51
|
Le S, Yu M, Yan J. Direct single-molecule quantification reveals unexpectedly high mechanical stability of vinculin-talin/α-catenin linkages. SCIENCE ADVANCES 2019; 5:eaav2720. [PMID: 31897422 PMCID: PMC6920023 DOI: 10.1126/sciadv.aav2720] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2019] [Indexed: 05/26/2023]
Abstract
The vinculin-mediated mechanosensing requires establishment of stable mechanical linkages between vinculin to integrin at focal adhesions and to cadherins at adherens junctions through associations with the respective adaptor proteins talin and α-catenin. However, the mechanical stability of these critical vinculin linkages has yet to be determined. Here, we developed a single-molecule detector assay to provide direct quantification of the mechanical lifetime of vinculin association with the vinculin binding sites in both talin and α-catenin, which reveals a surprisingly high mechanical stability of the vinculin-talin and vinculin-α-catenin interfaces that have a lifetime of >1000 s at forces up to 10 pN and can last for seconds to tens of seconds at 15 to 25 pN. Our results suggest that these force-bearing intermolecular interfaces provide sufficient mechanical stability to support the vinculin-mediated mechanotransduction at cell-matrix and cell-cell adhesions.
Collapse
Affiliation(s)
- Shimin Le
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| | - Miao Yu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| | - Jie Yan
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
- Centre for Bioimaging Sciences, National University of Singapore, Singapore 117546, Singapore
| |
Collapse
|
52
|
Morales W, Rezaie A, Barlow G, Pimentel M. Second-Generation Biomarker Testing for Irritable Bowel Syndrome Using Plasma Anti-CdtB and Anti-Vinculin Levels. Dig Dis Sci 2019; 64:3115-3121. [PMID: 31152332 DOI: 10.1007/s10620-019-05684-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND ELISA testing for anti-CdtB and anti-vinculin can discriminate patients with irritable bowel syndrome with diarrhea (IBS-D) from those with inflammatory bowel disease (IBD). However, recent findings suggest the antigens can suffer from epitope instability. AIM This study aimed to assess effects of incorporating epitope stabilization on test characteristics for distinguishing IBS-D from IBD subjects. METHODS Plasma samples from IBS-D subjects from a large-scale clinical trial and subjects with endoscopically active IBD without concurrent immunomodulator therapy were used. After epitope stabilization, CdtB and vinculin were used in ELISA testing. Optical density readings were compared between IBS-D and IBD subjects. RESULTS Samples from 100 IBS-D and 31 IBD (22 UC and 9 CD) subjects were tested. IBS-D subjects had higher anti-CdtB titers (P = 0.0001) and higher anti-vinculin titers (P = 0.004) than IBD subjects. The specificities of anti-CdtB and anti-vinculin to differentiate IBS-D from IBD were 93.5% and 90.9%, respectively, with sensitivities of 43.0% and 52.2%, respectively. The positive likelihood ratios of identifying IBS-D with anti-CdtB and anti-vinculin were 6.7 and 5.7, respectively. Assuming a pretest probability of 57% for diagnosis of IBS-D in patients with abdominal pain and change in bowel habits, testing positive for both antibodies resulted in a posttest probability of > 98%. CONCLUSIONS Performing epitope stabilization for CdtB and vinculin enhances the test characteristics of ELISAs for anti-CdtB and anti-vinculin in discriminating IBS-D from IBD. Measurement of anti-CdtB and anti-vinculin with this second-generation methodology may further advance our understanding of the role of immunity in functional bowel diseases.
Collapse
Affiliation(s)
- Walter Morales
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA
| | - Ali Rezaie
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA
| | - Gillian Barlow
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA
| | - Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, 8730 Alden Drive, Suite 240E, Los Angeles, CA, 90048, USA.
| |
Collapse
|
53
|
The planarian Vinculin is required for the regeneration of GABAergic neurons in Dugesia japonica. Exp Cell Res 2019; 383:111540. [PMID: 31369753 DOI: 10.1016/j.yexcr.2019.111540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 10/26/2022]
Abstract
Vinculin is a cytoskeletal protein associated with cell-cell and cell-matrix junctions, playing an important role in linkage of integrin adhesion molecules to the actin cytoskeleton. The planarian nervous system is a fascinating system for studying the organogenesis during regeneration. In this paper, a homolog gene of Vinculin, DjVinculin, was identified and characterized in Dugesia japonica. The DjVinculin sequence analysis revealed that it contains an opening reading frame encoding a putative protein of 975 amino acids with functionally domains that are highly conserved, including eight anti-parallel α-helical bundles organized into five distinct domains. Whole mount in situ hybridization showed that DjVinculin was predominantly expressed in the brain of intact and regenerating planarians. RNA interference of DjVinculin caused distinct defects in brain morphogenesis and influences the regeneration of planarian GABAergic neurons. The expression level of DjGAD protein was decreased in the DjVinculin-knockdown planarians. These findings suggest that DjVinculin is required for GABAergic neurons regeneration.
Collapse
|
54
|
Krokhotin A, Sarker M, Sevilla EA, Costantini LM, Griffith JD, Campbell SL, Dokholyan NV. Distinct Binding Modes of Vinculin Isoforms Underlie Their Functional Differences. Structure 2019; 27:1527-1536.e3. [PMID: 31422909 PMCID: PMC6774862 DOI: 10.1016/j.str.2019.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 05/23/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Vinculin and its splice isoform metavinculin play key roles in regulating cellular morphology, motility, and force transduction. Vinculin is distinct from metavinculin in its ability to bundle filamentous actin (F-actin). To elucidate the molecular basis for these differences, we employed computational and experimental approaches. Results from these analyses suggest that the C terminus of both vinculin and metavinculin form stable interactions with the F-actin surface. However, the metavinculin tail (MVt) domain contains a 68 amino acid insert, with helix 1 (H1) sequestered into a globular subdomain, which protrudes from the F-actin surface and prevents actin bundling by sterically occluding actin filaments. Consistent with our model, deletion and selective point mutations within the MVt H1 disrupt this protruding structure, and facilitate actin bundling similar to vinculin tail (Vt) domain.
Collapse
Affiliation(s)
- Andrey Krokhotin
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Departments of Pathology, Genetics and Developmental Biology, Howard Hughes Medical Institute, Stanford Medical School, Palo Alto, CA 94305, USA
| | - Muzaddid Sarker
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ernesto Alva Sevilla
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lindsey M Costantini
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jack D Griffith
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Departments of Pharmacology and Departments of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
55
|
Transcriptome sequencing reveals genetic mechanisms of reproduction performance stimulated by dietary daidzein in laying breeder hens. Theriogenology 2019; 142:120-130. [PMID: 31593879 DOI: 10.1016/j.theriogenology.2019.09.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/21/2019] [Accepted: 09/26/2019] [Indexed: 12/23/2022]
Abstract
Daidzein (DA) is a kind of isoflavone that is extracted primarily from soy plants and that has become increasingly popular as a dietary supplement. The objective of this study was to evaluate the effects of dietary DA supplementation for laying breeder hens on laying performance, reproductive organ development, hatching performance of seed eggs, and growth performance of offspring and to investigate the underlying molecular mechanisms. A total of 180 55-week-old laying breeder hens were randomly divided into 2 treatment groups and, after 3 weeks of acclimation, were fed either a control diet (CON) or a DA-supplemented diet (DAS, CON+30 mg/kg DA) for a total of 12 weeks. DAS treatment improved the laying rate, luteinizing hormone (LH) levels, and small yellow follicle (SYF) numbers without negative effects on the hatchability of breeder eggs or the growth performance of offspring. High-throughput RNA sequencing was utilized to identify differentially expressed genes in the SYF granulosa layer in the two groups. Transcriptome analysis showed that 161 genes (fold change ≥2 or ≤0.5; P-value<0.05) were significantly differentially expressed between the two groups, including 139 upregulated genes and 22 downregulated genes. Gene ontology (GO) functional annotation analysis revealed potential genes, processes and pathways involved in cell proliferation and differentiation related to the improvement of laying performance stimulated by DA. Dietary DA supplementation for laying breeder hens improved laying performance and reproductive performance with no negative impacts on hatchability or offspring growth. A series of differentially expressed genes in SYF granulosa cells were significantly upregulated in the DAS group relative to the CON group. This study provides insight into the genetic architecture of the transcriptome of the SYF granulosa layer in layer breeding hens and proposes candidate genes that respond to dietary DA.
Collapse
|
56
|
Mechanical Forces Regulate Cardiomyocyte Myofilament Maturation via the VCL-SSH1-CFL Axis. Dev Cell 2019; 51:62-77.e5. [PMID: 31495694 DOI: 10.1016/j.devcel.2019.08.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2019] [Accepted: 08/07/2019] [Indexed: 01/07/2023]
Abstract
Mechanical forces regulate cell behavior and tissue morphogenesis. During cardiac development, mechanical stimuli from the heartbeat are required for cardiomyocyte maturation, but the underlying molecular mechanisms remain unclear. Here, we first show that the forces of the contracting heart regulate the localization and activation of the cytoskeletal protein vinculin (VCL), which we find to be essential for myofilament maturation. To further analyze the role of VCL in this process, we examined its interactome in contracting versus non-contracting cardiomyocytes and, in addition to several known interactors, including actin regulators, identified the slingshot protein phosphatase SSH1. We show how VCL recruits SSH1 and its effector, the actin depolymerizing factor cofilin (CFL), to regulate F-actin rearrangement and promote cardiomyocyte myofilament maturation. Overall, our results reveal that mechanical forces generated by cardiac contractility regulate cardiomyocyte maturation through the VCL-SSH1-CFL axis, providing further insight into how mechanical forces are transmitted intracellularly to regulate myofilament maturation.
Collapse
|
57
|
Lee HT, Sharek L, O’Brien ET, Urbina FL, Gupton SL, Superfine R, Burridge K, Campbell SL. Vinculin and metavinculin exhibit distinct effects on focal adhesion properties, cell migration, and mechanotransduction. PLoS One 2019; 14:e0221962. [PMID: 31483833 PMCID: PMC6726196 DOI: 10.1371/journal.pone.0221962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/19/2019] [Indexed: 12/04/2022] Open
Abstract
Vinculin (Vcn) is a ubiquitously expressed cytoskeletal protein that links transmembrane receptors to actin filaments, and plays a key role in regulating cell adhesion, motility, and force transmission. Metavinculin (MVcn) is a Vcn splice isoform that contains an additional exon encoding a 68-residue insert within the actin binding tail domain. MVcn is selectively expressed at sub-stoichiometic amounts relative to Vcn in smooth and cardiac muscle cells. Mutations in the MVcn insert are linked to various cardiomyopathies. In vitro analysis has previously shown that while both proteins can engage filamentous (F)-actin, only Vcn can promote F-actin bundling. Moreover, we and others have shown that MVcn can negatively regulate Vcn-mediated F-actin bundling in vitro. To investigate functional differences between MVcn and Vcn, we stably expressed either Vcn or MVcn in Vcn-null mouse embryonic fibroblasts. While both MVcn and Vcn were observed at FAs, MVcn-expressing cells had larger but fewer focal adhesions per cell compared to Vcn-expressing cells. MVcn-expressing cells migrated faster and exhibited greater persistence compared to Vcn-expressing cells, even though Vcn-containing FAs assembled and disassembled faster. Magnetic tweezer measurements on Vcn-expressing cells show a typical cell stiffening phenotype in response to externally applied force; however, this was absent in Vcn-null and MVcn-expressing cells. Our findings that MVcn expression leads to larger but fewer FAs per cell, in conjunction with the inability of MVcn to bundle F-actin in vitro and rescue the cell stiffening response, are consistent with our previous findings of actin bundling deficient Vcn variants, suggesting that deficient actin-bundling may account for some of the differences between Vcn and MVcn.
Collapse
Affiliation(s)
- Hyunna T. Lee
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Lisa Sharek
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - E. Timothy O’Brien
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Fabio L. Urbina
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Stephanie L. Gupton
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Richard Superfine
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Keith Burridge
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Sharon L. Campbell
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
58
|
Merkel CD, Li Y, Raza Q, Stolz DB, Kwiatkowski AV. Vinculin anchors contractile actin to the cardiomyocyte adherens junction. Mol Biol Cell 2019; 30:2639-2650. [PMID: 31483697 PMCID: PMC6761764 DOI: 10.1091/mbc.e19-04-0216] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The adherens junction (AJ) couples the actin cytoskeletons of neighboring cells to allow mechanical integration and tissue organization. The physiological demands of intercellular adhesion require that the AJ be responsive to dynamic changes in force while maintaining mechanical load. These demands are tested in the heart, where cardiomyocyte AJs must withstand repeated cycles of actomyosin-mediated contractile force. Here we show that force-responsive cardiomyocyte AJs recruit actin-binding ligands to selectively couple actin networks. We employed a panel of N-cadherin-αE-catenin fusion proteins to rebuild AJs with specific actin linkages in N-cadherin-null cardiomyocytes. In this system, vinculin recruitment was required to rescue myofibril integration at nascent contacts. In contrast, loss of vinculin from the AJ disrupted junction morphology and blocked myofibril integration at cell–cell contacts. Our results identify vinculin as a critical link to contractile actomyosin and offer insight to how actin integration at the AJ is regulated to provide stability under mechanical load.
Collapse
Affiliation(s)
- Chelsea D Merkel
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Yang Li
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Qanber Raza
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Donna B Stolz
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | - Adam V Kwiatkowski
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| |
Collapse
|
59
|
Chang LY, Fan SMY, Liao YC, Wang WH, Chen YJ, Lin SJ. Proteomic Analysis Reveals Anti-Fibrotic Effects of Blue Light Photobiomodulation on Fibroblasts. Lasers Surg Med 2019; 52:358-372. [PMID: 31321797 DOI: 10.1002/lsm.23137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES This study was aimed at determining the effects of blue light photobiomodulation on primary adult mouse dermal fibroblasts (AMDFs) and the associated signaling pathways. STUDY DESIGN/MATERIALS AND METHODS Cultured AMDFs from adult C57BL/6 mice were irradiated by blue light from a light-emitting diode (wavelength = 463 ± 50 nm; irradiance = 5 mW/cm2 ; energy density = 4-8 J/cm2 ). The cells were analyzed using mass spectrometry for proteomics/phosphoproteomics, AlamarBlue assay for mitochondrial activity, time-lapse video for cell migration, quantitative polymerase chain reaction for gene expression, and immunofluorescence for protein expression. RESULTS Proteomic/phosphoproteomic analysis showed inhibition of extracellular signal-regulated kinases/mammalian target of rapamycin and casein kinase 2 pathways, cell motility-related networks, and multiple metabolic processes, including carbon metabolism, biosynthesis of amino acid, glycolysis/gluconeogenesis, and the pentose phosphate pathway. Functional analysis demonstrated inhibition of mitochondrial activities, cell migration, and mitosis. Expression of growth promoting insulin-like growth factor 1 and fibrosis-related genes, including transforming growth factor β1 (TGFβ1) and collagen type 1 ɑ2 chain diminished. Protein expression of α-smooth muscle actin, an important regulator of myofibroblast functions, was also suppressed. CONCLUSIONS Low-level blue light exerted suppressive effects on AMDFs, including suppression of mitochondrial activity, metabolism, cell motility, proliferation, TGFβ1 levels, and collagen I production. Low-level blue light can be a potential treatment for the prevention and reduction of tissue fibrosis, such as hypertrophic scar and keloids. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lo-Yu Chang
- School of Medicine, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Sabrina Mai-Yi Fan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yen-Chen Liao
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yu-Ju Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei 100, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
60
|
Viswanathan A, Sebastianelli G, Brown K, Raunio J, Sipilä V, Yli-Harja O, Candeias NR, Kandhavelu M. In vitro anti-glioblastoma activity of L-valine derived boroxazolidones. Eur J Pharmacol 2019; 854:194-200. [PMID: 30981767 DOI: 10.1016/j.ejphar.2019.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
Abstract
In the present study, a series of L-valine derived boroxazolidones, previously synthesized and reported to have residual activity in a human epithelial cell line, have been evaluated in vitro for their anti-glioblastoma activity. A boroxazolidone derivative containing 2,4-difluorophenyl moieties (6) was found to have higher cytotoxicity than the standard drug, Temozolomide (TMZ). Compound 6 was found to exhibit dose-dependent growth inhibitory effects with an IC50 of 49 μM and 53 μM for LN229 and SNB19 cells, respectively. Additionally, 6 was assessed for its role in apoptosis, caspase 3/7 activation and oxidative stress in SNB19 and LN229 cells. SNB19 cells treated with 6 showed 45.3% apoptosis in the population, while TMZ had 24.7%. In LN229 cells, the percentage of apoptotic cells treated with compound 6 and TMZ were the same. Both 6 and TMZ induced apoptosis through the activation of caspase 3/7 in SNB19 and LN229 cells. Interestingly, 6 exhibited a higher effectivity in promoting reactive oxygen species production in LN229, while it was 6-fold less in SNB19. Boroxazolidone-treated GBM cell lines increased reactive oxygen species production, suggesting that such species may be interlinked with the observed programmed cell death. Additionally, the treatment of both GBM cell lines with 6 led to G2/M phase arrest. The magnitude of anti-GBM effect of 6 is significantly higher than the known chemotherapeutic agent TMZ. This work further demonstrates the anticancer properties of L-valine derived boroxazolidones, adding another potential derivative to the collection of promising chemotherapeutic agents for GBM treatment.
Collapse
Affiliation(s)
- Anisha Viswanathan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland
| | - Giulia Sebastianelli
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland
| | - Kenna Brown
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland
| | - Jenna Raunio
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33101 Tampere, Finland
| | - Vili Sipilä
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland
| | - Olli Yli-Harja
- Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University and BioMedi Tech, P.O. Box 553, 33101 Tampere, Finland; Institute for Systems Biology, 1441N 34th Street, Seattle, WA 98103-8904, USA
| | - Nuno R Candeias
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 8, 33101 Tampere, Finland.
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland.
| |
Collapse
|
61
|
Carvalho JR, Fortunato IC, Fonseca CG, Pezzarossa A, Barbacena P, Dominguez-Cejudo MA, Vasconcelos FF, Santos NC, Carvalho FA, Franco CA. Non-canonical Wnt signaling regulates junctional mechanocoupling during angiogenic collective cell migration. eLife 2019; 8:e45853. [PMID: 31246175 PMCID: PMC6684320 DOI: 10.7554/elife.45853] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022] Open
Abstract
Morphogenesis of hierarchical vascular networks depends on the integration of multiple biomechanical signals by endothelial cells, the cells lining the interior of blood vessels. Expansion of vascular networks arises through sprouting angiogenesis, a process involving extensive cell rearrangements and collective cell migration. Yet, the mechanisms controlling angiogenic collective behavior remain poorly understood. Here, we show this collective cell behavior is regulated by non-canonical Wnt signaling. We identify that Wnt5a specifically activates Cdc42 at cell junctions downstream of ROR2 to reinforce coupling between adherens junctions and the actin cytoskeleton. We show that Wnt5a signaling stabilizes vinculin binding to alpha-catenin, and abrogation of vinculin in vivo and in vitro leads to uncoordinated polarity and deficient sprouting angiogenesis in Mus musculus. Our findings highlight how non-canonical Wnt signaling coordinates collective cell behavior during vascular morphogenesis by fine-tuning junctional mechanocoupling between endothelial cells.
Collapse
Affiliation(s)
- Joana R Carvalho
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Isabela C Fortunato
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Catarina G Fonseca
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Anna Pezzarossa
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Pedro Barbacena
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | | | | | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| | - Claudio A Franco
- Instituto de Medicina Molecular, Faculdade de MedicinaUniversidade de LisboaLisbonPortugal
| |
Collapse
|
62
|
Zhou C, Wang Q, Zhang D, Cai L, Du W, Xie J. Compliant substratum modulates vinculin expression in focal adhesion plaques in skeletal cells. Int J Oral Sci 2019; 11:18. [PMID: 31152146 PMCID: PMC6544630 DOI: 10.1038/s41368-019-0052-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 02/05/2023] Open
Abstract
The biophysical properties of the extracellular matrix (ECM) dictate tissue-specific cell behaviour. In the skeleton system, bone shows the potential to adapt its architecture and contexture to environmental rigidity via the bone remodelling process, which involves chondrocytes, osteoblasts, osteoclasts, osteocytes and even peripheral bone marrow-derived stem/stromal cells (BMSCs). In the current study, we generated stiff (~1 014 ± 56) kPa, Young's modulus) and soft (~46 ± 11) kPa silicon-based elastomer polydimethylsiloxane (PDMS) substrates by mixing curing agent into oligomeric base at 1:5 and 1:45 ratios, respectively, and investigated the influence of substrate stiffness on the cell behaviours by characterizing cell spreading area, cell cytoskeleton and cell adhesion capacity. The results showed that the cell spreading areas of chondrocytes, osteoblasts, osteoclasts, osteocytes and BMSCs were all reduced in the soft substrate relative to those in the stiff substrate. F-actin staining confirmed that the cytoskeleton was also changed in the soft group compared to that in the stiff group. Vinculin in focal adhesion plaques was significantly decreased in response to soft substrate compared to stiff substrate. This study establishes the potential correlation between microenvironmental mechanics and the skeletal system, and the results regarding changes in cell spreading area, cytoskeleton and cell adhesion further indicate the important role of biomechanics in the cell-matrix interaction.
Collapse
Affiliation(s)
- Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qingxuan Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
63
|
Zhang M, Liu P, Xu F, He Y, Xie X, Jiang X. Vinculin promotes gastric cancer proliferation and migration and predicts poor prognosis in patients with gastric cancer. J Cell Biochem 2019; 120:14107-14115. [PMID: 30989694 DOI: 10.1002/jcb.28686] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/10/2019] [Accepted: 02/14/2019] [Indexed: 12/17/2022]
Abstract
Vinculin is a highly conserved protein involved in cell proliferation, migration, and adhesion. However, the effects of vinculin on gastric cancer (GC) remain unclear. Therefore, we aimed to explore the functional role of vinculin in GC, as well as its underlying mechanism. Expression of vinculin in patients with GC was analyzed by real-time polymerase chain reaction, Western blot analysis, and immunohistochemistry. Overall survival was evaluated by the Kaplan-Meier method with the log-rank test. The relationship between vinculin and clinicopathological characteristics of patients with GC was further identified. In addition, we assessed the expression of vinculin in GC cell lines. Besides, vinculin was suppressed or overexpressed by transfection with small interfering (si-vinculin) or pcDNA-vinculin and then cell viability, cell apoptosis, and/or migration was respectively examined by the 3-(4, 5-dimethylthiazole-2-yl)-2, 5-biphenyl tetrazolium bromide assay, flow cytometer, and scratch assay, respectively. Moreover, the cell cycle- and apoptosis-related proteins were detected by Western blot analysis. The expression of vinculin was significantly increased in the GC tissues and cells compared with the nontumor tissues or cells. Vinculin protein positive staining was mainly located in the cell membrane and cytoplasm. Moreover, vinculin was significantly associated with Tumor Node Metastasis (TNM) and poor differentiation. Patients with high vinculin levels had significantly worse overall survival than those with low levels. Suppression of vinculin significantly decreased cell viability and migration and promoted cell apoptosis. However, overexpression of vinculin statistically increased cell viability but had no effects on cell apoptosis. Vinculin promotes GC proliferation and migration and predicts poor prognosis in patients with GC.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Gastroenterology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China
| | - Pei Liu
- Department of Infectious Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Famei Xu
- Department of Pathology, Zibo Central Hospital, Zibo, Shandong, China
| | - Yuanlong He
- Department of Gastroenterology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiangjun Xie
- Department of Gastroenterology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiangjun Jiang
- Department of Gastroenterology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
64
|
Rahikainen R, Öhman T, Turkki P, Varjosalo M, Hytönen VP. Talin-mediated force transmission and talin rod domain unfolding independently regulate adhesion signaling. J Cell Sci 2019; 132:jcs226514. [PMID: 30837291 DOI: 10.1242/jcs.226514] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/26/2019] [Indexed: 12/29/2022] Open
Abstract
Talin protein is one of the key components in integrin-mediated adhesion complexes. Talins transmit mechanical forces between β-integrin and actin, and regulate adhesion complex composition and signaling through the force-regulated unfolding of talin rod domain. Using modified talin proteins, we demonstrate that these functions contribute to different cellular processes and can be dissected. The transmission of mechanical forces regulates adhesion complex composition and phosphotyrosine signaling even in the absence of the mechanically regulated talin rod subdomains. However, the presence of the rod subdomains and their mechanical activation are required for the reinforcement of the adhesion complex, cell polarization and migration. Talin rod domain unfolding was also found to be essential for the generation of cellular signaling anisotropy, since both insufficient and excess activity of the rod domain severely inhibited cell polarization. Utilizing proteomics tools, we identified adhesome components that are recruited and activated either in a talin rod-dependent manner or independently of the rod subdomains. This study clarifies the division of roles between the force-regulated unfolding of a talin protein (talin 1) and its function as a physical linker between integrins and the cytoskeleton.
Collapse
Affiliation(s)
- Rolle Rahikainen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere 33014, Finland
- Fimlab Laboratories, Tampere 33520, Finland
| | - Tiina Öhman
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
| | - Paula Turkki
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere 33014, Finland
- Fimlab Laboratories, Tampere 33520, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere 33014, Finland
- Fimlab Laboratories, Tampere 33520, Finland
| |
Collapse
|
65
|
Sarker M, Lee HT, Mei L, Krokhotin A, de Los Reyes SE, Yen L, Costantini LM, Griffith J, Dokholyan NV, Alushin GM, Campbell SL. Cardiomyopathy Mutations in Metavinculin Disrupt Regulation of Vinculin-Induced F-Actin Assemblies. J Mol Biol 2019; 431:1604-1618. [PMID: 30844403 DOI: 10.1016/j.jmb.2019.02.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022]
Abstract
Debilitating heart conditions, notably dilated and hypertrophic cardiomyopathies (CMs), are associated with point mutations in metavinculin, a larger isoform of the essential cytoskeletal protein vinculin. Metavinculin is co-expressed with vinculin at sub-stoichiometric ratios in cardiac tissues. CM mutations in the metavinculin tail domain (MVt) occur within the extra 68-residue insert that differentiates it from the vinculin tail domain (Vt). Vt binds actin filaments (F-actin) and promotes vinculin dimerization to bundle F-actin into thick fibers. While MVt binds to F-actin in a similar manner to Vt, MVt is incapable of F-actin bundling and inhibits Vt-mediated F-actin bundling. We performed F-actin co-sedimentation and negative-stain EM experiments to dissect the coordinated roles of metavinculin and vinculin in actin fiber assembly and the effects of three known metavinculin CM mutations. These CM mutants were found to weakly induce the formation of disordered F-actin assemblies. Notably, they fail to inhibit Vt-mediated F-actin bundling and instead promote formation of large assemblies embedded with linear bundles. Computational models of MVt bound to F-actin suggest that MVt undergoes a conformational change licensing the formation of a protruding sub-domain incorporating the insert, which sterically prevents dimerization and bundling of F-actin by Vt. Sub-domain formation is destabilized by CM mutations, disrupting this inhibitory mechanism. These findings provide new mechanistic insights into the ability of metavinculin to tune actin organization by vinculin and suggest that dysregulation of this process by CM mutants could underlie their malfunction in disease.
Collapse
Affiliation(s)
- Muzaddid Sarker
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hyunna T Lee
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lin Mei
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Andrey Krokhotin
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Laura Yen
- Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY 10025, USA
| | - Lindsey M Costantini
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jack Griffith
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
66
|
Krüger-Genge A, Dietze S, Yan W, Liu Y, Fang L, Kratz K, Lendlein A, Jung F. Endothelial cell migration, adhesion and proliferation on different polymeric substrates. Clin Hemorheol Microcirc 2019; 70:511-529. [DOI: 10.3233/ch-189317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Anne Krüger-Genge
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Stefanie Dietze
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Wan Yan
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Yue Liu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Liang Fang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| | - Friedrich Jung
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| |
Collapse
|
67
|
Deacon DC, Happe CL, Chen C, Tedeschi N, Manso AM, Li T, Dalton ND, Peng Q, Farah EN, Gu Y, Tenerelli KP, Tran VD, Chen J, Peterson KL, Schork NJ, Adler ED, Engler AJ, Ross RS, Chi NC. Combinatorial interactions of genetic variants in human cardiomyopathy. Nat Biomed Eng 2019; 3:147-157. [PMID: 30923642 PMCID: PMC6433174 DOI: 10.1038/s41551-019-0348-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of morbidity and mortality worldwide; yet how genetic variation and environmental factors impact DCM heritability remains unclear. Here, we report that compound genetic interactions between DNA sequence variants contribute to the complex heritability of DCM. By using genetic data from a large family with a history of DCM, we discovered that heterozygous sequence variants in the TROPOMYOSIN 1 (TPM1) and VINCULIN (VCL) genes cose-gregate in individuals affected by DCM. In vitro studies of patient-derived and isogenic human-pluripotent-stem-cell-derived cardio-myocytes that were genome-edited via CRISPR to create an allelic series of TPM1 and VCL variants revealed that cardiomyocytes with both TPM1 and VCL variants display reduced contractility and sarcomeres that are less organized. Analyses of mice genetically engineered to harbour these human TPM1 and VCL variants show that stress on the heart may also influence the variable penetrance and expressivity of DCM-associated genetic variants in vivo. We conclude that compound genetic variants can interact combinatorially to induce DCM, particularly when influenced by other disease-provoking stressors.
Collapse
Affiliation(s)
- Dekker C Deacon
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Cassandra L Happe
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Chao Chen
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Neil Tedeschi
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ana Maria Manso
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Veterans Administration Healthcare San Diego, San Diego, CA, USA
| | - Ting Li
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nancy D Dalton
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Qian Peng
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- Department of Human Biology, J. Craig Venter Institute, La Jolla, CA, USA
| | - Elie N Farah
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yusu Gu
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kevin P Tenerelli
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Vivien D Tran
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Ju Chen
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kirk L Peterson
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nicholas J Schork
- Department of Human Biology, J. Craig Venter Institute, La Jolla, CA, USA
| | - Eric D Adler
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA.
| | - Robert S Ross
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Veterans Administration Healthcare San Diego, San Diego, CA, USA.
| | - Neil C Chi
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
68
|
Pinheiro D, Bellaïche Y. Mechanical Force-Driven Adherens Junction Remodeling and Epithelial Dynamics. Dev Cell 2019; 47:3-19. [PMID: 30300588 DOI: 10.1016/j.devcel.2018.09.014] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/02/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022]
Abstract
During epithelial tissue development, repair, and homeostasis, adherens junctions (AJs) ensure intercellular adhesion and tissue integrity while allowing for cell and tissue dynamics. Mechanical forces play critical roles in AJs' composition and dynamics. Recent findings highlight that beyond a well-established role in reinforcing cell-cell adhesion, AJ mechanosensitivity promotes junctional remodeling and polarization, thereby regulating critical processes such as cell intercalation, division, and collective migration. Here, we provide an integrated view of mechanosensing mechanisms that regulate cell-cell contact composition, geometry, and integrity under tension and highlight pivotal roles for mechanosensitive AJ remodeling in preserving epithelial integrity and sustaining tissue dynamics.
Collapse
Affiliation(s)
- Diana Pinheiro
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | - Yohanns Bellaïche
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne Universités, UPMC Univ Paris 06, CNRS, CNRS UMR 3215, INSERM U934, 75005 Paris, France.
| |
Collapse
|
69
|
Rosowski KA, Boltyanskiy R, Xiang Y, Van den Dries K, Schwartz MA, Dufresne ER. Vinculin and the mechanical response of adherent fibroblasts to matrix deformation. Sci Rep 2018; 8:17967. [PMID: 30568231 PMCID: PMC6299284 DOI: 10.1038/s41598-018-36272-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/14/2018] [Indexed: 12/21/2022] Open
Abstract
Cells respond to the mechanics of their environment. Mechanical cues include extracellular matrix (ECM) stiffness and deformation, which are primarily sensed through integrin-mediated adhesions. We investigated the impact of ECM deformation on cellular forces, measuring the time-evolution of traction forces of isolated mouse fibroblasts in response to stretch and release. Stretch triggered a marked increase of traction stresses and apparent stiffness. Expression of the focal adhesion protein vinculin not only increased baseline traction forces, but also increased dissipation of mechanical energy, which was correlated with the cells’ failure to recover baseline traction forces after release of stretch.
Collapse
Affiliation(s)
- Kathryn A Rosowski
- Department of Materials, ETH Zürich, 8093, Zürich, Switzerland.,Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06511, USA
| | - Rostislav Boltyanskiy
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06511, USA
| | - Yingjie Xiang
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06511, USA
| | - Koen Van den Dries
- Cardiovascular Research Center and Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT, 06511, USA.,Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin A Schwartz
- Cardiovascular Research Center and Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT, 06511, USA.,Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, 06511, USA
| | - Eric R Dufresne
- Department of Materials, ETH Zürich, 8093, Zürich, Switzerland. .,Department of Mechanical Engineering and Materials Science, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
70
|
Li T, Zheng Y, Li Y, Ye D. Comparison of in vitro Neuronal Differentiation Capacity Between Mouse Epiblast Stem Cells Derived From Nuclear Transfer and Naturally Fertilized Embryos. Front Mol Neurosci 2018; 11:392. [PMID: 30425619 PMCID: PMC6218595 DOI: 10.3389/fnmol.2018.00392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/09/2018] [Indexed: 11/18/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) can give rise to fertile adults, but the successful perinatal and postnatal developmental rates are inefficient, including delayed developmental behaviors, and respiratory failure. However, the molecular and cellular mechanisms remain elusive. Mouse epiblast stem cells (mEpiSCs) from E5.5-6.5 epiblasts share defining features with human embryonic stem cells (hESCs), providing a new opportunity to study early mammalian development in vitro. In this study, mEpiSCs were established from naturally fertilized mouse embryos (F-mEpiSCs) and SCNT mouse embryos (NT-mEpiSCs). Also, the in vitro neuronal differentiation capacity of F-mEpiSCs and NT-mEpiSCs was compared. Morphology analysis showed less and smaller neurospheres formation and lower percentage of early neurons generation in NT-mEpiSCs. The immunocytochemical analysis and altered mRNA expression levels of the neuronal markers in differentiated cells further confirmed that neurogenesis was slower in NT-mEpiSCs than in F-mEpiSCs. Moreover, neuronal differentiation capacity was correlated with the basal expression levels of Atox1 and Vinculin but not Brachyury and Otx2, emphasizing that developmental aberrations in neurogenesis were associated with the NT technique but not random variations between clones. This study provided an important in vitro platform using mEpiSCs to study early epigenetic and developmental processes associated with neurogenesis.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of Ophthalmology and Vision Science, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yi Zheng
- Department of Reproductive Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Li
- Department of Reproductive Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Danna Ye
- Department of Reproductive Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
71
|
Bai M, Xie J, Liu X, Chen X, Liu W, Wu F, Chen D, Sun Y, Li X, Wang C, Ye L. Microenvironmental Stiffness Regulates Dental Papilla Cell Differentiation: Implications for the Importance of Fibronectin-Paxillin-β-Catenin Axis. ACS APPLIED MATERIALS & INTERFACES 2018; 10:26917-26927. [PMID: 30004214 DOI: 10.1021/acsami.8b08450] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The mechanical stiffness of substrates is recognized to be an important physical cue in the microenvironment of local cellular residents in mammalian species due to their great capacity in regulating cell behavior. Dental papilla cells (DPCs) play an important role in the field of dental tissue engineering for their stem cell-like properties. Therefore, it is essential to provide the suitable microenvironment by combining with the physical cues of biomaterials for DPCs to carry out the function of effective tissue regeneration. However, how the substrate stiffness influences the odontogenic differentiation of DPCs is still unclear. Thus, we fabricated poly(dimethylsiloxane) substrates with varied stiffness for cell behavior. Both cell morphology and focal adhesion were shown to have significant changes in response to varied stiffness. Paxillin, an important protein adapter of focal adhesion kinase protein, was shown to interact with both ectoplasmic fibronectin and cytoplasmic β-catenin by coimmunoprecipitation. The resultant changes of β-catenin by varied stiffness were confirmed by immunofluorescent stain and western blotting. Further, the higher quantity nuclear translocation of β-catenin and the less phospho-β-catenin on the stiff substrate were detected. This nuclear translocation in the stiff substrate finally led to an increased mineralization of DPCs relative to the soft substrate detected by Von Kossa and Alizarin Red stain. Taken together, this work not only points out that the substrate stiffness can regulate the odontogenic differentiation potential of DPCs via fibronectin/paxillin/β-catenin pathway but also provides significant consequence for biomechanical control of cell behavior in cell-based tooth tissue regeneration.
Collapse
Affiliation(s)
- Mingru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Xiaoyu Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Xia Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Wenjing Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Dian Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Yimin Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| |
Collapse
|
72
|
Haining AWM, Rahikainen R, Cortes E, Lachowski D, Rice A, von Essen M, Hytönen VP, del Río Hernández A. Mechanotransduction in talin through the interaction of the R8 domain with DLC1. PLoS Biol 2018; 16:e2005599. [PMID: 30028837 PMCID: PMC6054372 DOI: 10.1371/journal.pbio.2005599] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 06/19/2018] [Indexed: 11/19/2022] Open
Abstract
The mechanical unfolding of proteins is a cellular mechanism for force transduction with potentially broad implications in cell fate. Despite this, the mechanism by which protein unfolding elicits differential downstream signalling pathways remains poorly understood. Here, we used protein engineering, atomic force microscopy, and biophysical tools to delineate how protein unfolding controls cell mechanics. Deleted in liver cancer 1 (DLC1) is a negative regulator of Ras homolog family member A (RhoA) and cell contractility that regulates cell behaviour when localised to focal adhesions bound to folded talin. Using a talin mutant resistant to force-induced unfolding of R8 domain, we show that talin unfolding determines DLC1 downstream signalling and, consequently, cell mechanics. We propose that this new mechanism of mechanotransduction may have implications for a wide variety of associated cellular processes.
Collapse
Affiliation(s)
- Alexander William M. Haining
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Rolle Rahikainen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Magdalena von Essen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Armando del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
73
|
Certhrax Is an Antivirulence Factor for the Anthrax-Like Organism Bacillus cereus Strain G9241. Infect Immun 2018; 86:IAI.00207-18. [PMID: 29610258 DOI: 10.1128/iai.00207-18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 03/28/2018] [Indexed: 11/20/2022] Open
Abstract
Bacillus cereus G9241 caused a life-threatening anthrax-like lung infection in a previously healthy human. This strain harbors two large virulence plasmids, pBCXO1 and pBC210, that are absent from typical B. cereus isolates. The pBCXO1 plasmid is nearly identical to pXO1 from Bacillus anthracis and carries genes (pagA1, lef, and cya) for anthrax toxin components (protective antigen [called PA1 in G9241], lethal factor [LF], and edema factor [EF], respectively). The plasmid also has an intact hyaluronic acid capsule locus. The pBC210 plasmid has a tetrasaccharide capsule locus, a gene for a PA1 homolog called PA2 (pagA2), and a gene (cer) for Certhrax, an ADP-ribosyltransferase toxin that inactivates vinculin. LF, EF, and Certhrax require PA for entry into cells. In this study, we asked what role PA1, PA2, LF, and Certhrax play in the pathogenicity of G9241. To answer this, we generated isogenic deletion mutations in the targeted toxin gene components and then assessed the strains for virulence in highly G9241-susceptible (A/J) and moderately G9241-sensitive (C57BL/6) mice. We found that full virulence of G9241 required PA1 and LF, while PA2 contributed minimally to pathogenesis of G9241 but could not functionally replace PA1 as a toxin-binding subunit in vivo Surprisingly, we discovered that Certhrax attenuated the virulence of G9241; i.e., a Δcer Δlef mutant strain was more virulent than a Δlef mutant strain following subcutaneous inoculation of A/J mice. Moreover, the enzymatic activity of Certhrax contributed to this phenotype. We concluded that Certhrax acts as an antivirulence factor in the anthrax-like organism B. cereus G9241.
Collapse
|
74
|
López-Escobar B, Caro-Vega JM, Vijayraghavan DS, Plageman TF, Sanchez-Alcazar JA, Moreno RC, Savery D, Márquez-Rivas J, Davidson LA, Ybot-González P. The non-canonical Wnt-PCP pathway shapes the mouse caudal neural plate. Development 2018; 145:dev.157487. [PMID: 29636380 DOI: 10.1242/dev.157487] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/03/2018] [Indexed: 01/03/2023]
Abstract
The last stage of neural tube (NT) formation involves closure of the caudal neural plate (NP), an embryonic structure formed by neuromesodermal progenitors and newly differentiated cells that becomes incorporated into the NT. Here, we show in mouse that, as cell specification progresses, neuromesodermal progenitors and their progeny undergo significant changes in shape prior to their incorporation into the NT. The caudo-rostral progression towards differentiation is coupled to a gradual reliance on a unique combination of complex mechanisms that drive tissue folding, involving pulses of apical actomyosin contraction and planar polarised cell rearrangements, all of which are regulated by the Wnt-PCP pathway. Indeed, when this pathway is disrupted, either chemically or genetically, the polarisation and morphology of cells within the entire caudal NP is disturbed, producing delays in NT closure. The most severe disruptions of this pathway prevent caudal NT closure and result in spina bifida. In addition, a decrease in Vangl2 gene dosage also appears to promote more rapid progression towards a neural fate, but not the specification of more neural cells.
Collapse
Affiliation(s)
- Beatriz López-Escobar
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - José Manuel Caro-Vega
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | | | | | - José A Sanchez-Alcazar
- Centro Andaluz de Biología del Desarrollo (CABD), and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC, Sevilla 41013, Spain
| | - Roberto Carlos Moreno
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - Dawn Savery
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Javier Márquez-Rivas
- Unidad de Gestión Clínica de Neurocirugía, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - Lance A Davidson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Patricia Ybot-González
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain .,Unidad de Gestión Clínica de Neurología y Neurofisiología, Hospital Universitario Virgen Macarena, Sevilla 41009, Spain
| |
Collapse
|
75
|
Ozawa M. Nonmuscle myosin IIA is involved in recruitment of apical junction components through activation of α-catenin. Biol Open 2018; 7:bio.031369. [PMID: 29654115 PMCID: PMC5992523 DOI: 10.1242/bio.031369] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
MDCK dog kidney epithelial cells express two isoforms of nonmuscle myosin heavy chain II, IIA and IIB. Using the CRISPR/Cas9 system, we established cells in which the IIA gene was ablated. These cells were then transfected with a vector that expresses GFP-IIA chimeric molecule under the control of a tetracycline-responsible element. In the absence of Dox (doxycyclin), when GFP-IIA is expressed (GFP-IIA+), the cells exhibit epithelial cell morphology, but in the presence of Dox, when expression of GFP-IIA is repressed (GFP-IIA-), the cells lose epithelial morphology and strong cell-cell adhesion. Consistent with these observations, GFP-IIA- cells failed to assemble junction components such as E-cadherin, desmoplakin, and occludin at cell-cell contact sites. Therefore, IIA is required for assembly of junction complexes. MDCK cells with an ablation of the α-catenin gene also exhibited the same phenotype. However, when in GFP-IIA- cells expressed α-catenin lacking the inhibitory region or E-cadherin/α-catenin chimeras, the cells acquired the ability to establish the junction complex. These experiments reveal that IIA acts as an activator of α-catenin in junction assembly.
Collapse
Affiliation(s)
- Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
76
|
Maziveyi M, Alahari SK. Cell matrix adhesions in cancer: The proteins that form the glue. Oncotarget 2018; 8:48471-48487. [PMID: 28476046 PMCID: PMC5564663 DOI: 10.18632/oncotarget.17265] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/27/2017] [Indexed: 12/28/2022] Open
Abstract
The main purposes of Integrin-mediated cell contacts are to interpret bi-directional signals between the extracellular environment and intracellular proteins, as well as, anchor the cell to a matrix. Many cell adhesion molecules have been discovered with a wide spectrum of responsibilities, including recruiting, activating, elongating, and maintaining. This review will perlustrate some of the key incidences that precede focal adhesion formation. Tyrosine phosphorylation is a key signaling initiation event that leads to the recruitment of multiple proteins to focal adhesion sites. Recruitment and concentration of proteins such as Paxillin and Vinculin to Integrin clutches is necessary for focal adhesion development. The assembled networks are responsible for transmitting signals back and forth from the extracellular matrix (ECM) to Actin and its binding proteins. Cancer cells exhibit highly altered focal adhesion dynamics. This review will highlight some key discoveries in cancer cell adhesion, as well as, identify current gaps in knowledge.
Collapse
Affiliation(s)
- Mazvita Maziveyi
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
77
|
Kindt LM, Coughlin AR, Perosino TR, Ersfeld HN, Hampton M, Liang JO. Identification of transcripts potentially involved in neural tube closure using RNA sequencing. Genesis 2018; 56:e23096. [PMID: 29488319 DOI: 10.1002/dvg.23096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 02/02/2018] [Accepted: 02/19/2018] [Indexed: 01/08/2023]
Abstract
Anencephaly is a fatal human neural tube defect (NTD) in which the anterior neural tube remains open. Zebrafish embryos with reduced Nodal signaling display an open anterior neural tube phenotype that is analogous to anencephaly. Previous work from our laboratory suggests that Nodal signaling acts through induction of the head mesendoderm and mesoderm. Head mesendoderm/mesoderm then, through an unknown mechanism, promotes formation of the polarized neuroepithelium that is capable of undergoing the movements required for closure. We compared the transcriptome of embryos treated with a Nodal signaling inhibitor at sphere stage, which causes NTDs, to embryos treated at 30% epiboly, which does not cause NTDs. This screen identified over 3,000 transcripts with potential roles in anterior neurulation. Expression of several genes encoding components of tight and adherens junctions was significantly reduced, supporting the model that Nodal signaling regulates formation of the neuroepithelium. mRNAs involved in Wnt, FGF, and BMP signaling were also differentially expressed, suggesting these pathways might regulate anterior neurulation. In support of this, we found that pharmacological inhibition of FGF-receptor function causes an open anterior NTD as well as loss of mesodermal derivatives. This suggests that Nodal and FGF signaling both promote anterior neurulation through induction of head mesoderm.
Collapse
Affiliation(s)
- Lexy M Kindt
- Department of Biology, University of Minnesota Duluth, Duluth.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth
| | - Alicia R Coughlin
- Department of Biology, University of Minnesota Duluth, Duluth.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth
| | | | - Haley N Ersfeld
- Department of Biology, University of Minnesota Duluth, Duluth
| | - Marshall Hampton
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth.,Department of Mathematics and Statistics, University of Minnesota Duluth, Duluth
| | - Jennifer O Liang
- Department of Biology, University of Minnesota Duluth, Duluth.,Integrated Biosciences Graduate Program, University of Minnesota, Duluth
| |
Collapse
|
78
|
Chen Z, Xu N, Chong D, Guan S, Jiang C, Yang Z, Li C. Geranylgeranyl pyrophosphate synthase facilitates the organization of cardiomyocytes during mid-gestation through modulating protein geranylgeranylation in mouse heart. Cardiovasc Res 2018; 114:965-978. [DOI: 10.1093/cvr/cvy042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 02/09/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Zhong Chen
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Na Xu
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Danyang Chong
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Shan Guan
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Chen Jiang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Zhongzhou Yang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| | - Chaojun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, #22 Hankou Road, Nanjing, Jiangsu 210093, People’s Republic of China
| |
Collapse
|
79
|
Liu H, Jia L, Guo W, Sun Y, Zhu R, Li S, Qu G, Jiang H, Wang J, Gu J, Sun C, Feng X, Han W, Lei L. Differential Protein Profiling of Cerebrospinal Fluid in Piglets with Severe Meningoencephalitis Caused by Streptococcus suis Type 2 Compared to Controls. Front Cell Infect Microbiol 2018; 8:35. [PMID: 29479521 PMCID: PMC5811643 DOI: 10.3389/fcimb.2018.00035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/25/2018] [Indexed: 11/19/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a zoonotic pathogen that can cause meningitis both in pigs and in human beings. However, the pathogenesis of central nervous system (CNS) infection caused by SS2 have not yet been elucidated. To find the key molecules in cerebrospinal fluid (CSF) needed for the pathogenesis, a SS2 meningoencephalitic pig model and a SS2 non-meningoencephalitic pig model were established in this study. CSF was collected from infected piglets, and protein profiling was performed with label-free proteomics technology. A total of 813 differential proteins, including 52 up-regulated proteins and 761 down-regulated proteins, were found in the CSF of meningoencephalitic pigs compared with both non-meningoencephalitic pigs and healthy pigs. These 813 differential proteins were clustered into three main categories, namely, cellular component, biological process, and molecular function by gene ontology (GO) analysis. The most enriched subclasses of differential proteins in each category were exosome (44.3%), energy pathway (25.0%) and catalytic activity (11.3%), respectively. The most enriched subclasses of upregulated proteins were extracellular (62.1%), protein metabolism (34.5%) and cysteine-type peptidase activity (6.9%), and of downregulated proteins were exosomes (45.0%), energy pathway (24.0%) and catalytic activity (9.4%). Then, the differential proteins were further investigated by using the KEGG database and were found to participate in 16 KEGGs. The most enriched KEGG was citrate cycle (56.6%), and some of these differential proteins are associated with brain diseases such as Huntington's disease (18.6%), Parkinson's disease (23.8%) and Alzheimer's disease (17.6%). Sixteen of the 813 differential proteins, chosen randomly as examples, were further confirmed by enzyme-linked immunosorbent assay (ELISA) to support the proteomic data. To our knowledge, this is the first study to analyze the differential protein profiling of CSF between SS2 meningoencephalitic piglets and non-meningoencephalitic piglets by employing proteomic technology. The discovery and bioinformatics analysis of these differential proteins provides reference data not only for research on pathogenesis of SS2 CNS infection but also for diagnosis and drug therapy research.
Collapse
Affiliation(s)
- Hongtao Liu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Jia
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenfei Guo
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yingying Sun
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Rining Zhu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shuguang Li
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Guanggang Qu
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, China
| | - Hexiang Jiang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Junjie Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingmin Gu
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Changjiang Sun
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Feng
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenyu Han
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Liancheng Lei
- College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
80
|
Juan-Rivera MC, Martínez-Ferrer M. Integrin Inhibitors in Prostate Cancer. Cancers (Basel) 2018; 10:E44. [PMID: 29415418 PMCID: PMC5836076 DOI: 10.3390/cancers10020044] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 01/20/2023] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer and the third highest cause of cancer-related deaths in men in the U.S. The development of chemotherapeutic agents that can bind PCa tumor cells with high specificity is critical in order to increase treatment effectiveness. Integrin receptors and their corresponding ligands have different expression patterns in PCa cells. They have been identified as promising targets to inhibit pathways involved in PCa progression. Currently, several compounds have proven to target specific integrins and their subunits in PCa cells. In this article, we review the role of integrins inhibitors in PCa and their potential as therapeutic targets for PCa treatments. We have discussed the following: natural compounds, monoclonal antibodies, statins, campothecins analog, aptamers, d-aminoacid, and snake venom. Recent studies have shown that their mechanisms of action result in decrease cell migration, cell invasion, cell proliferation, and metastasis of PCa cells.
Collapse
Affiliation(s)
- Maylein C Juan-Rivera
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA.
- University of Puerto Rico Comprehensive Cancer Center, Medical Sciences Campus, San Juan, PR 00936, USA.
| | - Magaly Martínez-Ferrer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA.
- University of Puerto Rico Comprehensive Cancer Center, Medical Sciences Campus, San Juan, PR 00936, USA.
| |
Collapse
|
81
|
Chopra A, Kutys ML, Zhang K, Polacheck WJ, Sheng CC, Luu RJ, Eyckmans J, Hinson JT, Seidman JG, Seidman CE, Chen CS. Force Generation via β-Cardiac Myosin, Titin, and α-Actinin Drives Cardiac Sarcomere Assembly from Cell-Matrix Adhesions. Dev Cell 2018; 44:87-96.e5. [PMID: 29316444 DOI: 10.1016/j.devcel.2017.12.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/16/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
Truncating mutations in the sarcomere protein titin cause dilated cardiomyopathy due to sarcomere insufficiency. However, it remains mechanistically unclear how these mutations decrease sarcomere content in cardiomyocytes. Utilizing human induced pluripotent stem cell-derived cardiomyocytes, CRISPR/Cas9, and live microscopy, we characterize the fundamental mechanisms of human cardiac sarcomere formation. We observe that sarcomerogenesis initiates at protocostameres, sites of cell-extracellular matrix adhesion, where nucleation and centripetal assembly of α-actinin-2-containing fibers provide a template for the fusion of Z-disk precursors, Z bodies, and subsequent striation. We identify that β-cardiac myosin-titin-protocostamere form an essential mechanical connection that transmits forces required to direct α-actinin-2 centripetal fiber assembly and sarcomere formation. Titin propagates diastolic traction stresses from β-cardiac myosin, but not α-cardiac myosin or non-muscle myosin II, to protocostameres during sarcomerogenesis. Ablating protocostameres or decoupling titin from protocostameres abolishes sarcomere assembly. Together these results identify the mechanical and molecular components critical for human cardiac sarcomerogenesis.
Collapse
Affiliation(s)
- Anant Chopra
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Matthew L Kutys
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - William J Polacheck
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Calvin C Sheng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rebeccah J Luu
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Cardiology Center, University of Connecticut Health, Farmington, CT 06030, USA.
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
82
|
Junghans D, Herzog S. Cnn3 regulates neural tube morphogenesis and neuronal stem cell properties. FEBS J 2018; 285:325-338. [PMID: 29151265 DOI: 10.1111/febs.14338] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/25/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Abstract
Calponin 3 (Cnn3) is a member of the Cnn family of actin-binding molecules that is highly expressed in the mammalian brain and has been shown to control dendritic spine morphology, density, and plasticity by regulating actin cytoskeletal reorganization and dynamics. However, little is known about the role of Cnn3 during embryonic development. In this study, we analyzed mutant animals deficient in Cnn3 to gain a better understanding of its role in brain morphogenesis. Embryos lacking Cnn3 exhibited massive malformation of the developing brain including exoencephaly, closure defects at the rostral neural tube, and strong enlargement of brain tissue. In wild-type animals, we found Cnn3 being localized to the apical lining of the neuroepithelium in close vicinity to beta-Catenin and N-cadherin. By performing immunohistochemistry on beta-Catenin and p-Smad, and furthermore taking advantage of Wnt-reporter animals, we provide evidence that the loss of Cnn3 during development can affect signaling pathways crucial for correct morphogenesis of the neural tube. In addition, we used embryonic neurosphere cultures to investigate the role of Cnn3 in embryonic neuronal stem cells (NSC). Here, we observed that Cnn3 deficiency in NSCs increased the number of newly formed neurospheres and increased neurosphere size without perturbing their differentiation potential. Together, our study provides evidence for an important role of Cnn3 during development of the embryonic brain and in regulating NSC function.
Collapse
Affiliation(s)
- Dirk Junghans
- Institute of Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Switzerland
| | - Sebastian Herzog
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Austria
| |
Collapse
|
83
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
84
|
Lausecker F, Tian X, Inoue K, Wang Z, Pedigo CE, Hassan H, Liu C, Zimmer M, Jinno S, Huckle AL, Hamidi H, Ross RS, Zent R, Ballestrem C, Lennon R, Ishibe S. Vinculin is required to maintain glomerular barrier integrity. Kidney Int 2017; 93:643-655. [PMID: 29241625 PMCID: PMC5846847 DOI: 10.1016/j.kint.2017.09.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/23/2017] [Accepted: 09/14/2017] [Indexed: 01/08/2023]
Abstract
Cell-matrix interactions and podocyte intercellular junctions are key for maintaining the glomerular filtration barrier. Vinculin, a cytoplasmic protein, couples actin filaments to integrin-mediated cell-matrix adhesions and to cadherin-based intercellular junctions. Here, we examined the role of vinculin in podocytes by the generation of a podocyte-specific knockout mouse. Mice lacking podocyte vinculin had increased albuminuria and foot process effacement following injury in vivo. Analysis of primary podocytes isolated from the mutant mice revealed defects in cell protrusions, altered focal adhesion size and signaling, as well as impaired cell migration. Furthermore, we found a marked mislocalization of the intercellular junction protein zonula occludens-1. In kidney sections from patients with focal segmental glomerulosclerosis, minimal change disease and membranous nephropathy, we observed dramatic differences in the expression levels and localization of vinculin. Thus, our results suggest that vinculin is necessary to maintain the integrity of the glomerular filtration barrier by modulating podocyte foot processes and stabilizing intercellular junctions.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhen Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Christopher E Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hossam Hassan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chang Liu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Margaret Zimmer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie Jinno
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Abby L Huckle
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hellyeh Hamidi
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert S Ross
- Department of Medicine/Cardiology, University of California, San Diego, School of Medicine, La Jolla, California, USA and Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Roy Zent
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Hospital, Nashville, Tennessee, USA
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; School of Biology, Faculty of Biology, Medicine and Health, University of Medicine, Manchester Academic Health Science Centre, UK.
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
85
|
Zhang SM, Yu LL, Qu T, Hu Y, Yuan DZ, Zhang S, Xu Q, Zhao YB, Zhang JH, Yue LM. The Changes of Cytoskeletal Proteins Induced by the Fast Effect of Estrogen in Mouse Blastocysts and Its Roles in Implantation. Reprod Sci 2017; 24:1639-1646. [PMID: 28299994 DOI: 10.1177/1933719117697126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
It is necessary for estrogen to activate mouse blastocysts, so that they can attach to endometrial epithelium in implantation and in our previous research, we have proved estrogen can induce a fast increase in intracellular calcium of mouse blastocysts through acting on G protein-coupled receptor 30 (GPR30), which further promotes their implantation. Moreover, there has been evidence that cytoskeletal proteins are involved in integrin-mediated adhesion of many kinds of cells, which also plays an important role in implantation. To prove estrogen induces rapidly the changes of cytoskeletal proteins in mouse blastocysts and its roles in implantation, we first used immunofluorescence staining and laser confocal microscopy to investigate the fast effect of estrogen on the expression and localization of cytoskeletal proteins in mouse blastocysts. Second, we used electroporation associated with RNA interference to knock down one of the important cytoskeletal proteins, talin, in the mouse blastocyst cells to investigate the fast effect of estrogen on the localization of integrins and the binding activity of integrins with their ligand fibronectin (FN). At last, mouse blastocysts with different treatments were cultured with FN or uterine epithelial cell line Ishikawa in vitro, respectively, and transferred into the bilateral uterine horns of recipient mice, to study the role of the fast effect of estrogen on cytoskeletal proteins in blastocysts adhesion and implantation. Our results indicated that estradiol (E2), E2 conjugated with bovine serum album (E2-BSA) and G-1 (a GPR30-specific agonist) could induce cytoskeletal protein talin, vinculin, and actin to cluster in the mouse blastocysts, while G15 (a GPR30-specific antagonist) and BAPTA (a calcium chelator) may block this effect induced by E2-BSA. Furthermore, E2-BSA could induce the clustering and relocalization of integrin β1 and β3 and increase the FN-binding activity of integrins in blastocyst cells, while E2-BSA could not induce these effects in the blastocysts pretreated with talin-small interfering RNA (siRNA). Meanwhile, the adhesion rate and implantation rate of blastocysts pretreated with talin-siRNA were significantly lower than those pretreated with control-siRNA. We provided the first evidence that the fast effect of estrogen might cause the clustering of the cytoskeletal proteins in mouse blastocyst cells and further induce the changes of localization and functional activity of integrins in the blastocyst cells, which play important roles in blastocyst implantation.
Collapse
Affiliation(s)
- Shi-Mao Zhang
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- 2 Chengdu Women and Children's Central Hospital, Chengdu, Sichuan, China
| | - Lin-Lin Yu
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- 2 Chengdu Women and Children's Central Hospital, Chengdu, Sichuan, China
| | - Ting Qu
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Ying Hu
- 3 Department of Obstetrics &Gynecology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, China
| | - Dong-Zhi Yuan
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Sheng Zhang
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qian Xu
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - You-Bo Zhao
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jin-Hu Zhang
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Li-Min Yue
- 1 Department of Physiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
86
|
Huang Q, Xi G, Alamdar A, Zhang J, Shen H. Comparative proteomic analysis reveals heart toxicity induced by chronic arsenic exposure in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 229:210-218. [PMID: 28599205 DOI: 10.1016/j.envpol.2017.05.077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/12/2017] [Accepted: 05/27/2017] [Indexed: 06/07/2023]
Abstract
Arsenic is a widespread metalloid in the environment, which poses a broad spectrum of adverse effects on human health. However, a global view of arsenic-induced heart toxicity is still lacking, and the underlying molecular mechanisms remain unclear. By performing a comparative quantitative proteomic analysis, the present study aims to investigate the alterations of proteome profile in rat heart after long-term exposure to arsenic. As a result, we found that the abundance of 81 proteins were significantly altered by arsenic treatment (35 up-regulated and 46 down-regulated). Among these, 33 proteins were specifically associated with cardiovascular system development and function, including heart development, heart morphology, cardiac contraction and dilation, and other cardiovascular functions. It is further proposed that the aberrant regulation of 14 proteins induced by arsenic would disturb cardiac contraction and relaxation, impair heart morphogenesis and development, and induce thrombosis in rats, which is mediated by the Akt/p38 MAPK signaling pathway. Overall, these findings will augment our knowledge of the involved mechanisms and develop useful biomarkers for cardiotoxicity induced by environmental arsenic exposure.
Collapse
Affiliation(s)
- Qingyu Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Guochen Xi
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Ambreen Alamdar
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Jie Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Heqing Shen
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China.
| |
Collapse
|
87
|
Lacolley P, Regnault V, Segers P, Laurent S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol Rev 2017; 97:1555-1617. [DOI: 10.1152/physrev.00003.2017] [Citation(s) in RCA: 483] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
The cushioning function of large arteries encompasses distension during systole and recoil during diastole which transforms pulsatile flow into a steady flow in the microcirculation. Arterial stiffness, the inverse of distensibility, has been implicated in various etiologies of chronic common and monogenic cardiovascular diseases and is a major cause of morbidity and mortality globally. The first components that contribute to arterial stiffening are extracellular matrix (ECM) proteins that support the mechanical load, while the second important components are vascular smooth muscle cells (VSMCs), which not only regulate actomyosin interactions for contraction but mediate also mechanotransduction in cell-ECM homeostasis. Eventually, VSMC plasticity and signaling in both conductance and resistance arteries are highly relevant to the physiology of normal and early vascular aging. This review summarizes current concepts of central pressure and tensile pulsatile circumferential stress as key mechanical determinants of arterial wall remodeling, cell-ECM interactions depending mainly on the architecture of cytoskeletal proteins and focal adhesion, the large/small arteries cross-talk that gives rise to target organ damage, and inflammatory pathways leading to calcification or atherosclerosis. We further speculate on the contribution of cellular stiffness along the arterial tree to vascular wall stiffness. In addition, this review provides the latest advances in the identification of gene variants affecting arterial stiffening. Now that important hemodynamic and molecular mechanisms of arterial stiffness have been elucidated, and the complex interplay between ECM, cells, and sensors identified, further research should study their potential to halt or to reverse the development of arterial stiffness.
Collapse
Affiliation(s)
- Patrick Lacolley
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Véronique Regnault
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Patrick Segers
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Stéphane Laurent
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| |
Collapse
|
88
|
Zhang Q, Deng S, Sun K, Lin S, Lin Y, Zhu B, Cai X. MMP-2 and Notch signal pathway regulate migration of adipose-derived stem cells and chondrocytes in co-culture systems. Cell Prolif 2017; 50. [PMID: 28925018 DOI: 10.1111/cpr.12385] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 08/15/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES The crosstalk between chondrocytes and adipose-derived stem cells (ADSCs) could regulate the secretion of multiple growth factors. However, it is not clear how the paracrine action in co-culture systems affect cell migration. This study focused on the changes of cell migration of ADSCs and chondrocytes in co-culture conditions. MATERIALS AND METHODS Primary ADSCs and chondrocytes were isolated from Sprague-Dawley rat. Transwell co-culture systems, inoculated with ADSCs and chondrocytes, were established in vitro. The morphology of the cells was observed 7 days post-seeding by inverted phase-contrast microscope. Additionally, the cytoskeleton changes were investigated by immunofluorescence staining. To detect the abundance of Vinculin, we used immunofluorescence and Western blotting. Additionally, the expression level of MMP-2, Hey1 and Hes1 was examined to determine the mechanisms of co-culture-induced cell migration changes. RESULTS The migration of ADSCs and chondrocytes in co-culture conditions significantly decreased compared with that in mono-culture groups, accompanied by the decrease of filopodia and the expression level of MMP-2. CONCLUSIONS The overall study showed that the migration of ADSCs and chondrocytes differs significantly depending on culture conditions. Moreover, the Notch signalling pathway may be involved in this process. Accordingly, by studying changes in migration caused by co-culture, we obtained new insight into the crosstalk between ADSCs and chondrocytes.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shuwen Deng
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke Sun
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Bofeng Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Shanxi, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Shanxi, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
89
|
Huang DL, Bax NA, Buckley CD, Weis WI, Dunn AR. Vinculin forms a directionally asymmetric catch bond with F-actin. Science 2017; 357:703-706. [PMID: 28818948 PMCID: PMC5821505 DOI: 10.1126/science.aan2556] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/11/2017] [Indexed: 11/02/2022]
Abstract
Vinculin is an actin-binding protein thought to reinforce cell-cell and cell-matrix adhesions. However, how mechanical load affects the vinculin-F-actin bond is unclear. Using a single-molecule optical trap assay, we found that vinculin forms a force-dependent catch bond with F-actin through its tail domain, but with lifetimes that depend strongly on the direction of the applied force. Force toward the pointed (-) end of the actin filament resulted in a bond that was maximally stable at 8 piconewtons, with a mean lifetime (12 seconds) 10 times as long as the mean lifetime when force was applied toward the barbed (+) end. A computational model of lamellipodial actin dynamics suggests that the directionality of the vinculin-F-actin bond could establish long-range order in the actin cytoskeleton. The directional and force-stabilized binding of vinculin to F-actin may be a mechanism by which adhesion complexes maintain front-rear asymmetry in migrating cells.
Collapse
Affiliation(s)
- Derek L Huang
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Nicolas A Bax
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA
| | - Craig D Buckley
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - William I Weis
- Biophysics Program, Stanford University, Stanford, CA 94305, USA.
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA
| | - Alexander R Dunn
- Biophysics Program, Stanford University, Stanford, CA 94305, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
90
|
Han MKL, van der Krogt GNM, de Rooij J. Zygotic vinculin is not essential for embryonic development in zebrafish. PLoS One 2017; 12:e0182278. [PMID: 28767718 PMCID: PMC5540497 DOI: 10.1371/journal.pone.0182278] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
The formation of multicellular tissues during development is governed by mechanical forces that drive cell shape and tissue architecture. Protein complexes at sites of adhesion to the extracellular matrix (ECM) and cell neighbors, not only transmit these mechanical forces, but also allow cells to respond to changes in force by inducing biochemical feedback pathways. Such force-induced signaling processes are termed mechanotransduction. Vinculin is a central protein in mechanotransduction that in both integrin-mediated cell-ECM and cadherin-mediated cell-cell adhesions mediates force-induced cytoskeletal remodeling and adhesion strengthening. Vinculin was found to be important for the integrity and remodeling of epithelial tissues in cell culture models and could therefore be expected to be of broad importance in epithelial morphogenesis in vivo. Besides a function in mouse heart development, however, the importance of vinculin in morphogenesis of other vertebrate tissues has remained unclear. To investigate this further, we knocked out vinculin functioning in zebrafish, which contain two fully functional isoforms designated as vinculin A and vinculin B that both show high sequence conservation with higher vertebrates. Using TALEN and CRISPR-Cas gene editing technology we generated vinculin-deficient zebrafish. While single vinculin A mutants are viable and able to reproduce, additional loss of zygotic vinculin B was lethal after embryonic stages. Remarkably, vinculin-deficient embryos do not show major developmental defects, apart from mild pericardial edemas. These results lead to the conclusion that vinculin is not of broad importance for the development and morphogenesis of zebrafish tissues.
Collapse
Affiliation(s)
- Mitchell K. L. Han
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard N. M. van der Krogt
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johan de Rooij
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
91
|
Shin JY, Kim HN, Bhang SH, Yoon JK, Suh KY, Jeon NL, Kim BS. Topography-Guided Control of Local Migratory Behaviors and Protein Expression of Cancer Cells. Adv Healthc Mater 2017; 6. [PMID: 28509381 DOI: 10.1002/adhm.201700155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 04/16/2017] [Indexed: 12/19/2022]
Abstract
In vivo cancer cell migration and invasion are directed by biophysical guidance mechanisms such as pre-existing microtracks and basement membrane extracellular matrices. Here, this paper reports the correlation of the local migratory behavior of cancer cells and the biochemical signal expression using the topography that can guide or inhibit cell behaviors. To this end, the local apparent migration and the protein expression level are investigated with respect to the topographical feature size (flat, nanoline, and microline) and orientation (microline, microconcentric, and microradial) with the collectively migrating (A431) and individually migrating (MDA-MB-231 and U-87-MG) cancer cells. The results show that the migration and the protein expression of focal adhesion kinase, rho-associated protein kinase, and extracellular signal-regulated kinase are localized in the periphery of cell colony. Furthermore, the inhibition of migratory behavior at the periphery recues the protein expression, while the guidance of migration enhances the aforementioned protein expression. The results may imply the employ of biophysical inhibitory factors can help to control invasiveness of cancer cells during the progression state.
Collapse
Affiliation(s)
- Jung-Youn Shin
- School of Chemical and Biological Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Hong Nam Kim
- Center for BioMicrosystems; Brain Science Institute; Korea Institute of Science and Technology (KIST); Seoul 02792 Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Kahp-Yang Suh
- School of Mechanical and Aerospace Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering; Seoul National University; Seoul 08826 Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering; Seoul National University; Seoul 08826 Republic of Korea
- Bio-MAX Institute; Institute for Chemical Processes; Seoul National University; Seoul 08826 Republic of Korea
| |
Collapse
|
92
|
De Pascalis C, Etienne-Manneville S. Single and collective cell migration: the mechanics of adhesions. Mol Biol Cell 2017; 28:1833-1846. [PMID: 28684609 PMCID: PMC5541834 DOI: 10.1091/mbc.e17-03-0134] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 12/11/2022] Open
Abstract
Chemical and physical properties of the environment control cell proliferation, differentiation, or apoptosis in the long term. However, to be able to move and migrate through a complex three-dimensional environment, cells must quickly adapt in the short term to the physical properties of their surroundings. Interactions with the extracellular matrix (ECM) occur through focal adhesions or hemidesmosomes via the engagement of integrins with fibrillar ECM proteins. Cells also interact with their neighbors, and this involves various types of intercellular adhesive structures such as tight junctions, cadherin-based adherens junctions, and desmosomes. Mechanobiology studies have shown that cell-ECM and cell-cell adhesions participate in mechanosensing to transduce mechanical cues into biochemical signals and conversely are responsible for the transmission of intracellular forces to the extracellular environment. As they migrate, cells use these adhesive structures to probe their surroundings, adapt their mechanical properties, and exert the appropriate forces required for their movements. The focus of this review is to give an overview of recent developments showing the bidirectional relationship between the physical properties of the environment and the cell mechanical responses during single and collective cell migration.
Collapse
Affiliation(s)
- Chiara De Pascalis
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur Paris, CNRS UMR3691, 75724 Paris Cedex 15, France
- UPMC Université Paris 06, IFD, Sorbonne Universités, 75252 Paris Cedex 05, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur Paris, CNRS UMR3691, 75724 Paris Cedex 15, France
| |
Collapse
|
93
|
Preliminary study on plasma proteins in pregnant and non-pregnant female dogs. Theriogenology 2017; 97:1-8. [DOI: 10.1016/j.theriogenology.2017.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/04/2017] [Accepted: 04/04/2017] [Indexed: 11/22/2022]
|
94
|
Dwivedi M, Winter R. Binding of Vinculin to Lipid Membranes in Its Inhibited and Activated States. Biophys J 2017; 111:1444-1453. [PMID: 27705767 DOI: 10.1016/j.bpj.2016.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 10/20/2022] Open
Abstract
Phosphoinositols are an important class of phospholipids that are involved in a myriad of cellular processes, from cell signaling to motility and adhesion. Vinculin (Vn) is a major adaptor protein that regulates focal adhesions in conjunction with PIP2 in lipid membranes and other cytoskeletal components. The binding and unbinding transitions of Vn at the membrane interface are an important link to understanding the coordination of cell signaling and motility. Using different biophysical tools, including atomic force microscopy combined with confocal fluorescence microscopy and Fourier transform infrared spectroscopy, we studied the nanoscopic interactions of activated and autoinhibited states of Vn with lipid membranes. We hypothesize that a weak interaction occurs between Vn and lipid membranes, which leads to binding of autoinhibited Vn to supported lipid bilayers, and to unbinding in freestanding lipid vesicles. Likely driving forces may include tethering of the C-terminus to the lipid membrane, as well as hydrophobic helix-membrane interactions. Conversely, activated Vn binds strongly to membranes through specific interactions with clusters of PIP2 embedded in lipid membranes. Activated Vn harbored on PIP2 clusters may form small oligomeric interaction platforms for further interaction partners, which is necessary for the proper function of focal adhesion points.
Collapse
Affiliation(s)
- Mridula Dwivedi
- Physical Chemistry I, Biophysical Chemistry, Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany.
| | - Roland Winter
- Physical Chemistry I, Biophysical Chemistry, Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany.
| |
Collapse
|
95
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
96
|
Rahikainen R, von Essen M, Schaefer M, Qi L, Azizi L, Kelly C, Ihalainen TO, Wehrle-Haller B, Bastmeyer M, Huang C, Hytönen VP. Mechanical stability of talin rod controls cell migration and substrate sensing. Sci Rep 2017; 7:3571. [PMID: 28620171 PMCID: PMC5472591 DOI: 10.1038/s41598-017-03335-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/26/2017] [Indexed: 11/17/2022] Open
Abstract
Cells adhere to the surrounding tissue and probe its mechanical properties by forming cell-matrix adhesions. Talin is a critical adhesion protein and participates in the transmission of mechanical signals between extracellular matrix and cell cytoskeleton. Force induced unfolding of talin rod subdomains has been proposed to act as a cellular mechanosensor, but so far evidence linking their mechanical stability and cellular response has been lacking. Here, by utilizing computationally designed mutations, we demonstrate that stepwise destabilization of the talin rod R3 subdomain decreases cellular traction force generation, which affects talin and vinculin dynamics in cell-matrix adhesions and results in the formation of talin-rich but unstable adhesions. We observed a connection between talin stability and the rate of cell migration and also found that talin destabilization affects the usage of different integrin subtypes and sensing of extracellular matrix proteins. Experiments with truncated forms of talin confirm the mechanosensory role of the talin R3 subdomain and exclude the possibility that the observed effects are caused by the release of talin head-rod autoinhibition. In conclusion, this study provides evidence into how the controlled talin rod domain unfolding acts as a key regulator of adhesion structure and function and consequently controls central cellular processes such as cell migration and substrate sensing.
Collapse
Affiliation(s)
- Rolle Rahikainen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Magdaléna von Essen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Markus Schaefer
- Zoological Institute, Cell- and Neurobiology, Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces (IFG), Karlsruhe, Germany
| | - Lei Qi
- Markey Cancer Center and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Latifeh Azizi
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Conor Kelly
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | - Teemu O Ihalainen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland
| | | | - Martin Bastmeyer
- Zoological Institute, Cell- and Neurobiology, Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces (IFG), Karlsruhe, Germany
| | - Cai Huang
- Markey Cancer Center and Department of Molecular and Biomedical Pharmacology, University of Kentucky, Lexington, KY, USA
| | - Vesa P Hytönen
- Faculty of Medicine and Life Sciences and BioMediTech, University of Tampere, Finland and Fimlab Laboratories, Tampere, Finland.
| |
Collapse
|
97
|
Hu X, Margadant FM, Yao M, Sheetz MP. Molecular stretching modulates mechanosensing pathways. Protein Sci 2017; 26:1337-1351. [PMID: 28474792 DOI: 10.1002/pro.3188] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/21/2023]
Abstract
For individual cells in tissues to create the diverse forms of biological organisms, it is necessary that they must reliably sense and generate the correct forces over the correct distances and directions. There is considerable evidence that the mechanical aspects of the cellular microenvironment provide critical physical parameters to be sensed. How proteins sense forces and cellular geometry to create the correct morphology is not understood in detail but protein unfolding appears to be a major component in force and displacement sensing. Thus, the crystallographic structure of a protein domain provides only a starting point to then analyze what will be the effects of physiological forces through domain unfolding or catch-bond formation. In this review, we will discuss the recent studies of cytoskeletal and adhesion proteins that describe protein domain dynamics. Forces applied to proteins can activate or inhibit enzymes, increase or decrease protein-protein interactions, activate or inhibit protein substrates, induce catch bonds and regulate interactions with membranes or nucleic acids. Further, the dynamics of stretch-relaxation can average forces or movements to reliably regulate morphogenic movements. In the few cases where single molecule mechanics are studied under physiological conditions such as titin and talin, there are rapid cycles of stretch-relaxation that produce mechanosensing signals. Fortunately, the development of new single molecule and super-resolution imaging methods enable the analysis of single molecule mechanics in physiologically relevant conditions. Thus, we feel that stereotypical changes in cell and tissue shape involve mechanosensing that can be analyzed at the nanometer level to determine the molecular mechanisms involved.
Collapse
Affiliation(s)
- Xian Hu
- Mechanobiology Institute, National University of Singapore, Singapore, 117411.,Department of Biosciences, University of Oslo, Oslo, 0316, Norway
| | | | - Mingxi Yao
- Mechanobiology Institute, National University of Singapore, Singapore, 117411
| | - Michael Patrick Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, 117411.,Department of Biological Sciences, University of Columbia, New York, 10027
| |
Collapse
|
98
|
Ohashi K, Fujiwara S, Mizuno K. Roles of the cytoskeleton, cell adhesion and rho signalling in mechanosensing and mechanotransduction. J Biochem 2017; 161:245-254. [PMID: 28082721 DOI: 10.1093/jb/mvw082] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/13/2016] [Indexed: 11/13/2022] Open
Abstract
All cells sense and respond to various mechanical forces in and mechanical properties of their environment. To respond appropriately, cells must be able to sense the location, direction, strength and duration of these forces. Recent progress in mechanobiology has provided a better understanding of the mechanisms of mechanoresponses underlying many cellular and developmental processes. Various roles of mechanoresponses in development and tissue homeostasis have been elucidated, and many molecules involved in mechanotransduction have been identified. However, the whole picture of the functions and molecular mechanisms of mechanotransduction remains to be understood. Recently, novel mechanisms for sensing and transducing mechanical stresses via the cytoskeleton, cell-substrate and cell-cell adhesions and related proteins have been identified. In this review, we outline the roles of the cytoskeleton, cell-substrate and cell-cell adhesions, and related proteins in mechanosensing and mechanotransduction. We also describe the roles and regulation of Rho-family GTPases in mechanoresponses.
Collapse
Affiliation(s)
- Kazumasa Ohashi
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Sachiko Fujiwara
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan.,Department of Mechanical Science and Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka 560-8531, Japan Osaka
| | - Kensaku Mizuno
- Department of Biomolecular Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| |
Collapse
|
99
|
Yu T, Wang C, Yang J, Guo Y, Wu Y, Li X. Metformin inhibits SUV39H1-mediated migration of prostate cancer cells. Oncogenesis 2017; 6:e324. [PMID: 28459432 PMCID: PMC5523061 DOI: 10.1038/oncsis.2017.28] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/20/2017] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer (PCa) is a leading cause of cancer-related death among men, largely due to incurable distant metastases. Metformin, the most common used anti-type-2 diabetes medicine, has been linked to reduced cancer risk and better diagnosis. We found that metformin was able to inhibit PCa cell migration, which correlates with tumor metastatic capability. The pathogenesis and progression of tumors are closely related to dysregulated gene expression in tumor cells through epigenetic alterations such as DNA methylation and histone modifications. We found that the level of SUV39H1, a histone methyltransferase of H3 Lys9, was reduced in metformin-treated PCa cells in a time-dependent manner. SUV39H1 overexpression increased PCa migration, whereas SUV39H1 depletion suppressed PCa cell migration. There is a positive correlation between SUV39H1 expression and PCa pathological stages. We further showed that both metformin treatment and SUV39H1 knockout in PCa cells can reduce integrin αV and β1 proteins, as well as their downstream phosphorylated focal adhesion kinase (FAK) levels, which is essential for functional adhesion signaling and tumor cell migration. Taken together, metformin reduced SUV39H1 to inhibit migration of PCa cells via disturbing the integrin-FAK signaling. Our study suggests SUV39H1 as a novel target to inhibit PCa cell migration.
Collapse
Affiliation(s)
- T Yu
- Institute of Gene Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry (NYUCD), New York, NY, USA
| | - C Wang
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - J Yang
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry (NYUCD), New York, NY, USA
| | - Y Guo
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry (NYUCD), New York, NY, USA
| | - Y Wu
- Institute of Gene Engineered Animal Models for Human Diseases, Dalian Medical University, Dalian, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry (NYUCD), New York, NY, USA
- The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - X Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry (NYUCD), New York, NY, USA
- Department of Urology, New York University Langone Medical Center, New York, NY, USA
- Perlmutter Cancer Institute, New York University, Langone Medical Center, New York, NY, USA
| |
Collapse
|
100
|
Bays JL, DeMali KA. Vinculin in cell-cell and cell-matrix adhesions. Cell Mol Life Sci 2017; 74:2999-3009. [PMID: 28401269 PMCID: PMC5501900 DOI: 10.1007/s00018-017-2511-3] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
Abstract
Vinculin was identified as a component of focal adhesions and adherens junctions nearly 40 years ago. Since that time, remarkable progress has been made in understanding its activation, regulation and function. Here we discuss the current understanding of the roles of vinculin in cell–cell and cell–matrix adhesions. Emphasis is placed on the how vinculin is recruited, activated and regulated. We also highlight the recent understanding of how vinculin responds to and transmits force at integrin- and cadherin-containing adhesion complexes to the cytoskeleton. Furthermore, we discuss roles of vinculin in binding to and rearranging the actin cytoskeleton.
Collapse
Affiliation(s)
- Jennifer L Bays
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, USA
| | - Kris A DeMali
- Department of Biochemistry, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|