51
|
Chen Y, Guo K, Jiang L, Zhu S, Ni Z, Xiang N. Microfluidic deformability cytometry: A review. Talanta 2022; 251:123815. [DOI: 10.1016/j.talanta.2022.123815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/23/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022]
|
52
|
Ahangar P, Strudwick XL, Cowin AJ. Wound Healing from an Actin Cytoskeletal Perspective. Cold Spring Harb Perspect Biol 2022; 14:a041235. [PMID: 35074864 PMCID: PMC9341468 DOI: 10.1101/cshperspect.a041235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing requires a complex cascade of highly controlled and conserved cellular and molecular processes. These involve numerous cell types and extracellular matrix molecules regulated by the actin cytoskeleton. This microscopic network of filaments is present within the cytoplasm of all cells and provides the shape and mechanical support required for cell movement and proliferation. Here, an overview of the processes of wound healing are described from the perspective of the cell in relation to the actin cytoskeleton. Key points of discussion include the role of actin, its binding proteins, signaling pathways, and events that play significant roles in the phases of wound healing. The identification of cytoskeletal targets that can be used to manipulate and improve wound healing is included as an emerging area of focus that may inform future therapeutic approaches to improve healing of complex wounds.
Collapse
Affiliation(s)
- Parinaz Ahangar
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Allison J Cowin
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| |
Collapse
|
53
|
Zhao Y, Sun Y, Hang R, Yao R, Zhang Y, Huang D, Yao X, Bai L, Hang R. Biocompatible silane adhesion layer on titanium implants improves angiogenesis and osteogenesis. BIOMATERIALS ADVANCES 2022; 139:213033. [PMID: 35882124 DOI: 10.1016/j.bioadv.2022.213033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/02/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
Silane adhesion layer strategy has been widely used to covalently graft biomolecules to the titanium implant surface, thereby conferring the implant bioactivity to ameliorate osseointegration. However, few researchers pay attention to the effects of silanization parameters on biocompatibility and biofunctionality of the silane adhesion layers. Accordingly, the present study successfully fabricated the silane adhesion layers with different thickness, intactness, and surface morphologies by introducing 3-aminopropyltriethoxysilane on the alkali-treated titanium surface in time-varied processing of silanization. The regulatory effects of the silane adhesion layers on angiogenesis and osteogenesis were assessed in vitro. Results showed that the prolonged silanization processing time increased the thickness and intactness of the silane adhesion layer and significantly improved its biocompatibility. Notably, the silane adhesion layer prepared after 12 h of silanization exhibited a brain-like surface morphology and benefited the adhesion and proliferation of endothelial cells (ECs) and osteoblasts (OBs). Moreover, the layer promoted angiogenesis via stimulating vascular endothelial growth factor (VEGF) secretion and nitric oxide (NO) production of ECs. Simultaneously, it improved osteogenesis by enhancing alkaline phosphatase (ALP) activity, collagen secretion, and extracellular matrix mineralization of OBs. This work systematically investigated the biocompatibility and biofunctionality of the modified silane adhesion layers, thus providing valuable references for their application in covalently grafting biomolecules on the titanium implant surface.
Collapse
Affiliation(s)
- Yuyu Zhao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yonghua Sun
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Runhua Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yi Zhang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444 China; Engineering Research Center for Biomedical Materials of Ministry of Education, College of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, China.
| |
Collapse
|
54
|
Liu HH, Xu Y, Li CJ, Hsu SJ, Lin XH, Zhang R, Chen J, Chen J, Gao DM, Cui JF, Yang XR, Ren ZG, Chen RX. An SCD1-dependent mechanoresponsive pathway promotes HCC invasion and metastasis through lipid metabolic reprogramming. Mol Ther 2022; 30:2554-2567. [PMID: 35358687 PMCID: PMC9263248 DOI: 10.1016/j.ymthe.2022.03.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 02/15/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
Matrix stiffness promotes hepatocellular carcinoma (HCC) metastasis. This study examined the contribution of lipid metabolic reprogramming to matrix stiffness-induced HCC metastasis. HCC cells were cultured on mechanically tunable polyacrylamide gels and subjected to lipidomic analysis. The key enzyme that responded to matrix stiffness and regulated lipid metabolism was identified. The comparative lipidomic screening revealed that stearoyl-CoA desaturase 1 (SCD1) is a mechanoresponsive enzyme that reprogrammed HCC cell lipid metabolism. The genetic and pharmacological inhibition of SCD1 expression/activity altered the cellular lipid composition, which in turn impaired plasma membrane fluidity and inhibited in vitro invasive motility of HCC cells in response to high matrix stiffness. Knockdown of SCD1 suppressed HCC invasion and metastasis in vivo. Conversely, the overexpression of SCD1 or exogenous administration of its product oleic acid augmented plasma membrane fluidity and rescued in vitro invasive migration in HCC cells cultured on soft substrates, mimicking the effects imposed by high matrix stiffness. In human HCC tissues, collagen content, a marker of increasing matrix stiffness, and increased expression of SCD1 together predicted poor survival of HCC patients. An SCD1-dependent mechanoresponsive pathway that responds to increasing matrix stiffness in the tumor microenvironment promotes HCC invasion and metastasis through lipid metabolic reprogramming.
Collapse
Affiliation(s)
- Hua-Hua Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Yang Xu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Cao-Jie Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Shu-Jung Hsu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Xia-Hui Lin
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Rui Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Jie Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Jun Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Dong-Mei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Jie-Feng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China
| | - Xin-Rong Yang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China.
| | - Zheng-Gang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Rong-Xin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 180 Fenglin Road, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| |
Collapse
|
55
|
Derisoud E, Jouneau L, Dubois C, Archilla C, Jaszczyszyn Y, Legendre R, Daniel N, Peynot N, Dahirel M, Auclair-Ronzaud J, Wimel L, Duranthon V, Chavatte-Palmer P. Maternal age affects equine day 8 embryo gene expression both in trophoblast and inner cell mass. BMC Genomics 2022; 23:443. [PMID: 35705916 PMCID: PMC9199136 DOI: 10.1186/s12864-022-08593-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Breeding a mare until she is not fertile or even until her death is common in equine industry but the fertility decreases as the mare age increases. Embryo loss due to reduced embryo quality is partly accountable for this observation. Here, the effect of mare's age on blastocysts' gene expression was explored. Day 8 post-ovulation embryos were collected from multiparous young (YM, 6-year-old, N = 5) and older (OM, > 10-year-old, N = 6) non-nursing Saddlebred mares, inseminated with the semen of one stallion. Pure or inner cell mass (ICM) enriched trophoblast, obtained by embryo bisection, were RNA sequenced. Deconvolution algorithm was used to discriminate gene expression in the ICM from that in the trophoblast. Differential expression was analyzed with embryo sex and diameter as cofactors. Functional annotation and classification of differentially expressed genes and gene set enrichment analysis were also performed. RESULTS Maternal aging did not affect embryo recovery rate, embryo diameter nor total RNA quantity. In both compartments, the expression of genes involved in mitochondria and protein metabolism were disturbed by maternal age, although more genes were affected in the ICM. Mitosis, signaling and adhesion pathways and embryo development were decreased in the ICM of embryos from old mares. In trophoblast, ion movement pathways were affected. CONCLUSIONS This is the first study showing that maternal age affects gene expression in the equine blastocyst, demonstrating significant effects as early as 10 years of age. These perturbations may affect further embryo development and contribute to decreased fertility due to aging.
Collapse
Affiliation(s)
- Emilie Derisoud
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France.
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.
| | - Luc Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Cédric Dubois
- IFCE, Plateau technique de Chamberet, 19370, Chamberet, France
| | - Catherine Archilla
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Yan Jaszczyszyn
- Institute for Integrative Biology of the Cell (I2BC), UMR 9198 CNRS, CEA, Paris-Sud University F-91198, Gif-sur-Yvette, France
| | - Rachel Legendre
- Institut Pasteur-Bioinformatics and Biostatistics Hub-Department of Computational Biology, Paris, France
| | - Nathalie Daniel
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Nathalie Peynot
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Michèle Dahirel
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | | | - Laurence Wimel
- IFCE, Plateau technique de Chamberet, 19370, Chamberet, France
| | - Véronique Duranthon
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350, Jouy-en-Josas, France.
- Ecole Nationale Vétérinaire d'Alfort, BREED, 94700, Maisons-Alfort, France.
| |
Collapse
|
56
|
Schmidt H, Mauer K, Hankeln T, Herlyn H. Host-dependent impairment of parasite development and reproduction in the acanthocephalan model. Cell Biosci 2022; 12:75. [PMID: 35642000 PMCID: PMC9153150 DOI: 10.1186/s13578-022-00818-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/19/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND A central question in parasitology is why parasites mature and reproduce in some host species but not in others. Yet, a better understanding of the inability of parasites to complete their life cycles in less suitable hosts may hold clues for their control. To shed light on the molecular basis of parasite (non-)maturation, we analyzed transcriptomes of thorny-headed worms (Acanthocephala: Pomphorhynchus laevis), and compared developmentally arrested worms excised from European eel (Anguilla anguilla) to developmentally unrestricted worms from barbel (Barbus barbus). RESULTS Based on 20 RNA-Seq datasets, we demonstrate that transcriptomic profiles are more similar between P. laevis males and females from eel than between their counterparts from barbel. Impairment of sexual phenotype development was reflected in gene ontology enrichment analyses of genes having differential transcript abundances. Genes having reproduction- and energy-related annotations were found to be affected by parasitizing either eel or barbel. According to this, the molecular machinery of male and female acanthocephalans from the eel is less tailored to reproduction and more to coping with the less suitable environment provided by this host. The pattern was reversed in their counterparts from the definitive host, barbel. CONCLUSIONS Comparative analysis of transcriptomes of developmentally arrested and reproducing parasites elucidates the challenges parasites encounter in hosts which are unsuitable for maturation and reproduction. By studying a gonochoric species, we were also able to highlight sex-specific traits. In fact, transcriptomic evidence for energy shortage in female acanthocephalans associates with their larger body size. Thus, energy metabolism and glycolysis should be promising targets for the treatment of acanthocephaliasis. Although inherently enabling a higher resolution in heterosexuals, the comparison of parasites from definitive hosts and less suitable hosts, in which the parasites merely survive, should be applicable to hermaphroditic helminths. This may open new perspectives in the control of other helminth pathogens of humans and livestock.
Collapse
Affiliation(s)
- Hanno Schmidt
- Anthropology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany.
| | - Katharina Mauer
- Anthropology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Hankeln
- Molecular Genetics and Genomic Analysis, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany
| | - Holger Herlyn
- Anthropology, Institute of Organismic and Molecular Evolution (iomE), Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
57
|
Differential Impacts on Tensional Homeostasis of Gastric Cancer Cells Due to Distinct Domain Variants of E-Cadherin. Cancers (Basel) 2022; 14:cancers14112690. [PMID: 35681670 PMCID: PMC9179447 DOI: 10.3390/cancers14112690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
In epithelia, breakdown of tensional homeostasis is closely associated with E-cadherin dysfunction and disruption of tissue function and integrity. In this study, we investigated the effect of E-cadherin mutations affecting distinct protein domains on tensional homeostasis of gastric cancer cells. We used micropattern traction microscopy to measure temporal fluctuations of cellular traction forces in AGS cells transfected with the wild-type E-cadherin or with variants affecting the extracellular, the juxtamembrane, and the intracellular domains of the protein. We focused on the dynamic aspect of tensional homeostasis, namely the ability of cells to maintain a consistent level of tension, with low temporal variability around a set point. Cells were cultured on hydrogels micropatterned with different extracellular matrix (ECM) proteins to test whether the ECM adhesion impacts cell behavior. A combination of Fibronectin and Vitronectin was used as a substrate that promotes the adhesive ability of E-cadherin dysfunctional cells, whereas Collagen VI was used to test an unfavorable ECM condition. Our results showed that mutations affecting distinct E-cadherin domains influenced differently cell tensional homeostasis, and pinpointed the juxtamembrane and intracellular regions of E-cadherin as the key players in this process. Furthermore, Fibronectin and Vitronectin might modulate cancer cell behavior towards tensional homeostasis.
Collapse
|
58
|
Gultian KA, Gandhi R, Sarin K, Sladkova-Faure M, Zimmer M, de Peppo GM, Vega SL. Human induced mesenchymal stem cells display increased sensitivity to matrix stiffness. Sci Rep 2022; 12:8483. [PMID: 35589731 PMCID: PMC9119934 DOI: 10.1038/s41598-022-12143-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical translation of mesenchymal stem cells (MSCs) is limited by population heterogeneity and inconsistent responses to engineered signals. Specifically, the extent in which MSCs respond to mechanical cues varies significantly across MSC lines. Although induced pluripotent stem cells (iPSCs) have recently emerged as a novel cell source for creating highly homogeneous MSC (iMSC) lines, cellular mechanosensing of iMSCs on engineered materials with defined mechanics is not well understood. Here, we tested the mechanosensing properties of three human iMSC lines derived from iPSCs generated using a fully automated platform. Stiffness-driven changes in morphology were comparable between MSCs and iMSCs cultured atop hydrogels of different stiffness. However, contrary to tissue derived MSCs, no significant changes in iMSC morphology were observed between iMSC lines atop different stiffness hydrogels, demonstrating a consistent response to mechanical signals. Further, stiffness-driven changes in mechanosensitive biomarkers were more pronounced in iMSCs than MSCs, which shows that iMSCs are more adaptive and responsive to mechanical cues than MSCs. This study reports that iMSCs are a promising stem cell source for basic and applied research due to their homogeneity and high sensitivity to engineered mechanical signals.
Collapse
Affiliation(s)
- Kirstene A Gultian
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, 08028, USA
| | - Roshni Gandhi
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, 08028, USA
| | - Khushi Sarin
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, 08028, USA
| | | | - Matthew Zimmer
- The New York Stem Cell Foundation Research Institute, New York, NY, 10019, USA
| | | | - Sebastián L Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, 08028, USA.
| |
Collapse
|
59
|
Kann AP, Hung M, Wang W, Nguyen J, Gilbert PM, Wu Z, Krauss RS. An injury-responsive Rac-to-Rho GTPase switch drives activation of muscle stem cells through rapid cytoskeletal remodeling. Cell Stem Cell 2022; 29:933-947.e6. [PMID: 35597234 PMCID: PMC9177759 DOI: 10.1016/j.stem.2022.04.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/14/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022]
Abstract
Many tissues harbor quiescent stem cells that are activated upon injury, subsequently proliferating and differentiating to repair tissue damage. Mechanisms by which stem cells sense injury and transition from quiescence to activation, however, remain largely unknown. Resident skeletal muscle stem cells (MuSCs) are essential orchestrators of muscle regeneration and repair. Here, with a combination of in vivo and ex vivo approaches, we show that quiescent MuSCs have elaborate, Rac GTPase-promoted cytoplasmic projections that respond to injury via the upregulation of Rho/ROCK signaling, facilitating projection retraction and driving downstream activation events. These early events involve rapid cytoskeletal rearrangements and occur independently of exogenous growth factors. This mechanism is conserved across a broad range of MuSC activation models, including injury, disease, and genetic loss of quiescence. Our results redefine MuSC activation and present a central mechanism by which quiescent stem cells initiate responses to injury.
Collapse
Affiliation(s)
- Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Margaret Hung
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wei Wang
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S3E1, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S3E1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada
| | - Zhuhao Wu
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
60
|
Khoonkari M, Liang D, Kamperman M, Kruyt FAE, van Rijn P. Physics of Brain Cancer: Multiscale Alterations of Glioblastoma Cells under Extracellular Matrix Stiffening. Pharmaceutics 2022; 14:pharmaceutics14051031. [PMID: 35631616 PMCID: PMC9145282 DOI: 10.3390/pharmaceutics14051031] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022] Open
Abstract
The biology and physics underlying glioblastoma is not yet completely understood, resulting in the limited efficacy of current clinical therapy. Recent studies have indicated the importance of mechanical stress on the development and malignancy of cancer. Various types of mechanical stress activate adaptive tumor cell responses that include alterations in the extracellular matrix (ECM) which have an impact on tumor malignancy. In this review, we describe and discuss the current knowledge of the effects of ECM alterations and mechanical stress on GBM aggressiveness. Gradual changes in the brain ECM have been connected to the biological and physical alterations of GBM cells. For example, increased expression of several ECM components such as glycosaminoglycans (GAGs), hyaluronic acid (HA), proteoglycans and fibrous proteins result in stiffening of the brain ECM, which alters inter- and intracellular signaling activity. Several mechanosensing signaling pathways have been identified that orchestrate adaptive responses, such as Hippo/YAP, CD44, and actin skeleton signaling, which remodel the cytoskeleton and affect cellular properties such as cell–cell/ECM interactions, growth, and migration/invasion of GBM cells. In vitro, hydrogels are used as a model to mimic the stiffening of the brain ECM and reconstruct its mechanics, which we also discuss. Overall, we provide an overview of the tumor microenvironmental landscape of GBM with a focus on ECM stiffening and its associated adaptive cellular signaling pathways and their possible therapeutic exploitation.
Collapse
Affiliation(s)
- Mohammad Khoonkari
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands;
| | - Dong Liang
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
| | - Marleen Kamperman
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands;
| | - Frank A. E. Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
- Correspondence: (F.A.E.K.); (P.v.R.)
| | - Patrick van Rijn
- Department of Biomedical Engineering-FB40, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Correspondence: (F.A.E.K.); (P.v.R.)
| |
Collapse
|
61
|
Mechanosignaling in vertebrate development. Dev Biol 2022; 488:54-67. [DOI: 10.1016/j.ydbio.2022.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 12/13/2022]
|
62
|
Wang JJ, Xue Q, Wang YJ, Zhang M, Chen YJ, Zhang Q. Engineered Chimeric Peptides with IGF-1 and Titanium-Binding Functions to Enhance Osteogenic Differentiation In Vitro under T2DM Condition. MATERIALS 2022; 15:ma15093134. [PMID: 35591468 PMCID: PMC9105221 DOI: 10.3390/ma15093134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/30/2022] [Accepted: 04/24/2022] [Indexed: 02/06/2023]
Abstract
Due to the complexity of the biomolecules and titanium (Ti) combination, it is a challenge to modify the implant surface with biological cytokines. The study proposed a new method for immobilizing cytokines on implant surface to solve the problem of low osseointegration under type 2 diabetes mellitus (T2DM) condition. This new modified protein that connected Ti-binding artificial aptamer minTBP-1 with Insulin-like growth factor I (IGF-I), had a special strong affinity with Ti and a therapeutic effect on diabetic bone loss. According to the copies of minTBP-1, three proteins were prepared, namely minTBP-1-IGF-1, 2minTBP-1-IGF-1 and 3minTBP-1-IGF-1. Compared with the other modified proteins, 3minTBP-1-IGF-1 adsorbed most on the Ti surface. Additionally, this biointerface demonstrated the most uniform state and the strongest hydrophilicity. In vitro results showed that the 3minTBP-1-IGF-1 significantly increased the adhesion, proliferation, and mineralization activity of osteoblasts under T2DM conditions when compared with the control group and the other modified IGF-1s groups. Real-time PCR assay results confirmed that 3minTBP-1-IGF-1 could effectively promote the expression of osteogenic genes, that is, ALP, BMP-2, OCN, OPG, and Runx2. All these data indicated that the 3minTBP-1-IGF-1 had the most efficacious effect in promoting osteoblasts osteogenesis in diabetic conditions, and may be a promising option for further clinical use.
Collapse
Affiliation(s)
| | | | | | - Min Zhang
- Correspondence: (M.Z.); (Y.-J.C.); (Q.Z.)
| | | | - Qian Zhang
- Correspondence: (M.Z.); (Y.-J.C.); (Q.Z.)
| |
Collapse
|
63
|
Pillai VV, Kei TG, Gurung S, Das M, Siqueira LGB, Cheong SH, Hansen PJ, Selvaraj V. RhoA/ROCK signaling antagonizes bovine trophoblast stem cell self-renewal and regulates preimplantation embryo size and differentiation. Development 2022; 149:274909. [DOI: 10.1242/dev.200115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/01/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Exponential proliferation of trophoblast stem cells (TSC) is crucial in Ruminantia to maximize numerical access to caruncles, the restricted uterine sites that permit implantation. When translating systems biology of the undifferentiated bovine trophectoderm, we uncovered that inhibition of RhoA/Rock promoted self-renewing proliferation and substantially increased blastocyst size. Analysis of transcripts suppressed by Rock inhibition revealed transforming growth factor β1 (TGFβ1) as a primary upstream effector. TGFβ1 treatment induced changes consistent with differentiation in bTSCs, a response that could be replicated by induced expression of the bovine ROCK2 transgene. Rocki could partially antagonize TGFβ1 effects, and TGFβ receptor inhibition promoted proliferation identical to Rocki, indicating an all-encompassing upstream regulation. Morphological differentiation included formation of binucleate cells and infrequent multinucleate syncytia, features we also localize in the in vivo bovine placenta. Collectively, we demonstrate a central role for TGFβ1, RhoA and Rock in inducing bTSC differentiation, attenuation of which is sufficient to sustain self-renewal and proliferation linked to blastocyst size and preimplantation development. Unraveling these mechanisms augments evolutionary/comparative physiology of the trophoblast cell lineage and placental development in eutherians.
Collapse
Affiliation(s)
- Viju Vijayan Pillai
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Tiffany G. Kei
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Shailesh Gurung
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Moubani Das
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Luiz G. B. Siqueira
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
- Embrapa Gado de Leite, Juiz de Fora, MG 36038-330, Brazil
| | - Soon Hon Cheong
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Peter J. Hansen
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
64
|
Khataee H, Fraser M, Neufeld Z. Modelling the Collective Mechanical Regulation of the Structure and Morphology of Epithelial Cell Layers. Front Cell Dev Biol 2022; 10:767688. [PMID: 35399530 PMCID: PMC8987200 DOI: 10.3389/fcell.2022.767688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
The morphology and function of epithelial sheets play an important role in healthy tissue development and cancer progression. The maintenance of structure of closely packed epithelial layers requires the coordination of various mechanical forces due to intracellular activities and interactions with other cells and tissues. However, a general model for the combination of mechanical properties which determine the cell shape and the overall structure of epithelial layers remains elusive. Here, we propose a computational model, based on the Cellular Potts Model, to analyse the interplay between mechanical properties of cells and dynamical transitions in epithelial cell shapes and structures. We map out phase diagrams as functions of cellular properties and the orientation of cell division. Results show that monolayers of squamous, cuboidal, and columnar cells are formed when the axis of cell proliferation is perpendicular to the substrate or along the major axis of the cells. Monolayer-to-multilayer transition is promoted via cell extrusion, depending on the mechanical properties of cells and the orientation of cell division. The results and model predictions are discussed in the context of experimental observations.
Collapse
|
65
|
Alksne M, Kalvaityte M, Simoliunas E, Gendviliene I, Barasa P, Rinkunaite I, Kaupinis A, Seinin D, Rutkunas V, Bukelskiene V. Dental pulp stem cell-derived extracellular matrix: autologous tool boosting bone regeneration. Cytotherapy 2022; 24:597-607. [PMID: 35304075 DOI: 10.1016/j.jcyt.2022.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/22/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND AIMS To facilitate artificial bone construct integration into a patient's body, scaffolds are enriched with different biologically active molecules. Among various scaffold decoration techniques, coating surfaces with cell-derived extracellular matrix (ECM) is a rapidly growing field of research. In this study, for the first time, this technology was applied using primary dental pulp stem cells (DPSCs) and tested for use in artificial bone tissue construction. METHODS Rat DPSCs were grown on three-dimensional-printed porous polylactic acid scaffolds for 7 days. After the predetermined time, samples were decellularized, and the remaining ECM detailed proteomic analysis was performed. Further, DPSC-secreated ECM impact to mesenchymal stromal cells (MSC) behaviour as well as its role in osteoregeneration induction were analysed. RESULTS It was identified that DPSC-specific ECM protein network ornamenting surface-enhanced MSC attachment, migration and proliferation and even promoted spontaneous stem cell osteogenesis. This protein network also demonstrated angiogenic properties and did not stimulate MSCs to secrete molecules associated with scaffold rejection. With regard to bone defects, DPSC-derived ECM recruited endogenous stem cells, initiating the bone self-healing process. Thus, the DPSC-secreted ECM network was able to significantly enhance artificial bone construct integration and induce successful tissue regeneration. CONCLUSIONS DPSC-derived ECM can be a perfect tool for decoration of various biomaterials in the context of bone tissue engineering.
Collapse
Affiliation(s)
- Milda Alksne
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Migle Kalvaityte
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Egidijus Simoliunas
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Gendviliene
- Institute of Odontology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Povilas Barasa
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Rinkunaite
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Algirdas Kaupinis
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dmitrij Seinin
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Vygandas Rutkunas
- Institute of Odontology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Virginija Bukelskiene
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
66
|
Huang SQ, Chen Y, Zhu Q, Zhang YM, Lei ZY, Zhou X, Fan DL. In Vivo and In Vitro Fibroblasts' Behavior and Capsular Formation in Correlation with Smooth and Textured Silicone Surfaces. Aesthetic Plast Surg 2022; 46:1164-1177. [PMID: 35237878 DOI: 10.1007/s00266-022-02769-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND As the most principal complication following breast augmentation with silicone breast implants, capsular contracture is greatly influenced by surface texture. However, there have long been widespread debates on the function of smooth or textured surface implants in reducing capsular contracture. MATERIALS AND METHODS Three commercially available silicone breast implants with smooth and textured surfaces were subjected to surface characterization, and in vitro and in vivo assessments were then implemented to investigate the effect of these different surfaces on the biological behaviors of fibroblasts and capsular formation in rat models. RESULTS Surface characterization demonstrated that all three samples were hydrophobic with distinct roughness values. Comparing the interactions of fibroblasts or tissues with different surfaces, we observed that as surface roughness increased, the adhesion and cell spreading of fibroblasts, the level of echogenicity, the density of collagen and α-SMA-positive immunoreactivity decreased, while the proliferation of fibroblasts and capsule thickness increased. CONCLUSIONS Our findings elucidated that the effect of silicone implant surface texture on fibroblasts' behaviors and capsular formation was associated with variations in surface roughness, and the number of myofibroblasts may have a more significant influence on the process of contracture than capsule thickness in the early stage of capsular formation. These results highlight that targeting myofibroblasts may be wielded in the prevention and treatment strategies of capsular contracture clinically. LEVEL OF EVIDENCE V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Shu-Qing Huang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing, 400037, People's Republic of China
| | - Yao Chen
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing, 400037, People's Republic of China
| | - Qiong Zhu
- Department of Ultrasound, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi-Ming Zhang
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing, 400037, People's Republic of China
| | - Ze-Yuan Lei
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing, 400037, People's Republic of China
| | - Xin Zhou
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing, 400037, People's Republic of China.
| | - Dong-Li Fan
- Department of Plastic and Cosmetic Surgery, Xinqiao Hospital, Army Medical University, Xinqiao Road, Sha Ping Ba District, Chongqing, 400037, People's Republic of China.
| |
Collapse
|
67
|
Zhang M, Meng N, Wang X, Chen W, Zhang Q. TRPV4 and PIEZO Channels Mediate the Mechanosensing of Chondrocytes to the Biomechanical Microenvironment. MEMBRANES 2022; 12:membranes12020237. [PMID: 35207158 PMCID: PMC8874592 DOI: 10.3390/membranes12020237] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023]
Abstract
Articular cartilage and their chondrocytes are physiologically submitted to diverse types of mechanical cues. Chondrocytes produce and maintain the cartilage by sensing and responding to changing mechanical loads. TRPV4 and PIEZOs, activated by mechanical cues, are important mechanosensing molecules of chondrocytes and have pivotal roles in articular cartilage during health and disease. The objective of this review is to introduce the recent progress indicating that the mechanosensitive ion channels, TRPV4 and PIEZOs, are involved in the chondrocyte sensing of mechanical and inflammatory cues. We present a focus on the important role of TRPV4 and PIEZOs in the mechanotransduction regulating diverse chondrocyte functions in the biomechanical microenvironment. The review synthesizes the most recent advances in our understanding of how mechanical stimuli affect various cellular behaviors and functions through differentially activating TRPV4 and PIEZO ion channels in chondrocyte. Advances in understanding the complex roles of TRPV4/PIEZO-mediated mechanosignaling mechanisms have the potential to recapitulate physiological biomechanical microenvironments and design cell-instructive biomaterials for cartilage tissue engineering.
Collapse
Affiliation(s)
- Min Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (M.Z.); (N.M.); (X.W.)
| | - Nan Meng
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (M.Z.); (N.M.); (X.W.)
| | - Xiaoxiao Wang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (M.Z.); (N.M.); (X.W.)
| | - Weiyi Chen
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (M.Z.); (N.M.); (X.W.)
- Correspondence: (W.C.); (Q.Z.); Tel.: +86-15364710252 (W.C.); +86-13700500252 (Q.Z.); Fax: +86-0351-3176651 (Q.Z.)
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (M.Z.); (N.M.); (X.W.)
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan 030001, China
- Correspondence: (W.C.); (Q.Z.); Tel.: +86-15364710252 (W.C.); +86-13700500252 (Q.Z.); Fax: +86-0351-3176651 (Q.Z.)
| |
Collapse
|
68
|
Aguado BA, Walker CJ, Grim JC, Schroeder ME, Batan D, Vogt BJ, Rodriguez AG, Schwisow JA, Moulton KS, Weiss RM, Heistad DD, Leinwand LA, Anseth KS. Genes That Escape X Chromosome Inactivation Modulate Sex Differences in Valve Myofibroblasts. Circulation 2022; 145:513-530. [PMID: 35000411 PMCID: PMC8844107 DOI: 10.1161/circulationaha.121.054108] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aortic valve stenosis is a sexually dimorphic disease, with women often presenting with sustained fibrosis and men with more extensive calcification. However, the intracellular molecular mechanisms that drive these clinically important sex differences remain underexplored. METHODS Hydrogel biomaterials were designed to recapitulate key aspects of the valve tissue microenvironment and to serve as a culture platform for sex-specific valvular interstitial cells (VICs; precursors to profibrotic myofibroblasts). The hydrogel culture system was used to interrogate intracellular pathways involved in sex-dependent VIC-to-myofibroblast activation and deactivation. RNA sequencing was used to define pathways involved in driving sex-dependent activation. Interventions with small molecule inhibitors and siRNA transfections were performed to provide mechanistic insight into sex-specific cellular responses to microenvironmental cues, including matrix stiffness and exogenously delivered biochemical factors. RESULTS In both healthy porcine and human aortic valves, female leaflets had higher baseline activation of the myofibroblast marker α-smooth muscle actin compared with male leaflets. When isolated and cultured, female porcine and human VICs had higher levels of basal α-smooth muscle actin stress fibers that further increased in response to the hydrogel matrix stiffness, both of which were higher than in male VICs. A transcriptomic analysis of male and female porcine VICs revealed Rho-associated protein kinase signaling as a potential driver of this sex-dependent myofibroblast activation. Furthermore, we found that genes that escape X-chromosome inactivation such as BMX and STS (encoding for Bmx nonreceptor tyrosine kinase and steroid sulfatase, respectively) partially regulate the elevated female myofibroblast activation through Rho-associated protein kinase signaling. This finding was confirmed by treating male and female VICs with endothelin-1 and plasminogen activator inhibitor-1, factors that are secreted by endothelial cells and known to drive myofibroblast activation through Rho-associated protein kinase signaling. CONCLUSIONS Together, in vivo and in vitro results confirm sex dependencies in myofibroblast activation pathways and implicate genes that escape X-chromosome inactivation in regulating sex differences in myofibroblast activation and subsequent aortic valve stenosis progression. Our results underscore the importance of considering sex as a biological variable to understand the molecular mechanisms of aortic valve stenosis and to help guide sex-based precision therapies.
Collapse
Affiliation(s)
- Brian A. Aguado
- Department of Chemical and Biological Engineering, University of Colorado Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Cierra J. Walker
- Materials Science and Engineering Program, University of Colorado Boulder, CO 80309, USA
- Department of Biochemistry, University of Colorado Boulder, CO 80303, USA
| | - Joseph C. Grim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
| | - Megan E. Schroeder
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
- Materials Science and Engineering Program, University of Colorado Boulder, CO 80309, USA
| | - Dilara Batan
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
- Department of Biochemistry, University of Colorado Boulder, CO 80303, USA
| | - Brandon J. Vogt
- Department of Chemical and Biological Engineering, University of Colorado Boulder, CO 80303, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Andrea Gonzalez Rodriguez
- Department of Chemical and Biological Engineering, University of Colorado Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
| | - Jessica A. Schwisow
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Karen S. Moulton
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert M. Weiss
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242
| | - Donald D. Heistad
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242
| | - Leslie A. Leinwand
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, CO 80309, USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, CO 80303, USA
- BioFrontiers Institute, University of Colorado Boulder, CO 80309, USA
- Materials Science and Engineering Program, University of Colorado Boulder, CO 80309, USA
| |
Collapse
|
69
|
Panzetta V, Musella I, Fusco S, Netti PA. ECM Mechanoregulation in Malignant Pleural Mesothelioma. Front Bioeng Biotechnol 2022; 10:797900. [PMID: 35237573 PMCID: PMC8883334 DOI: 10.3389/fbioe.2022.797900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/05/2022] [Indexed: 01/16/2023] Open
Abstract
Malignant pleural mesothelioma is a relatively rare, but devastating tumor, because of the difficulties in providing early diagnosis and effective treatments with conventional chemo- and radiotherapies. Patients usually present pleural effusions that can be used for diagnostic purposes by cytological analysis. This effusion cytology may take weeks or months to establish and has a limited sensitivity (30%-60%). Then, it is becoming increasingly urgent to develop alternative investigative methods to support the diagnosis of mesothelioma at an early stage when this cancer can be treated successfully. To this purpose, mechanobiology provides novel perspectives into the study of tumor onset and progression and new diagnostic tools for the mechanical characterization of tumor tissues. Here, we report a mechanical and biophysical characterization of malignant pleural mesothelioma cells as additional support to the diagnosis of pleural effusions. In particular, we examined a normal mesothelial cell line (Met5A) and two epithelioid mesothelioma cell lines (REN and MPP89), investigating how malignant transformation can influence cellular function like proliferation, cell migration, and cell spreading area with respect to the normal ones. These alterations also correlated with variations in cytoskeletal mechanical properties that, in turn, were measured on substrates mimicking the stiffness of patho-physiological ECM.
Collapse
Affiliation(s)
- Valeria Panzetta
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, IIT@CRIB, Naples, Italy
| | - Ida Musella
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, IIT@CRIB, Naples, Italy
| | - Sabato Fusco
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Paolo A. Netti
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, IIT@CRIB, Naples, Italy
| |
Collapse
|
70
|
Sivaraj D, Padmanabhan J, Chen K, Henn D, Noishiki C, Trotsyuk AA, Kussie HC, Leeolou MC, Magbual NJ, Andrikopoulos S, Perrault DP, Barrera JA, Januszyk M, Gurtner GC. IQGAP1-mediated mechanical signaling promotes the foreign body response to biomedical implants. FASEB J 2022; 36:e22007. [PMID: 35051300 DOI: 10.1096/fj.202101354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022]
Abstract
The aim of this study was to further elucidate the molecular mechanisms that mediate pathologic foreign body response (FBR) to biomedical implants. The longevity of biomedical implants is limited by the FBR, which leads to implant failure and patient morbidity. Since the specific molecular mechanisms underlying fibrotic responses to biomedical implants have yet to be fully described, there are currently no targeted approaches to reduce pathologic FBR. We utilized proteomics analysis of human FBR samples to identify potential molecular targets for therapeutic inhibition of FBR. We then employed a murine model of FBR to further evaluate the role of this potential target. We performed histological and immunohistochemical analysis on the murine FBR capsule tissue, as well as single-cell RNA sequencing (scRNA-seq) on cells isolated from the capsules. We identified IQ motif containing GTPase activating protein 1 (IQGAP1) as the most promising of several targets, serving as a central molecular mediator in human and murine FBR compared to control subcutaneous tissue. IQGAP1-deficient mice displayed a significantly reduced FBR compared to wild-type mice as evidenced by lower levels of collagen deposition and maturity. Our scRNA-seq analysis revealed that decreasing IQGAP1 resulted in diminished transcription of mechanotransduction, inflammation, and fibrosis-related genes, which was confirmed on the protein level with immunofluorescent staining. The deficiency of IQGAP1 significantly attenuates FBR by deactivating downstream mechanotransduction signaling, inflammation, and fibrotic pathways. IQGAP1 may be a promising target for rational therapeutic design to mitigate pathologic FBR around biomedical implants.
Collapse
Affiliation(s)
- Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Chikage Noishiki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Artem A Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hudson C Kussie
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Melissa C Leeolou
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Noah J Magbual
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Sophia Andrikopoulos
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - David P Perrault
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Janos A Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Geoffrey C Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
71
|
Babayeva DB, Shishkin MM, Fayzrakhmanov RR, Konovalova KI. [Vitreopapillary traction syndrome in patients with proliferative diabetic retinopathy]. Vestn Oftalmol 2021; 137:38-44. [PMID: 34965066 DOI: 10.17116/oftalma202113706138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE To draw attention to characteristic features of the clinical manifestations of vitreopapillary traction syndrome (VPTS) in patients with proliferative diabetic retinopathy (PDR), to determine the role of the biomechanical factor in pathophysiology of this syndrome, and to evaluate the results of surgical treatment. MATERIAL AND METHODS The features of clinical manifestations of VPTS of 50 patients with PDR who were treated at the ophthalmology center of the National Medical and Surgical Center named after N.I. Pirogov in the period from 2014 to 2018 were analyzed retrospectively. The preoperative study included standard ophthalmological examination methods, as well as ultrasound B-scan, computer perimetry, optical coherence tomography (OCT), fluorescence angiography (FA) in the first two years of observation. The comparison group consisted of 45 patients with PDR without signs of VPTS, who were treated in the center during the same period of time. RESULTS The obtained results suggest that the pathophysiology of VPTS in diabetes mellitus has distinctive features and is determined by the topography of the peripapillary zone, as well as choroidal vascularization of the optic nerve head; biomechanical factor plays an important role in the formation of a fibrovascular stem (FVS) in VPTS. CONCLUSION The results indicate that patients with VPTS have a worse prognosis for achieving best corrected visual acuity (BCVA) after surgery than patients without signs of VPTS. Correct and timely diagnosis, early treatment by vitreoretinal surgery give a greater chance of achieving higher BCVA in the postoperative period in this category of patients.
Collapse
Affiliation(s)
- D B Babayeva
- National Medical and Surgical Center named after N.I. Pirogov, Moscow, Russia
| | - M M Shishkin
- National Medical and Surgical Center named after N.I. Pirogov, Moscow, Russia
| | - R R Fayzrakhmanov
- National Medical and Surgical Center named after N.I. Pirogov, Moscow, Russia
| | - K I Konovalova
- National Medical and Surgical Center named after N.I. Pirogov, Moscow, Russia
| |
Collapse
|
72
|
Babi M, Riesco R, Boyer L, Fatona A, Accardo A, Malaquin L, Moran-Mirabal J. Tuning the Nanotopography and Chemical Functionality of 3D Printed Scaffolds through Cellulose Nanocrystal Coatings. ACS APPLIED BIO MATERIALS 2021; 4:8443-8455. [PMID: 35005920 DOI: 10.1021/acsabm.1c00970] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In nature, cells exist in three-dimensional (3D) microenvironments with topography, stiffness, surface chemistry, and biological factors that strongly dictate their phenotype and behavior. The cellular microenvironment is an organized structure or scaffold that, together with the cells that live within it, make up living tissue. To mimic these systems and understand how the different properties of a scaffold, such as adhesion, proliferation, or function, influence cell behavior, we need to be able to fabricate cellular microenvironments with tunable properties. In this work, the nanotopography and functionality of scaffolds for cell culture were modified by coating 3D printed materials (DS3000 and poly(ethylene glycol)diacrylate, PEG-DA) with cellulose nanocrystals (CNCs). This general approach was demonstrated on a variety of structures designed to incorporate macro- and microscale features fabricated using photopolymerization and 3D printing techniques. Atomic force microscopy was used to characterize the CNC coatings and showed the ability to tune their density and in turn the surface nanoroughness from isolated nanoparticles to dense surface coverage. The ability to tune the density of CNCs on 3D printed structures could be leveraged to control the attachment and morphology of prostate cancer cells. It was also possible to introduce functionalization onto the surface of these scaffolds, either by directly coating them with CNCs grafted with the functionality of interest or with a more general approach of functionalizing the CNCs after coating using biotin-streptavidin coupling. The ability to carefully tune the nanostructure and functionalization of different 3D-printable materials is a step forward to creating in vitro scaffolds that mimic the nanoscale features of natural microenvironments, which are key to understanding their impact on cells and developing artificial tissues.
Collapse
Affiliation(s)
- Mouhanad Babi
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Roberto Riesco
- LAAS-CNRS, Université Toulouse III─Paul Sabatier, 31400 Toulouse, France
| | - Louisa Boyer
- LAAS-CNRS, Université Toulouse III─Paul Sabatier, 31400 Toulouse, France
| | - Ayodele Fatona
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Angelo Accardo
- Department of Precision and Microsystems Engineering, Delft University of Technology, Mekelweg 2, 2628 CD Delft, Netherlands
| | - Laurent Malaquin
- LAAS-CNRS, Université Toulouse III─Paul Sabatier, 31400 Toulouse, France
| | - Jose Moran-Mirabal
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada.,Centre for Advanced Light Microscopy, McMaster University, Hamilton, Ontario L8S 4M1, Canada.,Brockhouse Institute for Materials Research, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| |
Collapse
|
73
|
Harberts J, Bours K, Siegmund M, Hedrich C, Glatza M, Schöler HR, Haferkamp U, Pless O, Zierold R, Blick RH. Culturing human iPSC-derived neural progenitor cells on nanowire arrays: mapping the impact of nanowire length and array pitch on proliferation, viability, and membrane deformation. NANOSCALE 2021; 13:20052-20066. [PMID: 34842880 DOI: 10.1039/d1nr04352h] [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
Nanowire arrays used as cell culture substrates build a potent tool for advanced biological applications such as cargo delivery and biosensing. The unique topography of nanowire arrays, however, renders them a challenging growth environment for cells and explains why only basic cell lines have been employed in existing studies. Here, we present the culturing of human induced pluripotent stem cell-derived neural progenitor cells on rectangularly arranged nanowire arrays: In detail, we mapped the impact on proliferation, viability, and topography-induced membrane deformation across a multitude of array pitches (1, 3, 5, 10 μm) and nanowire lengths (1.5, 3, 5 μm). Against the intuitive expectation, a reduced proliferation was found on the arrays with the smallest array pitch of 1 μm and long NWs. Typically, cells settle in a fakir-like state on such densely-spaced nanowires and thus experience no substantial stress caused by nanowires indenting the cell membrane. However, imaging of F-actin showed a distinct reorganization of the cytoskeleton along the nanowire tips in the case of small array pitches interfering with regular proliferation. For larger pitches, the cell numbers depend on the NW lengths but proliferation generally continued although heavy deformations of the cell membrane were observed caused by the encapsulation of the nanowires. Moreover, we noticed a strong interaction of the nanowires with the nucleus in terms of squeezing and indenting. Remarkably, the cell viability is maintained at about 85% despite the massive deformation of the cells. Considering the enormous potential of human induced stem cells to study neurodegenerative diseases and the high cellular viability combined with a strong interaction with nanowire arrays, we believe that our results pave the way to apply nanowire arrays to human stem cells for future applications in stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Katja Bours
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Malte Siegmund
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Carina Hedrich
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Michael Glatza
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Robert Zierold
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Robert H Blick
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
- Material Science and Engineering, College of Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
74
|
Muntz I, Fenu M, van Osch GJVM, Koenderink G. The role of cell-matrix interactions in connective tissue mechanics. Phys Biol 2021; 19. [PMID: 34902848 DOI: 10.1088/1478-3975/ac42b8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/13/2021] [Indexed: 11/12/2022]
Abstract
Living tissue is able to withstand large stresses in everyday life, yet it also actively adapts to dynamic loads. This remarkable mechanical behaviour emerges from the interplay between living cells and their non-living extracellular environment. Here we review recent insights into the biophysical mechanisms involved in the reciprocal interplay between cells and the extracellular matrix and how this interplay determines tissue mechanics, with a focus on connective tissues. We first describe the roles of the main macromolecular components of the extracellular matrix in regards to tissue mechanics. We then proceed to highlight the main routes via which cells sense and respond to their biochemical and mechanical extracellular environment. Next we introduce the three main routes via which cells can modify their extracellular environment: exertion of contractile forces, secretion and deposition of matrix components, and matrix degradation. Finally we discuss how recent insights in the mechanobiology of cell-matrix interactions are furthering our understanding of the pathophysiology of connective tissue diseases and cancer, and facilitating the design of novel strategies for tissue engineering.
Collapse
Affiliation(s)
- Iain Muntz
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HC, NETHERLANDS
| | - Michele Fenu
- Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gerjo J V M van Osch
- Orthopaedics; Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Zuid-Holland, 3000 CA, NETHERLANDS
| | - Gijsje Koenderink
- Bionanoscience, TU Delft, Kavli Institute of Nanoscience Delft, Delft University of Technology, Van der Maasweg 9, Delft, Zuid-Holland, 2629 HZ, NETHERLANDS
| |
Collapse
|
75
|
Valencia FR, Sandoval E, Du J, Iu E, Liu J, Plotnikov SV. Force-dependent activation of actin elongation factor mDia1 protects the cytoskeleton from mechanical damage and promotes stress fiber repair. Dev Cell 2021; 56:3288-3302.e5. [PMID: 34822787 DOI: 10.1016/j.devcel.2021.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/02/2021] [Accepted: 11/02/2021] [Indexed: 01/16/2023]
Abstract
Plasticity of cell mechanics underlies a wide range of cell and tissue behaviors allowing cells to migrate through narrow spaces, resist shear forces, and safeguard against mechanical damage. Such plasticity depends on spatiotemporal regulation of the actomyosin cytoskeleton, but mechanisms of adaptive change in cell mechanics remain elusive. Here, we report a mechanism of mechanically activated actin polymerization at focal adhesions (FAs), specifically requiring the actin elongation factor mDia1. By combining live-cell imaging with mathematical modeling, we show that actin polymerization at FAs exhibits pulsatile dynamics where spikes of mDia1 activity are triggered by contractile forces. The suppression of mDia1-mediated actin polymerization increases tension on stress fibers (SFs) leading to an increased frequency of spontaneous SF damage and decreased efficiency of zyxin-mediated SF repair. We conclude that tension-controlled actin polymerization acts as a safety valve dampening excessive tension on the actin cytoskeleton and safeguarding SFs against mechanical damage.
Collapse
Affiliation(s)
- Fernando R Valencia
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Eduardo Sandoval
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joy Du
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Ernest Iu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jian Liu
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
76
|
Petzold J, Gentleman E. Intrinsic Mechanical Cues and Their Impact on Stem Cells and Embryogenesis. Front Cell Dev Biol 2021; 9:761871. [PMID: 34820380 PMCID: PMC8606660 DOI: 10.3389/fcell.2021.761871] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Collapse
Affiliation(s)
- Jonna Petzold
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
77
|
Hou Y, Xie W, Fan X, Tang P, Yu L, Haag R. "Raspberry" Hierarchical Topographic Features Regulate Human Mesenchymal Stem Cell Adhesion and Differentiation via Enhanced Mechanosensing. ACS APPLIED MATERIALS & INTERFACES 2021; 13:54840-54849. [PMID: 34756008 DOI: 10.1021/acsami.1c18722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An understanding of cellular mechanoresponses to well-defined synthetic topographic features is crucial for the fundamental research and biomedical applications of stem cells. Structured biointerfaces, in particular the ones with nanometer and/or micrometer surficial features, have drawn more attention in the past few decades. However, it is still difficult to integrate nanostructures and microstructures onto the synthesized biointerfaces to mimic the hierarchical architecture of the native extracellular matrix (ECM). Herein, a series of "raspberry"-like hierarchical surfaces with well-defined nanofeatures and tunable nano/microfeatures have been achieved via a catecholic polymer coating technique. Cellular responses to these hierarchical interfaces were systemically studied, indicating that the nanofeatures on the raspberry surfaces significantly enhanced the mechanosensing of human mesenchymal stem cells (hMSCs) to interfacial physical cues. Cell mechanotransduction was further investigated by analyzing focal adhesion assembling, cytoskeleton organization, cell nuclear mechanics, and transcriptional activity. The results suggest that nanosize surficial features could increase cellular mechanosensing to environment physical cues. The mechanotransduction and cell fate specification were greatly enhanced by the ECM mimicking nano/microhierarchical biointerfaces but the features should be in an optimized size.
Collapse
Affiliation(s)
- Yong Hou
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Wenyan Xie
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany
| | - Xin Fan
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Peng Tang
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Leixiao Yu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
78
|
Kim S, Choi C, Cha C. Mechanotopography-Driven Design of Dispersible Nanofiber-Laden Hydrogel as a 3D Cell Culture Platform for Investigating Tissue Fibrosis. Adv Healthc Mater 2021; 10:e2101109. [PMID: 34494395 DOI: 10.1002/adhm.202101109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/26/2021] [Indexed: 11/07/2022]
Abstract
Fibrosis is one of the most frequent occurrences during one's lifetime, identified by various physiological changes including, most notably, excessive deposition of extracellular matrix (ECM). Despite its physiological importance, it is still a significant challenge to conduct a systematic investigation of tissue fibrosis, mainly due to the lack of in vitro 3D tissue model that can accurately portray the characteristic features of fibrotic events. Herein, a hybrid hydrogel system incorporating dispersible nanofibers is developed to emulate highly collagenous deposits formed within a fibrotic tissue leading to altered mechanotopographical properties. Micrometer-length, aqueous-stable nanofibers consisting of crosslinked gelatin network embedded with graphene oxide (GO) or reduced graphene (rGO) are infused into hydrogel, resulting in controllable mechanotopographical properties while maintaining permeability sufficiently enough for various cellular activities. Ultimately, the ability to induce fibrotic behavior of fibroblasts cultured in these mechanotopography-controlled, nanofiber-laden hydrogels is investigated in detail.
Collapse
Affiliation(s)
- Suntae Kim
- Center for Multidimensional Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Cholong Choi
- Center for Multidimensional Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Chaenyung Cha
- Center for Multidimensional Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| |
Collapse
|
79
|
Huang WY, Suye SI, Fujita S. Cell Trapping via Migratory Inhibition within Density-Tuned Electrospun Nanofibers. ACS APPLIED BIO MATERIALS 2021; 4:7456-7466. [PMID: 35006712 DOI: 10.1021/acsabm.1c00700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell migration is an essential bioprocess that occurs during wound healing and tissue regeneration. Abnormal cell migration is observed in various pathologies, including cancer metastasis. Glioblastoma multiforme (GBM) is an aggressive and highly infiltrative brain tumor. The white matter tracts are considered the preferred routes for GBM invasion and the subsequent spread throughout the brain tissue. In the present study, a platform based on electrospun nanofibers with a consistent alignment and controlled density was designed to inhibit cell migration. The observation of the cells cultured on the nanofibers with different fiber densities revealed an inverse correlation between the cell migration velocity and nanofiber density. This was attributed to the formation of focal adhesions (FAs). The FAs in the sparse fiber matrix were small, whereas those in the dense fiber matrix were large, aligned with the nanofibers, and distributed throughout the cells. A nanofiber-based platform with stepwise different fiber densities was designed based on the aforementioned observation. A time-lapse observation of the GBM cells cultured on the platform revealed a directional one-way migration that induced the entrapment of cells in the dense-fiber zone. The designed platform mimicked the structure of the white matter tracts and enabled the entrapment of migrating cells. The demonstrated approach is suitable for inhibiting metastasis and understanding the biology of invasion, thereby functioning as a promising therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Wan-Ying Huang
- Department of Advanced Interdisciplinary Science and Technology, Graduate School of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Shin-Ichiro Suye
- Department of Advanced Interdisciplinary Science and Technology, Graduate School of Engineering, University of Fukui, Fukui 910-8507, Japan.,Department of Frontier Fiber Technology and Science, University of Fukui, Fukui 910-8507, Japan.,Organization for Life Science Advancement Programs, University of Fukui, Fukui 910-8507, Japan
| | - Satoshi Fujita
- Department of Advanced Interdisciplinary Science and Technology, Graduate School of Engineering, University of Fukui, Fukui 910-8507, Japan.,Department of Frontier Fiber Technology and Science, University of Fukui, Fukui 910-8507, Japan.,Organization for Life Science Advancement Programs, University of Fukui, Fukui 910-8507, Japan
| |
Collapse
|
80
|
Luo Y, Li J, Li B, Xia Y, Wang H, Fu C. Physical Cues of Matrices Reeducate Nerve Cells. Front Cell Dev Biol 2021; 9:731170. [PMID: 34646825 PMCID: PMC8502847 DOI: 10.3389/fcell.2021.731170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/20/2021] [Indexed: 11/15/2022] Open
Abstract
The behavior of nerve cells plays a crucial role in nerve regeneration. The mechanical, topographical, and electrical microenvironment surrounding nerve cells can activate cellular signaling pathways of mechanical transduction to affect the behavior of nerve cells. Recently, biological scaffolds with various physical properties have been developed as extracellular matrix to regulate the behavior conversion of nerve cell, such as neuronal neurite growth and directional differentiation of neural stem cells, providing a robust driving force for nerve regeneration. This review mainly focused on the biological basis of nerve cells in mechanical transduction. In addition, we also highlighted the effect of the physical cues, including stiffness, mechanical tension, two-dimensional terrain, and electrical conductivity, on neurite outgrowth and differentiation of neural stem cells and predicted their potential application in clinical nerve tissue engineering.
Collapse
Affiliation(s)
- Yiqian Luo
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jie Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Baoqin Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
81
|
Qi C, Min P, Wang Q, Wang Y, Song Y, Zhang Y, Bibi M, Du J. MICAL2 Contributes to Gastric Cancer Cell Proliferation by Promoting YAP Dephosphorylation and Nuclear Translocation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9955717. [PMID: 34650666 PMCID: PMC8510804 DOI: 10.1155/2021/9955717] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/12/2021] [Accepted: 09/16/2021] [Indexed: 01/19/2023]
Abstract
Dynamic cytoskeletal rearrangements underlie the changes that occur during cell division in proliferating cells. MICAL2 has been reported to possess reactive oxygen species- (ROS-) generating properties and act as an important regulator of cytoskeletal dynamics. However, whether it plays a role in gastric cancer cell proliferation is not known. In the present study, we found that MICAL2 was highly expressed in gastric cancer tissues, and this high expression level was associated with carcinogenesis and poor overall survival in gastric cancer patients. The knockdown of MICAL2 led to cell cycle arrest in the S phase and attenuated cell proliferation. Concomitant with S-phase arrest, a decrease in CDK6 and cyclin D protein levels was observed. Furthermore, MICAL2 knockdown attenuated intracellular ROS generation, while MICAL2 overexpression led to a decrease in the p-YAP/YAP ratio and promoted YAP nuclear localization and cell proliferation, effects that were reversed by pretreatment with the ROS scavenger N-acetyl-L-cysteine (NAC) and SOD-mimetic drug tempol. We further found that MICAL2 induced Cdc42 activation, and activated Cdc42 mediated the effect of MICAL2 on YAP dephosphorylation and nuclear translocation. Collectively, our results showed that MICAL2 has a promotive effect on gastric cancer cell proliferation through ROS generation and Cdc42 activation, both of which independently contribute to YAP dephosphorylation and its nuclear translocation.
Collapse
Affiliation(s)
- Chenxiang Qi
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Pengxiang Min
- Key Laboratory of Cardio Vascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qianwen Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yueyuan Wang
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yixuan Song
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maria Bibi
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
82
|
Nie Y, Wang W, Xu X, Ma N, Lendlein A. The response of human induced pluripotent stem cells to cyclic temperature changes explored by BIO-AFM. MRS ADVANCES 2021; 6:745-749. [DOI: 10.1557/s43580-021-00110-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 01/06/2025]
Abstract
AbstractHuman induced pluripotent stem cells (hiPSCs) are highly sensitive to extrinsic physical and biochemical signals from their extracellular microenvironments. In this study, we analyzed the effect of cyclic temperature changes on hiPSCs behaviors, especially by means of scanning force microscopy (BIO-AFM). The alternation in cellular mechanics, as well as the secretion and pattern of deposition of extracellular matrix (ECM) protein in hiPSCs were evaluated. The arrangement of the actin cytoskeleton changed with the variation of the temperature. The rearranged cytoskeleton architecture led to the subsequent changes in cell mechanics (Young's modulus of hiPSCs). With the exposure to the cyclic cold stimuli, an increase in the average surface roughness (Ra) and roughness mean square (RMS) was detected. This observation might be at least in part due to the upregulated secretion of Laminin α5 during repeated temporary cooling. The expression of pluripotent markers, NANOG and SOX2, was not impaired in hiPSCs, when exposed to the cyclic cold stimuli for 24 h. Our findings provide an insight into the effect of temperature on the hiPSC behaviors, which may contribute to a better understanding of the application of locally controlled therapeutic hypothermia.Graphic abstractThe cyclic temperature changes, from 37 to 10 °C, rapidly increased the mechanical strength of human-induced pluripotent stem cells (hiPSCs), which could be explained by the re-arrangement of cytoskeletons. The capacity of hiPSCs to remodel the extracellular matrix was also altered by the repeated temporary cooling, as they exhibit an enhanced ability to physically remodulate and secrete the ECM components.
Collapse
|
83
|
Yuzhalin AE. Parallels between the extracellular matrix roles in developmental biology and cancer biology. Semin Cell Dev Biol 2021; 128:90-102. [PMID: 34556419 DOI: 10.1016/j.semcdb.2021.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 12/28/2022]
Abstract
Interaction of a tumor with its microenvironment is an emerging field of investigation, and the crosstalk between cancer cells and the extracellular matrix is of particular interest, since cancer patients with abundant and stiff extracellular matrices display a poorer prognosis. At the post-juvenile stage, the extracellular matrix plays predominantly a structural role by providing support to cells and tissues; however, during development, matrix proteins exert a plethora of diverse signals to guide the movement and determine the fate of pluripotent cells. Taking a closer look at the communication between the extracellular matrix and cells of a developing body may bring new insights into cancer biology and identify cancer weaknesses. This review discusses parallels between the extracellular matrix roles during development and tumor growth.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
84
|
Kim MH, Thanuthanakhun N, Fujimoto S, Kino-Oka M. Effect of initial seeding density on cell behavior-driven epigenetic memory and preferential lineage differentiation of human iPSCs. Stem Cell Res 2021; 56:102534. [PMID: 34530397 DOI: 10.1016/j.scr.2021.102534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/22/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022] Open
Abstract
Understanding the cellular behavioral mechanisms underlying memory formation and maintenance in human induced pluripotent stem cell (hiPSC) culture provides key strategies for achieving stability and robustness of cell differentiation. Here, we show that changes in cell behavior-driven epigenetic memory of hiPSC cultures alter their pluripotent state and subsequent differentiation. Interestingly, pluripotency-associated genes were activated during the entire cell growth phases along with increased active modifications and decreased repressive modifications. This memory effect can last several days in the long-term stationary phase and was sustained in the aspect of cell behavioral changes after subculture. Further, changes in growth-related cell behavior were found to induce nucleoskeletal reorganization and active versus repressive modifications, thereby enabling hiPSCs to change their differentiation potential. Overall, we discuss the cell behavior-driven epigenetic memory induced by the culture environment, and the effect of previous memory on cell lineage specification in the process of hiPSC differentiation.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shun Fujimoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
85
|
Reye G, Huang X, Haupt LM, Murphy RJ, Northey JJ, Thompson EW, Momot KI, Hugo HJ. Mechanical Pressure Driving Proteoglycan Expression in Mammographic Density: a Self-perpetuating Cycle? J Mammary Gland Biol Neoplasia 2021; 26:277-296. [PMID: 34449016 PMCID: PMC8566410 DOI: 10.1007/s10911-021-09494-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 07/05/2021] [Indexed: 12/23/2022] Open
Abstract
Regions of high mammographic density (MD) in the breast are characterised by a proteoglycan (PG)-rich fibrous stroma, where PGs mediate aligned collagen fibrils to control tissue stiffness and hence the response to mechanical forces. Literature is accumulating to support the notion that mechanical stiffness may drive PG synthesis in the breast contributing to MD. We review emerging patterns in MD and other biological settings, of a positive feedback cycle of force promoting PG synthesis, such as in articular cartilage, due to increased pressure on weight bearing joints. Furthermore, we present evidence to suggest a pro-tumorigenic effect of increased mechanical force on epithelial cells in contexts where PG-mediated, aligned collagen fibrous tissue abounds, with implications for breast cancer development attributable to high MD. Finally, we summarise means through which this positive feedback mechanism of PG synthesis may be intercepted to reduce mechanical force within tissues and thus reduce disease burden.
Collapse
Affiliation(s)
- Gina Reye
- School of Biomedical Sciences, Gardens Point, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia
- Translational Research Institute, Woolloongabba, QLD, Australia
| | - Xuan Huang
- School of Biomedical Sciences, Gardens Point, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia
- Translational Research Institute, Woolloongabba, QLD, Australia
| | - Larisa M Haupt
- Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Ryan J Murphy
- School of Mathematical Sciences, Gardens Point, Queensland University of Technology (QUT), Kelvin Grove, QLD, Australia
| | - Jason J Northey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Erik W Thompson
- School of Biomedical Sciences, Gardens Point, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia
- Translational Research Institute, Woolloongabba, QLD, Australia
| | - Konstantin I Momot
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Honor J Hugo
- School of Biomedical Sciences, Gardens Point, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia.
- Translational Research Institute, Woolloongabba, QLD, Australia.
| |
Collapse
|
86
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
87
|
Kałuzińska Ż, Kołat D, Bednarek AK, Płuciennik E. PLEK2, RRM2, GCSH: A Novel WWOX-Dependent Biomarker Triad of Glioblastoma at the Crossroads of Cytoskeleton Reorganization and Metabolism Alterations. Cancers (Basel) 2021; 13:2955. [PMID: 34204789 PMCID: PMC8231639 DOI: 10.3390/cancers13122955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is one of the deadliest human cancers. Its malignancy depends on cytoskeleton reorganization, which is related to, e.g., epithelial-to-mesenchymal transition and metastasis. The malignant phenotype of glioblastoma is also affected by the WWOX gene, which is lost in nearly a quarter of gliomas. Although the role of WWOX in the cytoskeleton rearrangement has been found in neural progenitor cells, its function as a modulator of cytoskeleton in gliomas was not investigated. Therefore, this study aimed to investigate the role of WWOX and its collaborators in cytoskeleton dynamics of glioblastoma. Methodology on RNA-seq data integrated the use of databases, bioinformatics tools, web-based platforms, and machine learning algorithm, and the obtained results were validated through microarray data. PLEK2, RRM2, and GCSH were the most relevant WWOX-dependent genes that could serve as novel biomarkers. Other genes important in the context of cytoskeleton (BMP4, CCL11, CUX2, DUSP7, FAM92B, GRIN2B, HOXA1, HOXA10, KIF20A, NF2, SPOCK1, TTR, UHRF1, and WT1), metabolism (MTHFD2), or correlation with WWOX (COL3A1, KIF20A, RNF141, and RXRG) were also discovered. For the first time, we propose that changes in WWOX expression dictate a myriad of alterations that affect both glioblastoma cytoskeleton and metabolism, rendering new therapeutic possibilities.
Collapse
Affiliation(s)
- Żaneta Kałuzińska
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752 Lodz, Poland; (D.K.); (A.K.B.); (E.P.)
| | | | | | | |
Collapse
|
88
|
The Hippo Pathway: A Master Regulatory Network Important in Cancer. Cells 2021; 10:cells10061416. [PMID: 34200285 PMCID: PMC8226666 DOI: 10.3390/cells10061416] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 12/11/2022] Open
Abstract
The Hippo pathway is pervasively activated and has been well recognized to play critical roles in human cancer. The deregulation of Hippo signaling involved in cancer development, progression, and resistance to cancer treatment have been confirmed in several human cancers. Its biological significance and deregulation in cancer have drawn increasing interest in the past few years. A fundamental understanding of the complexity of the Hippo pathway in cancer is crucial for improving future clinical interventions and therapy for cancers. In this review, we try to clarify the complex regulation and function of the Hippo signaling network in cancer development, including its role in signal transduction, metabolic regulation, and tumor development, as well as tumor therapies targeting the Hippo pathway.
Collapse
|
89
|
Ngo MT, Harley BAC. Progress in mimicking brain microenvironments to understand and treat neurological disorders. APL Bioeng 2021; 5:020902. [PMID: 33869984 PMCID: PMC8034983 DOI: 10.1063/5.0043338] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders including traumatic brain injury, stroke, primary and metastatic brain tumors, and neurodegenerative diseases affect millions of people worldwide. Disease progression is accompanied by changes in the brain microenvironment, but how these shifts in biochemical, biophysical, and cellular properties contribute to repair outcomes or continued degeneration is largely unknown. Tissue engineering approaches can be used to develop in vitro models to understand how the brain microenvironment contributes to pathophysiological processes linked to neurological disorders and may also offer constructs that promote healing and regeneration in vivo. In this Perspective, we summarize features of the brain microenvironment in normal and pathophysiological states and highlight strategies to mimic this environment to model disease, investigate neural stem cell biology, and promote regenerative healing. We discuss current limitations and resulting opportunities to develop tissue engineering tools that more faithfully recapitulate the aspects of the brain microenvironment for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Mai T. Ngo
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Brendan A. C. Harley
- Author to whom correspondence should be addressed:. Tel.: (217) 244-7112. Fax: (217) 333-5052
| |
Collapse
|
90
|
Mahmoud M, Mayer M, Cancel LM, Bartosch AM, Mathews R, Tarbell JM. The glycocalyx core protein Glypican 1 protects vessel wall endothelial cells from stiffness-mediated dysfunction and disease. Cardiovasc Res 2021; 117:1592-1605. [PMID: 32647868 PMCID: PMC8152694 DOI: 10.1093/cvr/cvaa201] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/22/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Arterial stiffness is an underlying risk factor and a hallmark of cardiovascular diseases. The endothelial cell (EC) glycocalyx is a glycan rich surface layer that plays a key role in protecting against EC dysfunction and vascular disease. However, the mechanisms by which arterial stiffness promotes EC dysfunction and vascular disease are not fully understood, and whether the mechanism involves the protective endothelial glycocalyx is yet to be determined. We hypothesized that endothelial glycocalyx protects the endothelial cells lining the vascular wall from dysfunction and disease in response to arterial stiffness. METHODS AND RESULTS Cells cultured on polyacrylamide (PA) gels of substrate stiffness 10 kPa (mimicking the subendothelial stiffness of aged, unhealthy arteries) showed a significant inhibition of glycocalyx expression compared to cells cultured on softer PA gels (2.5 kPa, mimicking the subendothelial stiffness of young, healthy arteries). Specifically, gene and protein analyses revealed that a glycocalyx core protein Glypican 1 was inhibited in cells cultured on stiff PA gels. These cells had enhanced endothelial cell dysfunction as determined by enhanced cell inflammation (enhanced inflammatory gene expression, monocyte adhesion, and inhibited nitric oxide expression), proliferation, and EndMT. Removal of Glypican 1 using gene-specific silencing with siRNA or gene overexpression using a plasmid revealed that Glypican 1 is required to protect against stiffness-mediated endothelial cell dysfunction. Consistent with this, using a model of age-mediated stiffness, older mice exhibited a reduced expression of Glypican 1 and enhanced endothelial cell dysfunction compared to young mice. Glypican 1 gene deletion in knockout mice (GPC1-/-) exacerbated endothelial dysfunction in young mice, which normally had high endothelial expression, but not in old mice that normally expressed low levels. Endothelial cell dysfunction was exacerbated in young, but not aged, Glypican 1 knockout mice (GPC1-/-). CONCLUSION Arterial stiffness promotes EC dysfunction and vascular disease at least partly through the suppression of the glycocalyx protein Glypican 1. Glypican 1 contributes to the protection against endothelial cell dysfunction and vascular disease in endothelial cells.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Mariya Mayer
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Limary M Cancel
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Anne Marie Bartosch
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Rick Mathews
- Oregon Health & Science University, School of Medicine, Portland, OR, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| |
Collapse
|
91
|
Kwon TY, Jeong J, Park E, Cho Y, Lim D, Ko UH, Shin JH, Choi J. Physical analysis reveals distinct responses of human bronchial epithelial cells to guanidine and isothiazolinone biocides. Toxicol Appl Pharmacol 2021; 424:115589. [PMID: 34029620 DOI: 10.1016/j.taap.2021.115589] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/18/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
Changes in the physical state of the cells can serve as important indicators of stress responses because they are closely linked with the changes in the pathophysiological functions of the cells. Physical traits can be conveniently assessed by analyzing the morphological features and the stresses at the cell-matrix and cell-cell adhesions in both single-cell and monolayer model systems in 2D. In this study, we investigated the mechano-stress responses of human bronchial epithelial cells, BEAS-2B, to two functionally distinct groups of biocides identified during the humidifier disinfectant accident, namely, guanidine (PHMG) and isothiazolinone (CMIT/MIT). We analyzed the physical traits, including cell area, nuclear area, and nuclear shape. While the results showed inconsistent average responses to the biocides, the degree of dispersion in the data set, measured by standard deviation, was remarkably higher in CMIT/MIT treated cells for all traits. As mechano-stress endpoints, traction and intercellular stresses were also measured, and the cytoskeletal actin structures were analyzed using immunofluorescence. This study demonstrates the versatility of the real-time imaging-based biomechanical analysis, which will contribute to identifying the temporally sensitive cellular behaviors as well as the emergence of heterogeneity in response to exogenously imposed stress factors. This study will also shed light on a comparative understanding of less studied substance, CMIT/MIT, in relation to a more studied substance, PHMG, which will further contribute to more strategic planning for proper risk management of the ingredients involved in toxicological accidents.
Collapse
Affiliation(s)
- Tae Yoon Kwon
- Department of Mechanical Engineering, KAIST, 291 Daehakro, Yuseong-gu, Daejeon 34034, Republic of Korea
| | - Jaeseong Jeong
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Eunyoung Park
- Department of Mechanical Engineering, KAIST, 291 Daehakro, Yuseong-gu, Daejeon 34034, Republic of Korea
| | - Youngbin Cho
- Department of Mechanical Engineering, KAIST, 291 Daehakro, Yuseong-gu, Daejeon 34034, Republic of Korea
| | - Dongyoung Lim
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic of Korea
| | - Ung Hyun Ko
- Department of Mechanical Engineering, KAIST, 291 Daehakro, Yuseong-gu, Daejeon 34034, Republic of Korea
| | - Jennifer H Shin
- Department of Mechanical Engineering, KAIST, 291 Daehakro, Yuseong-gu, Daejeon 34034, Republic of Korea.
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, Republic of Korea.
| |
Collapse
|
92
|
Lee JS, Park TH, Ryu JY, Kim DK, Oh EJ, Kim HM, Shim JH, Yun WS, Huh JB, Moon SH, Kang SS, Chung HY. Osteogenesis of 3D-Printed PCL/TCP/bdECM Scaffold Using Adipose-Derived Stem Cells Aggregates; An Experimental Study in the Canine Mandible. Int J Mol Sci 2021; 22:ijms22115409. [PMID: 34063742 PMCID: PMC8196585 DOI: 10.3390/ijms22115409] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional (3D) printing is perceived as an innovative tool for change in tissue engineering and regenerative medicine based on research outcomes on the development of artificial organs and tissues. With advances in such technology, research is underway into 3D-printed artificial scaffolds for tissue recovery and regeneration. In this study, we fabricated artificial scaffolds by coating bone demineralized and decellularized extracellular matrix (bdECM) onto existing 3D-printed polycaprolactone/tricalcium phosphate (PCL/TCP) to enhance osteoconductivity and osteoinductivity. After injecting adipose-derived stem cells (ADSCs) in an aggregate form found to be effective in previous studies, we examined the effects of the scaffold on ossification during mandibular reconstruction in beagle dogs. Ten beagles were divided into two groups: group A (PCL/TCP/bdECM + ADSC injection; n = 5) and group B (PCL/TCP/bdECM; n = 5). The results were analyzed four and eight weeks after intervention. Computed tomography (CT) findings showed that group A had more diffuse osteoblast tissue than group B. Evidence of infection or immune rejection was not detected following histological examination. Goldner trichrome (G/T) staining revealed rich ossification in scaffold pores. ColI, Osteocalcin, and Runx2 gene expressions were determined using real-time polymerase chain reaction. Group A showed greater expression of these genes. Through Western blotting, group A showed a greater expression of genes that encode ColI, Osteocalcin, and Runx2 proteins. In conclusion, intervention group A, in which the beagles received the additional ADSC injection together with the 3D-printed PCL/TCP coated with bdECM, showed improved mandibular ossification in and around the pores of the scaffold.
Collapse
Affiliation(s)
- Joon Seok Lee
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (T.H.P.); (J.Y.R.); (E.J.O.); (H.M.K.)
| | - Tae Hyun Park
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (T.H.P.); (J.Y.R.); (E.J.O.); (H.M.K.)
| | - Jeong Yeop Ryu
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (T.H.P.); (J.Y.R.); (E.J.O.); (H.M.K.)
| | - Dong Kyu Kim
- TINA Aesthetic Surgical Clinic, Daegu 41938, Korea;
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (T.H.P.); (J.Y.R.); (E.J.O.); (H.M.K.)
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hyun Mi Kim
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (T.H.P.); (J.Y.R.); (E.J.O.); (H.M.K.)
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jin-Hyung Shim
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-Ro, Siheung-si 15073, Gyeonggi-do, Korea; (J.-H.S.); (W.-S.Y.)
- Research Institute, T&R Biofab Co., Ltd. 242 Pangyo-ro, Seongnam-si 13487, Gyeonggi-do, Korea;
| | - Won-Soo Yun
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-Ro, Siheung-si 15073, Gyeonggi-do, Korea; (J.-H.S.); (W.-S.Y.)
- Research Institute, T&R Biofab Co., Ltd. 242 Pangyo-ro, Seongnam-si 13487, Gyeonggi-do, Korea;
| | - Jung Bo Huh
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Science, School of Dentistry, Pusan National University, Yangsan-si 50612, Korea;
| | - Sung Hwan Moon
- Research Institute, T&R Biofab Co., Ltd. 242 Pangyo-ro, Seongnam-si 13487, Gyeonggi-do, Korea;
| | - Seong Soo Kang
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Korea;
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (J.S.L.); (T.H.P.); (J.Y.R.); (E.J.O.); (H.M.K.)
- Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Science for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: or ; Tel.: +82-53-420-5692; Fax: +82-53-425-3879
| |
Collapse
|
93
|
Martirosyan A, Poghosyan D, Ghonyan S, Mkrtchyan N, Amaryan G, Manukyan G. Transmigration of Neutrophils From Patients With Familial Mediterranean Fever Causes Increased Cell Activation. Front Immunol 2021; 12:672728. [PMID: 34079554 PMCID: PMC8165278 DOI: 10.3389/fimmu.2021.672728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Familial Mediterranean fever (FMF) is caused by pyrin-encoding MEFV gene mutations and characterized by the self-limiting periods of intense inflammation, which are mainly mediated by a massive influx of polymorphonuclear neutrophils (PMNs) into the inflamed sites. Perturbation of actin polymerization by different pathogens was shown to activate the pyrin inflammasome. Our aim was to test whether cytoskeletal dynamics in the absence of pathogens may cause abnormal activation of PMNs from FMF patients. We also aimed to characterize immunophenotypes of circulating neutrophils and their functional activity. Circulating PMNs displayed heterogeneity in terms of cell size, granularity and immunophenotypes. Particularly, PMNs from the patients in acute flares (FMF-A) exhibited a characteristic of aged/activated cells (small cell size and granularity, up-regulated CXCR4), while PMNs form the patients in remission period (FMF-R) displayed mixed fresh/aged cell characteristics (normal cell size and granularity, up-regulated CD11b, CD49d, CXCR4, and CD62L). The findings may suggest that sterile tissue-infiltrated PMNs undergo reverse migration back to bone marrow and may explain why these PMNs do not cause immune-mediated tissue damage. A multidirectional expression of FcγRs on neutrophils during acute flares was also noteworthy: up-regulation of FcγRI and down-regulation of FcγRII/FcγRIII. We also observed spontaneous and fMPL-induced activation of PMNs from the patients after transmigration through inserts as seen by the increased expression of CD11b and intracellular expression of IL-1β. Our study suggests heightened sensitivity of mutated pyrin inflammasome towards cytoskeletal modifications in the absence of pathogens.
Collapse
Affiliation(s)
- Anush Martirosyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology National Academy of Sciences of the Republic of Armenia (NAS RA), Yerevan, Armenia
| | - David Poghosyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology National Academy of Sciences of the Republic of Armenia (NAS RA), Yerevan, Armenia
| | - Susanna Ghonyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology National Academy of Sciences of the Republic of Armenia (NAS RA), Yerevan, Armenia
| | - Nune Mkrtchyan
- National Pediatrics Center of Familial Mediterranean Fever "Arabkir" Joint Medical Center- Institute of Child and Adolescent Health, Yerevan, Armenia.,Department of Pediatrics, Yerevan State Medical University, Yerevan, Armenia
| | - Gayane Amaryan
- National Pediatrics Center of Familial Mediterranean Fever "Arabkir" Joint Medical Center- Institute of Child and Adolescent Health, Yerevan, Armenia.,Department of Pediatrics, Yerevan State Medical University, Yerevan, Armenia
| | - Gayane Manukyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology National Academy of Sciences of the Republic of Armenia (NAS RA), Yerevan, Armenia
| |
Collapse
|
94
|
A spatial model of YAP/TAZ signaling reveals how stiffness, dimensionality, and shape contribute to emergent outcomes. Proc Natl Acad Sci U S A 2021; 118:2021571118. [PMID: 33990464 DOI: 10.1073/pnas.2021571118] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
YAP/TAZ is a master regulator of mechanotransduction whose functions rely on translocation from the cytoplasm to the nucleus in response to diverse physical cues. Substrate stiffness, substrate dimensionality, and cell shape are all input signals for YAP/TAZ, and through this pathway, regulate critical cellular functions and tissue homeostasis. Yet, the relative contributions of each biophysical signal and the mechanisms by which they synergistically regulate YAP/TAZ in realistic tissue microenvironments that provide multiplexed input signals remain unclear. For example, in simple two-dimensional culture, YAP/TAZ nuclear localization correlates strongly with substrate stiffness, while in three-dimensional (3D) environments, YAP/TAZ translocation can increase with stiffness, decrease with stiffness, or remain unchanged. Here, we develop a spatial model of YAP/TAZ translocation to enable quantitative analysis of the relationships between substrate stiffness, substrate dimensionality, and cell shape. Our model couples cytosolic stiffness to nuclear mechanics to replicate existing experimental trends, and extends beyond current data to predict that increasing substrate activation area through changes in culture dimensionality, while conserving cell volume, forces distinct shape changes that result in nonlinear effect on YAP/TAZ nuclear localization. Moreover, differences in substrate activation area versus total membrane area can account for counterintuitive trends in YAP/TAZ nuclear localization in 3D culture. Based on this multiscale investigation of the different system features of YAP/TAZ nuclear translocation, we predict that how a cell reads its environment is a complex information transfer function of multiple mechanical and biochemical factors. These predictions reveal a few design principles of cellular and tissue engineering for YAP/TAZ mechanotransduction.
Collapse
|
95
|
Mascharak S, desJardins-Park HE, Davitt MF, Griffin M, Borrelli MR, Moore AL, Chen K, Duoto B, Chinta M, Foster DS, Shen AH, Januszyk M, Kwon SH, Wernig G, Wan DC, Lorenz HP, Gurtner GC, Longaker MT. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science 2021; 372:372/6540/eaba2374. [PMID: 33888614 DOI: 10.1126/science.aba2374] [Citation(s) in RCA: 333] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 10/01/2020] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
Skin scarring, the end result of adult wound healing, is detrimental to tissue form and function. Engrailed-1 lineage-positive fibroblasts (EPFs) are known to function in scarring, but Engrailed-1 lineage-negative fibroblasts (ENFs) remain poorly characterized. Using cell transplantation and transgenic mouse models, we identified a dermal ENF subpopulation that gives rise to postnatally derived EPFs by activating Engrailed-1 expression during adult wound healing. By studying ENF responses to substrate mechanics, we found that mechanical tension drives Engrailed-1 activation via canonical mechanotransduction signaling. Finally, we showed that blocking mechanotransduction signaling with either verteporfin, an inhibitor of Yes-associated protein (YAP), or fibroblast-specific transgenic YAP knockout prevents Engrailed-1 activation and promotes wound regeneration by ENFs, with recovery of skin appendages, ultrastructure, and mechanical strength. This finding suggests that there are two possible outcomes to postnatal wound healing: a fibrotic response (EPF-mediated) and a regenerative response (ENF-mediated).
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heather E desJardins-Park
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael F Davitt
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mimi R Borrelli
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alessandra L Moore
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kellen Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bryan Duoto
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Malini Chinta
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deshka S Foster
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Abra H Shen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sun Hyung Kwon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gerlinde Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Peter Lorenz
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
96
|
Yang Y, Lin Y, Zhang Z, Xu R, Yu X, Deng F. Micro/nano-net guides M2-pattern macrophage cytoskeleton distribution via Src-ROCK signalling for enhanced angiogenesis. Biomater Sci 2021; 9:3334-3347. [PMID: 33725044 DOI: 10.1039/d1bm00116g] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Implant surface topography has been proven to determine the fate of adhered macrophage polarization, which is closely related to the cytoskeletal arrangement during adhesion. Our purpose was to establish a topography that is favourable to M2 macrophage switching by regulating macrophage cytoskeleton distribution. Two micro/nano-net structures with different pore sizes were generated by alkali bathing at medium (SAM) or high (SAH) temperature based on the micro-level surface. Their surface characteristics, in vitro macrophage polarization and impact on endothelial cells were analysed. The in vivo macrophage response and osseointegration were also tested. The results showed that the micro/nano-net has high hydrophilicity and moderate roughness. In the SAH and SAM groups, macrophages exhibited an elongated cytoskeleton with tiny protrusions and had a high M2/M1 polarization ratio with enhanced angiogenic ability, and in vivo studies also showed faster angiogenesis and bone formation in these groups. SAH showed even better results than SAM. For cytoskeleton related pathway explanation, ROCK expression was upregulated and Src expression was downregulated at the early or late adhesion stage in both the SAH and SAM groups. These results indicated that the micro/nano-net structure guides elongated macrophage adhesion states via Src-ROCK signalling and switches macrophages towards the M2 phenotype, which provides a cytoskeleton-oriented topography design for an ideal immune response.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Yujing Lin
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Zhengchuan Zhang
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Ruogu Xu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Xiaoran Yu
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| | - Feilong Deng
- Department of Oral Implantology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, PR China. and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| |
Collapse
|
97
|
Xu Z, Orkwis JA, Harris GM. Cell Shape and Matrix Stiffness Impact Schwann Cell Plasticity via YAP/TAZ and Rho GTPases. Int J Mol Sci 2021; 22:ijms22094821. [PMID: 34062912 PMCID: PMC8124465 DOI: 10.3390/ijms22094821] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Schwann cells (SCs) are a highly plastic cell type capable of undergoing phenotypic changes following injury or disease. SCs are able to upregulate genes associated with nerve regeneration and ultimately achieve functional recovery. During the regeneration process, the extracellular matrix (ECM) and cell morphology play a cooperative, critical role in regulating SCs, and therefore highly impact nerve regeneration outcomes. However, the roles of the ECM and mechanotransduction relating to SC phenotype are largely unknown. Here, we describe the role that matrix stiffness and cell morphology play in SC phenotype specification via known mechanotransducers YAP/TAZ and RhoA. Using engineered microenvironments to precisely control ECM stiffness, cell shape, and cell spreading, we show that ECM stiffness and SC spreading downregulated SC regenerative associated proteins by the activation of RhoA and YAP/TAZ. Additionally, cell elongation promoted a distinct SC regenerative capacity by the upregulation of Rac1/MKK7/JNK, both necessary for the ECM and morphology changes found during nerve regeneration. These results confirm the role of ECM signaling in peripheral nerve regeneration as well as provide insight to the design of future biomaterials and cellular therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Zhenyuan Xu
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
| | - Jacob A. Orkwis
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
| | - Greg M. Harris
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-(513)-556-4167
| |
Collapse
|
98
|
Abstract
The development of molecular nanostructures with well-defined particle size and shape is of eminent interest in biomedicine. Among many studied nanostructures, dendrimers represent the group of those most thoroughly characterized ones. Due to their unique structure and properties, dendrimers are very attractive for medical and pharmaceutical applications. Owing to the controllable cavities inside the dendrimer, guest molecules may be encapsulated, and highly reactive terminal groups are susceptible to further modifications, e.g., to facilitate target delivery. To understand the potential of these nanoparticles and to predict and avoid any adverse cellular reactions, it is necessary to know the mechanisms responsible for an efficient dendrimer uptake and the destination of their intracellular journey. In this article, we summarize the results of studies describing the dendrimer uptake, traffic, and efflux mechanisms depending on features of specific nanoparticles and cell types. We also present mechanisms of dendrimers responsible for toxicity and alteration in signal transduction pathways at the cellular level.
Collapse
Affiliation(s)
- Barbara Ziemba
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
| | - Maciej Borowiec
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
| | - Ida Franiak-Pietryga
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland.,Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
99
|
Chen X, Lu F, Yuan Y. The Application and Mechanism of Action of External Volume Expansion in Soft Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 27:181-197. [PMID: 32821009 DOI: 10.1089/ten.teb.2020.0137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Xihang Chen
- Department of Plastic and Cosmetic Surgery, Southern Medical University, Nanfang Hospital, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Southern Medical University, Nanfang Hospital, Guangzhou, China
| | - Yi Yuan
- Department of Plastic and Cosmetic Surgery, Southern Medical University, Nanfang Hospital, Guangzhou, China
| |
Collapse
|
100
|
Leek CC, Soulas JM, Sullivan AL, Killian ML. Using tools in mechanobiology to repair tendons. ACTA ACUST UNITED AC 2021; 1:31-40. [PMID: 33585822 DOI: 10.1007/s43152-020-00005-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Purpose of review The purpose of this review is to describe the mechanobiological mechanisms of tendon repair as well as outline current and emerging tools in mechanobiology that might be useful for improving tendon healing and regeneration. Over 30 million musculoskeletal injuries are reported in the US per year and nearly 50% involve soft tissue injuries to tendons and ligaments. Yet current therapeutic strategies for treating tendon injuries are not always successful in regenerating and returning function of the healing tendon. Recent findings The use of rehabilitative strategies to control the motion and transmission of mechanical loads to repairing tendons following surgical reattachment is beneficial for some, but not all, tendon repairs. Scaffolds that are designed to recapitulate properties of developing tissues show potential to guide the mechanical and biological healing of tendon following rupture. The incorporation of biomaterials to control alignment and reintegration, as well as promote scar-less healing, are also promising. Improving our understanding of damage thresholds for resident cells and how these cells respond to bioelectrical cues may offer promising steps forward in the field of tendon regeneration. Summary The field of orthopaedics continues to advance and improve with the development of regenerative approaches for musculoskeletal injuries, especially for tendon, and deeper exploration in this area will lead to improved clinical outcomes.
Collapse
Affiliation(s)
- Connor C Leek
- College of Engineering, Department of Biomedical Engineering, 5 Innovation Way, Suite 200, University of Delaware, Newark, Delaware 19716
| | - Jaclyn M Soulas
- College of Engineering, Department of Biomedical Engineering, 5 Innovation Way, Suite 200, University of Delaware, Newark, Delaware 19716.,College of Agriculture and Natural Resources, Department of Animal Biosciences, 531 South College Avenue, University of Delaware, Newark, Delaware 19716
| | - Anna Lia Sullivan
- College of Engineering, Department of Biomedical Engineering, 5 Innovation Way, Suite 200, University of Delaware, Newark, Delaware 19716.,College of Agriculture and Natural Resources, Department of Animal Biosciences, 531 South College Avenue, University of Delaware, Newark, Delaware 19716
| | - Megan L Killian
- College of Engineering, Department of Biomedical Engineering, 5 Innovation Way, Suite 200, University of Delaware, Newark, Delaware 19716.,College of Medicine, Department of Orthopaedic Surgery, 109 Zina Pitcher Place, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|