UR
Food and Biobased Research, 6708 WG Wageningen, The Netherlands
| | - Frits A. de Wolf
- Wageningen UR
Food and Biobased Research, 6708 WG Wageningen, The Netherlands
| | - Renko de Vries
- Physical
Chemistry and Soft Matter, Wageningen University Stippeneng 4, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
41
|
Yamaba H, Haba M, Kunita M, Sakaida T, Tanaka H, Yashiro Y, Nakata S. Morphological change of skin fibroblasts induced by UV Irradiation is involved in photoaging. Exp Dermatol 2016; 25 Suppl 3:45-51. [DOI: 10.1111/exd.13084] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Hiroyuki Yamaba
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Manami Haba
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Mayumi Kunita
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Tsutomu Sakaida
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Hiroshi Tanaka
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Youichi Yashiro
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Satoru Nakata
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| |
Collapse
|
42
|
Peñalver Bernabé B, Shin S, Rios PD, Broadbelt LJ, Shea LD, Seidlits SK. Dynamic transcription factor activity networks in response to independently altered mechanical and adhesive microenvironmental cues. Integr Biol (Camb) 2016; 8:844-60. [PMID: 27470442 DOI: 10.1039/c6ib00093b] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple aspects of the local extracellular environment profoundly affect cell phenotype and function. Physical and chemical cues in the environment trigger intracellular signaling cascades that ultimately activate transcription factors (TFs) - powerful regulators of the cell phenotype. TRACER (TRanscriptional Activity CEll aRrays) was employed for large-scale, dynamic quantification of TF activity in human fibroblasts cultured on hydrogels with a controlled elastic modulus and integrin ligand density. We identified three groups of TFs: responders to alterations in ligand density alone, substrate stiffness or both. Dynamic networks of regulatory TFs were constructed computationally and revealed distinct TF activity levels, directionality (i.e., activation or inhibition), and dynamics for adhesive and mechanical cues. Moreover, TRACER networks predicted conserved hubs of TF activity across multiple cell types, which are significantly altered in clinical fibrotic tissues. Our approach captures the distinct and overlapping effects of adhesive and mechanical stimuli, identifying conserved signaling mechanisms in normal and disease states.
Collapse
|
43
|
Peela N, Sam FS, Christenson W, Truong D, Watson AW, Mouneimne G, Ros R, Nikkhah M. A three dimensional micropatterned tumor model for breast cancer cell migration studies. Biomaterials 2015; 81:72-83. [PMID: 26724455 DOI: 10.1016/j.biomaterials.2015.11.039] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/29/2015] [Indexed: 12/14/2022]
Abstract
Breast cancer cell invasion is a highly orchestrated process driven by a myriad of complex microenvironmental stimuli, making it difficult to isolate and assess the effects of biochemical or biophysical cues (i.e. tumor architecture, matrix stiffness) on disease progression. In this regard, physiologically relevant tumor models are becoming instrumental to perform studies of cancer cell invasion within well-controlled conditions. Herein, we explored the use of photocrosslinkable hydrogels and a novel, two-step photolithography technique to microengineer a 3D breast tumor model. The microfabrication process enabled precise localization of cell-encapsulated circular constructs adjacent to a low stiffness matrix. To validate the model, breast cancer cell lines (MDA-MB-231, MCF7) and non-tumorigenic mammary epithelial cells (MCF10A) were embedded separately within the tumor model, all of which maintained high viability throughout the experiments. MDA-MB-231 cells exhibited extensive migratory behavior and invaded the surrounding matrix, whereas MCF7 or MCF10A cells formed clusters that stayed confined within the circular tumor regions. Additionally, real-time cell tracking indicated that the speed and persistence of MDA-MB-231 cells were substantially higher within the surrounding matrix compared to the circular constructs. Z-stack imaging of F-actin/α-tubulin cytoskeletal organization revealed unique 3D protrusions in MDA-MB-231 cells and an abundance of 3D clusters formed by MCF7 and MCF10A cells. Our results indicate that gelatin methacrylate (GelMA) hydrogel, integrated with the two-step photolithography technique, has great promise in the development of 3D tumor models with well-defined architecture and tunable stiffness.
Collapse
Affiliation(s)
- Nitish Peela
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA
| | - Feba S Sam
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA
| | - Wayne Christenson
- Center for Biological Physics, Arizona State University, Tempe, AZ 85287, USA; Department of Physics, Arizona State University, Tempe, AZ 85287, USA
| | - Danh Truong
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA
| | - Adam W Watson
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, Tucson, AZ 85724, USA
| | - Ghassan Mouneimne
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, Tucson, AZ 85724, USA
| | - Robert Ros
- Center for Biological Physics, Arizona State University, Tempe, AZ 85287, USA; Department of Physics, Arizona State University, Tempe, AZ 85287, USA; Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
44
|
Cell type-specific adaptation of cellular and nuclear volume in micro-engineered 3D environments. Biomaterials 2015; 69:121-32. [DOI: 10.1016/j.biomaterials.2015.08.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 12/14/2022]
|
45
|
Greene T, Lin CC. Modular Cross-Linking of Gelatin-Based Thiol–Norbornene Hydrogels for in Vitro 3D Culture of Hepatocellular Carcinoma Cells. ACS Biomater Sci Eng 2015; 1:1314-1323. [DOI: 10.1021/acsbiomaterials.5b00436] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Tanja Greene
- Department
of Biomedical
Engineering, Indiana University—Purdue University Indianapolis, Indianapolis, Indiana, 46202 United States
| | - Chien-Chi Lin
- Department
of Biomedical
Engineering, Indiana University—Purdue University Indianapolis, Indianapolis, Indiana, 46202 United States
| |
Collapse
|
46
|
Dyson RJ, Green JEF, Whiteley JP, Byrne HM. An investigation of the influence of extracellular matrix anisotropy and cell–matrix interactions on tissue architecture. J Math Biol 2015; 72:1775-809. [DOI: 10.1007/s00285-015-0927-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 05/21/2015] [Indexed: 12/25/2022]
|
47
|
Kan-Dapaah K, Rahbar N, Tahlil A, Crosson D, Yao N, Soboyejo W. Mechanical and hyperthermic properties of magnetic nanocomposites for biomedical applications. J Mech Behav Biomed Mater 2015; 49:118-28. [DOI: 10.1016/j.jmbbm.2015.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/13/2015] [Accepted: 04/20/2015] [Indexed: 10/23/2022]
|
48
|
Abstract
PURPOSE Regional heterogeneity in mitral annular contraction, which is generally ascribed to the fibrous vs. muscular annular composition, ensures proper leaflet motion and timing of coaptation. It is unknown whether the fibroblast-like cells in the annulus modulate this heterogeneity, even though valvular interstitial cells (VICs) can be mechanically "activated." METHODS Fourteen sheep underwent implantation of radiopaque markers around the mitral annulus defining four segments: septal (SEPT), lateral (LAT), and anterior (ANT-C) and posterior (POST-C) commissures. Segmental annular contraction was calculated using biplane videofluoroscopy. Immunohistochemistry of annular cross sections assessed regional matrix content, matrix turnover, and cell phenotype. Micropipette aspiration measured the Young's modulus of the leaflets adjacent to the myocardial border. RESULTS Whereas SEPT contained more collagen I and III, LAT demonstrated more collagen and elastin turnover as shown by greater decorin, lysyl oxidase, and matrix metalloprotease (MMP)-13 and smooth muscle alpha-actin (SMaA). This greater matrix turnover paralleled greater annular contraction in LAT vs. SEPT (22.5% vs. 4.1%). Similarly, POST-C had more SMaA and MMP13 than ANT-C, consistent with greater annular contraction in POST-C (18.8% vs. 11.1%). Interestingly, POST-C had the greatest effective modulus, significantly higher than LAT. CONCLUSIONS These data suggest that matrix turnover by activated VICs relates to annular motion heterogeneity, maintains steady-state mechanical properties in the annulus, and could be a therapeutic target when annular motion is impaired. Conversely, alterations in this heterogeneous annular contraction, whether through disease or secondary to ring annuloplasty, could disrupt this normal pattern of cell-mediated matrix remodeling and further adversely impact mitral valve function.
Collapse
|
49
|
PANZETTA VALERIA, DE MENNA MARTA, BUCCI DEBORA, GIOVANNINI VITTORIA, PUGLIESE MARIAGABRIELLA, QUARTO MARIA, FUSCO SABATO, NETTI PAOLO. X-RAY IRRADIATION AFFECTS MORPHOLOGY, PROLIFERATION AND MIGRATION RATE OF HEALTHY AND CANCER CELLS. J MECH MED BIOL 2015. [DOI: 10.1142/s0219519415400229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cytoskeleton plays a central role in many cellular processes, such as migration, adhesion and proliferation. Alterations of its structural properties are commonly associated with different diseases (malignancy, cardiac hypertrophy, etc.). In this work, we studied the effects of X-radiations on cytoskeleton architecture of two cell lines: BALBc/3T3 and Simian virus 40-transformed BALBc/3T3 (SVT2) cells. In agreement with the current literature, we observed reduced adhesion and increased motility of SVT2 cells respect to non-transformed BALBc/3T3. In addition, we showed that two different doses of X-rays (1 and 2 Gy) increased cell-dish adhesiveness and reduced cell proliferation and cell motility of transformed cells, whereas minor effects were measured on the normal counterpart. These results suggested that low doses or fractioning of X-rays may have a normalization effect on the investigated parameters for the transformed cell phenotype.
Collapse
Affiliation(s)
- VALERIA PANZETTA
- Center for Advanced Biomaterials for Health Care@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53, 80125 Napoli, Italy
| | - MARTA DE MENNA
- Interdisciplinary Research Centre on Biomaterials, Federico II University of Naples, Piazzale Tecchio, 80, 80126, Napoli, Italy
| | - DEBORA BUCCI
- Interdisciplinary Research Centre on Biomaterials, Federico II University of Naples, Piazzale Tecchio, 80, 80126, Napoli, Italy
| | - VITTORIA GIOVANNINI
- Interdisciplinary Research Centre on Biomaterials, Federico II University of Naples, Piazzale Tecchio, 80, 80126, Napoli, Italy
| | - MARIAGABRIELLA PUGLIESE
- Dipartimento di Fisica, Università Federico II and INFN-Sezione di Napoli, Monte S. Angelo, Via Cintia, 80126 Napoli, Italy
| | - MARIA QUARTO
- Dipartimento di Fisica, Università Federico II and INFN-Sezione di Napoli, Monte S. Angelo, Via Cintia, 80126 Napoli, Italy
| | - SABATO FUSCO
- Center for Advanced Biomaterials for Health Care@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53, 80125 Napoli, Italy
| | - PAOLO NETTI
- Center for Advanced Biomaterials for Health Care@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53, 80125 Napoli, Italy
- Interdisciplinary Research Centre on Biomaterials, Federico II University of Naples, Piazzale Tecchio, 80, 80126, Napoli, Italy
- Dipartimento di Ingegneria dei Materiali e della Produzione, Universita di Napoli Federico II, 'Piazzale Tecchio 80, 80126, Napoli, Italy
| |
Collapse
|
50
|
Chow MJ, Turcotte R, Lin CP, Zhang Y. Arterial extracellular matrix: a mechanobiological study of the contributions and interactions of elastin and collagen. Biophys J 2015; 106:2684-92. [PMID: 24940786 DOI: 10.1016/j.bpj.2014.05.014] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 03/04/2014] [Accepted: 05/05/2014] [Indexed: 01/01/2023] Open
Abstract
The complex network structure of elastin and collagen extracellular matrix (ECM) forms the primary load bearing components in the arterial wall. The structural and mechanobiological interactions between elastin and collagen are important for properly functioning arteries. Here, we examined the elastin and collagen organization, realignment, and recruitment by coupling mechanical loading and multiphoton imaging. Two-photon excitation fluorescence and second harmonic generation methods were performed with a multiphoton video-rate microscope to capture real time changes to the elastin and collagen structure during biaxial deformation. Enzymatic removal of elastin was performed to assess the structural changes of the remaining collagen structure. Quantitative analysis of the structural changes to elastin and collagen was made using a combination of two-dimensional fast Fourier transform and fractal analysis, which allows for a more complete understanding of structural changes. Our study provides new quantitative evidence, to our knowledge on the sequential engagement of different arterial ECM components in response to mechanical loading. The adventitial collagen exists as large wavy bundles of fibers that exhibit fiber engagement after 20% strain. The medial collagen is engaged throughout the stretching process, and prominent elastic fiber engagement is observed up to 20% strain after which the engagement plateaus. The fiber orientation distribution functions show remarkably different changes in the ECM structure in response to mechanical loading. The medial collagen shows an evident preferred circumferential distribution, however the fiber families of adventitial collagen are obscured by their waviness at no or low mechanical strains. Collagen fibers in both layers exhibit significant realignment in response to unequal biaxial loading. The elastic fibers are much more uniformly distributed and remained relatively unchanged due to loading. Removal of elastin produces similar structural changes in collagen as mechanical loading. Our study suggests that the elastic fibers are under tension and impart an intrinsic compressive stress on the collagen.
Collapse
Affiliation(s)
- Ming-Jay Chow
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts
| | - Raphaël Turcotte
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts; Center for Systems Biology, Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Charles P Lin
- Center for Systems Biology, Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yanhang Zhang
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts; Department of Biomedical Engineering, Boston University, Boston, Massachusetts.
| |
Collapse
|