1
|
Park J, Na CS. Weighted single-step genome-wide association study to reveal new candidate genes for productive traits of Landrace pig in Korea. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:702-716. [PMID: 39165735 PMCID: PMC11331376 DOI: 10.5187/jast.2024.e104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/12/2023] [Accepted: 10/04/2023] [Indexed: 08/22/2024]
Abstract
The objective of this study was to identify genomic regions and candidate genes associated with productive traits using a total of 37,099 productive records and 6,683 single nucleotide polymorphism (SNP) data obtained from five Great-Grand-Parents (GGP) farms in Landrace. The estimated of heritabilities for days to 105 kg (AGE), average daily gain (ADG), backfat thickness (BF), and eye muscle area (EMA) were 0.49, 0.49, 0.56, and 0.23, respectively. We identified a genetic window that explained 2.05%-2.34% for each trait of the total genetic variance. We observed a clear partitioning of the four traits into two groups, and the most significant genomic region for AGE and ADG were located on the Sus scrofa chromosome (SSC) 1, while BF and EMA were located on SSC 2. We conducted Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), which revealed results in three biological processes, four cellular component, three molecular function, and six KEGG pathway. Significant SNPs can be used as markers for quantitative trait loci (QTL) investigation and genomic selection (GS) for productive traits in Landrace pig.
Collapse
Affiliation(s)
- Jun Park
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| | - Chong-Sam Na
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
2
|
Wu Z, Wu D, Zhong Q, Zou X, Liu Z, Long H, Wei J, Li X, Dai F. The role of zyxin in signal transduction and its relationship with diseases. Front Mol Biosci 2024; 11:1371549. [PMID: 38712343 PMCID: PMC11070705 DOI: 10.3389/fmolb.2024.1371549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
This review highlighted the pivotal role of zyxin, an essential cell focal adhesions protein, in cellular biology and various diseases. Zyxin can orchestrate the restructuring and dynamic alterations of the cellular cytoskeleton, which is involved in cell proliferation, adhesion, motility, and gene transcription. Aberrant zyxin expression is closely correlated with tumor cell activity and cardiac function in both tumorigenesis and cardiovascular diseases. Moreover, in fibrotic and inflammatory conditions, zyxin can modulate cellular functions and inflammatory responses. Therefore, a comprehensive understanding of zyxin is crucial for deciphering signal transduction networks and disease pathogenesis. Investigating its role in diseases holds promise for novel avenues in early diagnosis and therapeutic strategies. Nevertheless, targeting zyxin as a therapeutic focal point presents challenges in terms of specificity, safety, drug delivery, and resistance. Nonetheless, in-depth studies on zyxin and the application of precision medicine could offer new possibilities for personalized treatment modalities.
Collapse
Affiliation(s)
- Zelan Wu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Daiqin Wu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qin Zhong
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xue Zou
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhongjing Liu
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hehua Long
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Jing Wei
- Department of Endocrinology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xia Li
- Guizhou Precision Medicine Institute, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fangjie Dai
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
3
|
Kliewe F, Siegerist F, Hammer E, Al-Hasani J, Amling TRJ, Hollemann JZE, Schindler M, Drenic V, Simm S, Amann K, Daniel C, Lindenmeyer M, Hecker M, Völker U, Endlich N. Zyxin is important for the stability and function of podocytes, especially during mechanical stretch. Commun Biol 2024; 7:446. [PMID: 38605154 PMCID: PMC11009394 DOI: 10.1038/s42003-024-06125-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Podocyte detachment due to mechanical stress is a common issue in hypertension-induced kidney disease. This study highlights the role of zyxin for podocyte stability and function. We have found that zyxin is significantly up-regulated in podocytes after mechanical stretch and relocalizes from focal adhesions to actin filaments. In zyxin knockout podocytes, we found that the loss of zyxin reduced the expression of vinculin and VASP as well as the expression of matrix proteins, such as fibronectin. This suggests that zyxin is a central player in the translation of mechanical forces in podocytes. In vivo, zyxin is highly up-regulated in patients suffering from diabetic nephropathy and in hypertensive DOCA-salt treated mice. Furthermore, zyxin loss in mice resulted in proteinuria and effacement of podocyte foot processes that was measured by super resolution microscopy. This highlights the essential role of zyxin for podocyte maintenance in vitro and in vivo, especially under mechanical stretch.
Collapse
Affiliation(s)
- Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jaafar Al-Hasani
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | | | | | - Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Vedran Drenic
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Kerstin Amann
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| |
Collapse
|
4
|
Schmitt MS, Colen J, Sala S, Devany J, Seetharaman S, Caillier A, Gardel ML, Oakes PW, Vitelli V. Machine learning interpretable models of cell mechanics from protein images. Cell 2024; 187:481-494.e24. [PMID: 38194965 PMCID: PMC11225795 DOI: 10.1016/j.cell.2023.11.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/20/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Cellular form and function emerge from complex mechanochemical systems within the cytoplasm. Currently, no systematic strategy exists to infer large-scale physical properties of a cell from its molecular components. This is an obstacle to understanding processes such as cell adhesion and migration. Here, we develop a data-driven modeling pipeline to learn the mechanical behavior of adherent cells. We first train neural networks to predict cellular forces from images of cytoskeletal proteins. Strikingly, experimental images of a single focal adhesion (FA) protein, such as zyxin, are sufficient to predict forces and can generalize to unseen biological regimes. Using this observation, we develop two approaches-one constrained by physics and the other agnostic-to construct data-driven continuum models of cellular forces. Both reveal how cellular forces are encoded by two distinct length scales. Beyond adherent cell mechanics, our work serves as a case study for integrating neural networks into predictive models for cell biology.
Collapse
Affiliation(s)
- Matthew S Schmitt
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA; Department of Physics, University of Chicago, Chicago, IL 60637, USA; Kadanoff Center for Theoretical Physics, University of Chicago, Chicago, IL 60637, USA
| | - Jonathan Colen
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA; Department of Physics, University of Chicago, Chicago, IL 60637, USA; Kadanoff Center for Theoretical Physics, University of Chicago, Chicago, IL 60637, USA
| | - Stefano Sala
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - John Devany
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA; Department of Physics, University of Chicago, Chicago, IL 60637, USA
| | - Shailaja Seetharaman
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA; Department of Physics, University of Chicago, Chicago, IL 60637, USA
| | - Alexia Caillier
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Margaret L Gardel
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA; Department of Physics, University of Chicago, Chicago, IL 60637, USA.
| | - Patrick W Oakes
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA.
| | - Vincenzo Vitelli
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA; Department of Physics, University of Chicago, Chicago, IL 60637, USA; Kadanoff Center for Theoretical Physics, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
5
|
McGowan SE. Discoidin domain receptor-2 enhances secondary alveolar septation in mice by activating integrins and modifying focal adhesions. Am J Physiol Lung Cell Mol Physiol 2023; 324:L307-L324. [PMID: 36719983 PMCID: PMC9988528 DOI: 10.1152/ajplung.00169.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/20/2022] [Accepted: 01/09/2023] [Indexed: 02/02/2023] Open
Abstract
The extracellular matrix (ECM) of the pulmonary parenchyma must maintain the structural relationships among resident cells during the constant distortion imposed by respiration. This dictates that both the ECM and cells adapt to changes in shape, while retaining their attachment. Membrane-associated integrins and discoidin domain receptors (DDR) bind collagen and transmit signals to the cellular cytoskeleton. Although the contributions of DDR2 to collagen deposition and remodeling during osseous development are evident, it is unclear how DDR2 contributes to lung development. Using mice (smallie, Slie/Slie, DDR2Δ) bearing a spontaneous inactivating deletion within the DDR2 coding region, we observed a decrease in gas-exchange surface area and enlargement of alveolar ducts. Compared with fibroblasts isolated from littermate controls, DDR2Δ fibroblasts, spread more slowly, developed fewer lamellipodia, and were less responsive to the rigidity of neighboring collagen fibers. Activated β1-integrin (CD29) was reduced in focal adhesions (FA) of DDR2Δ fibroblasts, less phospho-zyxin localized to and fewer FA developed over ventral actin stress fibers, and the adhesions had a lower aspect ratio compared with controls. However, DDR2 deletion did not reduce cellular displacement of the ECM. Our findings indicate that DDR2, in concert with collagen-binding β1-integrins, regulates the timing and location of focal adhesion formation and how lung fibroblasts respond to ECM rigidity. Reduced rigidity sensing and mechano-responsiveness may contribute to the distortion of alveolar ducts, where the fiber cable-network is enriched and tensile forces are concentrated. Strategies targeting DDR2 could help guide fibroblasts to locations where tensile forces organize parenchymal repair.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| |
Collapse
|
6
|
Zhang S, Chong LH, Woon JYX, Chua TX, Cheruba E, Yip AK, Li HY, Chiam KH, Koh CG. Zyxin regulates embryonic stem cell fate by modulating mechanical and biochemical signaling interface. Commun Biol 2023; 6:62. [PMID: 36653484 PMCID: PMC9849324 DOI: 10.1038/s42003-023-04421-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Biochemical signaling and mechano-transduction are both critical in regulating stem cell fate. How crosstalk between mechanical and biochemical cues influences embryonic development, however, is not extensively investigated. Using a comparative study of focal adhesion constituents between mouse embryonic stem cell (mESC) and their differentiated counterparts, we find while zyxin is lowly expressed in mESCs, its levels increase dramatically during early differentiation. Interestingly, overexpression of zyxin in mESCs suppresses Oct4 and Nanog. Using an integrative biochemical and biophysical approach, we demonstrate involvement of zyxin in regulating pluripotency through actin stress fibres and focal adhesions which are known to modulate cellular traction stress and facilitate substrate rigidity-sensing. YAP signaling is identified as an important biochemical effector of zyxin-induced mechanotransduction. These results provide insights into the role of zyxin in the integration of mechanical and biochemical cues for the regulation of embryonic stem cell fate.
Collapse
Affiliation(s)
- Songjing Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lor Huai Chong
- Bioinformatics Institute A*STAR, Singapore, Singapore.,School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jessie Yong Xing Woon
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Theng Xuan Chua
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Ai Kia Yip
- Bioinformatics Institute A*STAR, Singapore, Singapore
| | - Hoi-Yeung Li
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
7
|
Hakeem RM, Subramanian BC, Hockenberry MA, King ZT, Butler MT, Legant WR, Bear JE. A Photopolymerized Hydrogel System with Dual Stiffness Gradients Reveals Distinct Actomyosin-Based Mechano-Responses in Fibroblast Durotaxis. ACS NANO 2023; 17:197-211. [PMID: 36475639 PMCID: PMC9839609 DOI: 10.1021/acsnano.2c05941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Durotaxis, migration of cells directed by a stiffness gradient, is critical in development and disease. To distinguish durotaxis-specific migration mechanisms from those on uniform substrate stiffnesses, we engineered an all-in-one photopolymerized hydrogel system containing areas of stiffness gradients with dual slopes (steep and shallow), adjacent to uniform stiffness (soft and stiff) regions. While fibroblasts rely on nonmuscle myosin II (NMII) activity and the LIM-domain protein Zyxin, ROCK and the Arp2/3 complex are surprisingly dispensable for durotaxis on either stiffness gradient. Additionally, loss of either actin-elongator Formin-like 3 (FMNL3) or actin-bundler fascin has little impact on durotactic response on stiffness gradients. However, lack of Arp2/3 activity results in a filopodia-based durotactic migration that is equally as efficient as that of lamellipodia-based durotactic migration. Importantly, we uncover essential and specific roles for FMNL3 and fascin in the formation and asymmetric distribution of filopodia during filopodia-based durotaxis response to the stiffness gradients. Together, our tunable all-in-one hydrogel system serves to identify both conserved as well as distinct molecular mechanisms that underlie mechano-responses of cells experiencing altered slopes of stiffness gradients.
Collapse
Affiliation(s)
- Reem M Hakeem
- Department of Biochemistry and Biophysics, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Bhagawat C Subramanian
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Max A Hockenberry
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Zayna T King
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Wesley R Legant
- Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - James E Bear
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
8
|
Martynova NY, Parshina EA, Zaraisky AG. Cytoskeletal protein Zyxin in embryonic development: from controlling cell movements and pluripotency to regulating embryonic patterning. FEBS J 2023; 290:66-72. [PMID: 34854244 DOI: 10.1111/febs.16308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/29/2021] [Accepted: 11/30/2021] [Indexed: 01/14/2023]
Abstract
The Lim-domain protein Zyxin was initially identified as a minor actin cytoskeleton protein that regulates the assembly and repair of actin filaments. At the same time, additional functions revealed for Zyxin in recent decades indicate that this protein can also play an important role in regulating gene expression and cell differentiation. In this review, we analysed the data in the literature pointing to Zyxin as one of the possible molecular hubs linking morphogenetic cell movements with gene expression, stem cell status regulation and pattern formation during the most complex processes in organism life, embryogenesis.
Collapse
Affiliation(s)
- Natalia Y Martynova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Elena A Parshina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Andrey G Zaraisky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
9
|
Seetharaman S, Sala S, Gardel ML, Oakes PW. Quantifying Strain-Sensing Protein Recruitment During Stress Fiber Repair. Methods Mol Biol 2023; 2600:169-182. [PMID: 36587097 DOI: 10.1007/978-1-0716-2851-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A family of proteins have been identified that recognize damaged, strained actin filaments in stress fibers. These proteins are often referred to as strain- or force-sensing and trigger downstream signaling mechanisms that can facilitate repair at these strain sites. Here we describe a method using high-resolution microscopy to screen and quantify the mechanosensitive recruitment of proteins to these stress fiber strain sites. Strain sites are induced using spatially controlled illumination of UV light to locally damage actin stress fibers. Recruitment of potential strain-sensing proteins can then either be compared to (Blanchoin, Physiol Rev 94, 235-263, 2014) a known control (e.g., zyxin-GFP) or (Hoffman, Mol Biol Cell 23, 1846-1859, 2012) the pre-damaged stress fiber protein distribution. With this method, strain-sensing proteins and their dynamic association with stress fiber strain sites can be reproducibly measured and compared.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Department of Physics, Institute for Biophysical Dynamics, and James Franck Institute, University of Chicago, Chicago, IL, USA
| | - Stefano Sala
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Margaret L Gardel
- Department of Physics, Institute for Biophysical Dynamics, and James Franck Institute, University of Chicago, Chicago, IL, USA.
| | - Patrick W Oakes
- Department of Cell & Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
10
|
Hoffman LM, Jensen CC, Beckerle MC. Phosphorylation of the small heat shock protein HspB1 regulates cytoskeletal recruitment and cell motility. Mol Biol Cell 2022; 33:ar100. [PMID: 35767320 DOI: 10.1091/mbc.e22-02-0057] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The small heat shock protein HspB1, also known as Hsp25/27, is a ubiquitously expressed molecular chaperone that responds to mechanical cues. Uniaxial cyclic stretch activates the p38 mitogen-activated protein kinase (MAPK) signaling cascade and increases the phosphorylation of HspB1. Similar to the mechanosensitive cytoskeletal regulator zyxin, phospho-HspB1 is recruited to features of the stretch-stimulated actin cytoskeleton. To evaluate the role of HspB1 and its phosphoregulation in modulating cell function, we utilized CRISPR/Cas9-edited HspB1-null cells and determined they were altered in behaviors such as actin cytoskeletal remodeling, cell spreading, and cell motility. In our model system, expression of WT HspB1, but not nonphosphorylatable HspB1, rescued certain characteristics of the HspB1-null cells including the enhanced cell motility of HspB1-null cells and the deficient actin reinforcement of stretch-stimulated HspB1-null cells. The recruitment of HspB1 to high-tension structures in geometrically constrained cells, such as actin comet tails emanating from focal adhesions, also required a phosphorylatable HspB1. We show that mechanical signals activate posttranslational regulation of the molecular chaperone, HspB1, and are required for normal cell behaviors including actin cytoskeletal remodeling, cell spreading, and cell migration.
Collapse
Affiliation(s)
- Laura M Hoffman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Biology, University of Utah, Salt Lake City, UT 84112
| | | | - Mary C Beckerle
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Biology, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
11
|
Madhavan SM, Konieczkowski M, Bruggeman LA, DeWalt M, Nguyen JK, O'Toole JF, Sedor JR. Essential role of Wtip in mouse development and maintenance of the glomerular filtration barrier. Am J Physiol Renal Physiol 2022; 323:F272-F287. [PMID: 35862649 PMCID: PMC9394782 DOI: 10.1152/ajprenal.00051.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Wilms' tumor interacting protein (Wtip) has been implicated in cell junction assembly and cell differentiation and interacts with proteins in the podocyte slit diaphragm, where it regulates podocyte phenotype. To define Wtip expression and function in the kidney, we created a Wtip-deleted mouse model using β-galactosidase-neomycin (β-geo) gene trap technology. Wtip gene trap mice were embryonic lethal, suggesting additional developmental roles outside kidney function. Using β-geo heterozygous and normal mice, Wtip expression was identified in the developing kidneys, heart, and eyes. In the kidney, expression was restricted to podocytes, which appeared initially at the capillary loop stage coinciding with terminal podocyte differentiation. Heterozygous mice had an expected lifespan and showed no evidence of proteinuria or glomerular pathology. However, heterozygous mice were more susceptible to glomerular injury than wild-type littermates and developed more significant and prolonged proteinuria in response to lipopolysaccharide or adriamycin. In normal human kidneys, WTIP expression patterns were consistent with observations in mice and were lost in glomeruli concurrent with loss of synaptopodin expression in disease. Mechanistically, we identified the Rho guanine nucleotide exchange factor 12 (ARHGEF12) as a binding partner for WTIP. ARHGEF12 was expressed in human podocytes and formed high-affinity interactions through their LIM- and PDZ-binding domains. Our findings suggest that Wtip is essential for early murine embryonic development and maintaining normal glomerular filtration barrier function, potentially regulating slit diaphragm and foot process function through Rho effector proteins.NEW & NOTEWORTHY This study characterized dynamic expression patterns of Wilms' tumor interacting protein (Wtip) and demonstrates the novel role of Wtip in murine development and maintenance of the glomerular filtration barrier.
Collapse
Affiliation(s)
- Sethu M Madhavan
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Leslie A Bruggeman
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Nephrology, Cleveland Clinic, Cleveland, Ohio
| | - Megan DeWalt
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Jane K Nguyen
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio
| | - John F O'Toole
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Nephrology, Cleveland Clinic, Cleveland, Ohio
| | - John R Sedor
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio
- Department of Nephrology, Cleveland Clinic, Cleveland, Ohio
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
12
|
Legerstee K, Houtsmuller AB. A Layered View on Focal Adhesions. BIOLOGY 2021; 10:biology10111189. [PMID: 34827182 PMCID: PMC8614905 DOI: 10.3390/biology10111189] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary The cytoskeleton is a network of protein fibres within cells that provide structure and support intracellular transport. Focal adhesions are protein complexes associated with the outer cell membrane that are found at the ends of specialised actin fibres of this cytoskeleton. They mediate cell adhesion by connecting the cytoskeleton to the extracellular matrix, a protein and sugar network that surrounds cells in tissues. Focal adhesions also translate forces on actin fibres into forces contributing to cell migration. Cell adhesion and migration are crucial to diverse biological processes such as embryonic development, proper functioning of the immune system or the metastasis of cancer cells. Advances in fluorescence microscopy and data analysis methods provided a more detailed understanding of the dynamic ways in which proteins bind and dissociate from focal adhesions and how they are organised within these protein complexes. In this review, we provide an overview of the advances in the current scientific understanding of focal adhesions and summarize relevant imaging techniques. One of the key insights is that focal adhesion proteins are organised into three layers parallel to the cell membrane. We discuss the relevance of this layered nature for the functioning of focal adhesion. Abstract The cytoskeleton provides structure to cells and supports intracellular transport. Actin fibres are crucial to both functions. Focal Adhesions (FAs) are large macromolecular multiprotein assemblies at the ends of specialised actin fibres linking these to the extracellular matrix. FAs translate forces on actin fibres into forces contributing to cell migration. This review will discuss recent insights into FA protein dynamics and their organisation within FAs, made possible by advances in fluorescence imaging techniques and data analysis methods. Over the last decade, evidence has accumulated that FAs are composed of three layers parallel to the plasma membrane. We focus on some of the most frequently investigated proteins, two from each layer, paxillin and FAK (bottom, integrin signalling layer), vinculin and talin (middle, force transduction layer) and zyxin and VASP (top, actin regulatory layer). Finally, we discuss the potential impact of this layered nature on different aspects of FA behaviour.
Collapse
|
13
|
Essential role of zyxin in platelet biogenesis and glycoprotein Ib-IX surface expression. Cell Death Dis 2021; 12:955. [PMID: 34657146 PMCID: PMC8520529 DOI: 10.1038/s41419-021-04246-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
Platelets are generated from the cytoplasm of megakaryocytes (MKs) via actin cytoskeleton reorganization. Zyxin is a focal adhesion protein and wildly expressed in eukaryotes to regulate actin remodeling. Zyxin is upregulated during megakaryocytic differentiation; however, the role of zyxin in thrombopoiesis is unknown. Here we show that zyxin ablation results in profound macrothrombocytopenia. Platelet lifespan and thrombopoietin level were comparable between wild-type and zyxin-deficient mice, but MK maturation, demarcation membrane system formation, and proplatelet generation were obviously impaired in the absence of zyxin. Differential proteomic analysis of proteins associated with macrothrombocytopenia revealed that glycoprotein (GP) Ib-IX was significantly reduced in zyxin-deficient platelets. Moreover, GPIb-IX surface level was decreased in zyxin-deficient MKs. Knockdown of zyxin in a human megakaryocytic cell line resulted in GPIbα degradation by lysosomes leading to the reduction of GPIb-IX surface level. We further found that zyxin was colocalized with vasodilator-stimulated phosphoprotein (VASP), and loss of zyxin caused diffuse distribution of VASP and actin cytoskeleton disorganization in both platelets and MKs. Reconstitution of zyxin with VASP binding site in zyxin-deficient hematopoietic progenitor cell-derived MKs restored GPIb-IX surface expression and proplatelet generation. Taken together, our findings identify zyxin as a regulator of platelet biogenesis and GPIb-IX surface expression through VASP-mediated cytoskeleton reorganization, suggesting possible pathogenesis of macrothrombocytopenia.
Collapse
|
14
|
Abstract
TRIP6, a member of the ZYXIN-family of LIM domain proteins, is a focal adhesion component. Trip6 deletion in the mouse, reported here, reveals a function in the brain: ependymal and choroid plexus epithelial cells are carrying, unexpectedly, fewer and shorter cilia, are poorly differentiated, and the mice develop hydrocephalus. TRIP6 carries numerous protein interaction domains and its functions require homodimerization. Indeed, TRIP6 disruption in vitro (in a choroid plexus epithelial cell line), via RNAi or inhibition of its homodimerization, confirms its function in ciliogenesis. Using super-resolution microscopy, we demonstrate TRIP6 localization at the pericentriolar material and along the ciliary axoneme. The requirement for homodimerization which doubles its interaction sites, its punctate localization along the axoneme, and its co-localization with other cilia components suggest a scaffold/co-transporter function for TRIP6 in cilia. Thus, this work uncovers an essential role of a LIM-domain protein assembly factor in mammalian ciliogenesis.
Collapse
|
15
|
Norizadeh Abbariki T, Gonda Z, Kemler D, Urbanek P, Wagner T, Litfin M, Wang ZQ, Herrlich P, Kassel O. The LIM domain protein nTRIP6 modulates the dynamics of myogenic differentiation. Sci Rep 2021; 11:12904. [PMID: 34145356 PMCID: PMC8213751 DOI: 10.1038/s41598-021-92331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 06/02/2021] [Indexed: 11/11/2022] Open
Abstract
The process of myogenesis which operates during skeletal muscle regeneration involves the activation of muscle stem cells, the so-called satellite cells. These then give rise to proliferating progenitors, the myoblasts which subsequently exit the cell cycle and differentiate into committed precursors, the myocytes. Ultimately, the fusion of myocytes leads to myofiber formation. Here we reveal a role for the transcriptional co-regulator nTRIP6, the nuclear isoform of the LIM-domain protein TRIP6, in the temporal control of myogenesis. In an in vitro model of myogenesis, the expression of nTRIP6 is transiently up-regulated at the transition between proliferation and differentiation, whereas that of the cytosolic isoform TRIP6 is not altered. Selectively blocking nTRIP6 function results in accelerated early differentiation followed by deregulated late differentiation and fusion. Thus, the transient increase in nTRIP6 expression appears to prevent premature differentiation. Accordingly, knocking out the Trip6 gene in satellite cells leads to deregulated skeletal muscle regeneration dynamics in the mouse. Thus, dynamic changes in nTRIP6 expression contributes to the temporal control of myogenesis.
Collapse
Affiliation(s)
- Tannaz Norizadeh Abbariki
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Zita Gonda
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Denise Kemler
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Pavel Urbanek
- Leibniz Institute for Age Research (Fritz Lipmann Institute, FLI), Jena, Germany
| | - Tabea Wagner
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Margarethe Litfin
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Zhao-Qi Wang
- Leibniz Institute for Age Research (Fritz Lipmann Institute, FLI), Jena, Germany
| | - Peter Herrlich
- Leibniz Institute for Age Research (Fritz Lipmann Institute, FLI), Jena, Germany
| | - Olivier Kassel
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
16
|
Konno K, Kulkeaw K, Sasada M, Nii T, Kaneyuki A, Ishitani T, Arai F, Sugiyama D. A novel method to purify neutrophils enables functional analysis of zebrafish hematopoiesis. Genes Cells 2020; 25:770-781. [PMID: 33006802 DOI: 10.1111/gtc.12810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/26/2020] [Accepted: 09/24/2020] [Indexed: 01/12/2023]
Abstract
Zebrafish is a useful model to study vertebrate hematopoiesis, but lack of antibodies to zebrafish proteins has limited purification of hematopoietic cells. Here, we purified neutrophils from larval and adult zebrafish using the lectin Phaseolus vulgaris erythroagglutinin (PHA-E) and DRAQ5, a DNA-staining fluorescent dye. In adult kidney marrow, we purified neutrophil-like PHA-E4low DRAQ5low cells, which neutrophil-type granules. Specifically, at 96-hr post-fertilization, we sorted large-sized cells from larvae using forward scatter and found that they consisted of PHA-Elow DRAQ5low populations. These cells had myeloperoxidase activity, were Sudan Black B-positive and expressed high levels of neutrophil-specific (csf3r and mpx) mRNAs, all neutrophil characteristics. Using this method, we conducted functional analysis suggesting that zyxin (Zyx) plays a role in neutrophil generation in zebrafish larvae. Overall, PHA-E and DRAQ5-based flow cytometry serves as a tool to purify zebrafish neutrophils.
Collapse
Affiliation(s)
- Katsuhiro Konno
- Incubation Center for Advanced Medical Science, Kyushu University, Fukuoka, Japan.,Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kasem Kulkeaw
- Department of Research and Development of Next Generation Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Manabu Sasada
- Clinical Research Center in Hiroshima University Hospital, Hiroshima, Japan
| | - Takenobu Nii
- Incubation Center for Advanced Medical Science, Kyushu University, Fukuoka, Japan
| | - Ayako Kaneyuki
- Incubation Center for Advanced Medical Science, Kyushu University, Fukuoka, Japan
| | - Tohru Ishitani
- Division of Cellular and Molecular Biology, Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.,Lab of Integrated Signaling Systems, Department of Molecular Medicine, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
| | - Fumio Arai
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Sugiyama
- Incubation Center for Advanced Medical Science, Kyushu University, Fukuoka, Japan.,Department of Clinical Research, Hiroshima-Nishi Medical Center, Hiroshima, Japan.,Translational Research Center in Hiroshima University, Hiroshima, Japan
| |
Collapse
|
17
|
Dynamics and distribution of paxillin, vinculin, zyxin and VASP depend on focal adhesion location and orientation. Sci Rep 2019; 9:10460. [PMID: 31320676 PMCID: PMC6639384 DOI: 10.1038/s41598-019-46905-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/05/2019] [Indexed: 01/07/2023] Open
Abstract
Focal adhesions (FAs) are multiprotein structures that link the intracellular cytoskeleton to the extracellular matrix. They mediate cell adhesion and migration, crucial to many (patho-) physiological processes. We examined in two cell types from different species the binding dynamics of functionally related FA protein pairs: paxillin and vinculin versus zyxin and VASP. In photobleaching experiments ~40% of paxillin and vinculin remained stably associated with a FA for over half an hour. Zyxin and VASP predominantly displayed more transient interactions. We show protein binding dynamics are influenced by FA location and orientation. In FAs located close to the edge of the adherent membrane paxillin, zyxin and VASP were more dynamic and had larger bound fractions. Zyxin and VASP were also more dynamic and had larger bound fractions at FAs perpendicular compared to parallel to this edge. Finally, we developed a photoconversion assay to specifically visualise stably bound proteins within subcellular structures and organelles. This revealed that while paxillin and vinculin are distributed evenly throughout FAs, their stably bound fractions form small clusters within the FA-complex. These clusters are more concentrated for paxillin than for vinculin and are mostly found at the proximal half of the FA where actin also enters.
Collapse
|
18
|
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: 36] [Impact Index Per Article: 4.5] [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
|
19
|
Kouwaki T, Okamoto M, Tsukamoto H, Fukushima Y, Matsumoto M, Seya T, Oshiumi H. Zyxin stabilizes RIG-I and MAVS interactions and promotes type I interferon response. Sci Rep 2017; 7:11905. [PMID: 28928438 PMCID: PMC5605516 DOI: 10.1038/s41598-017-12224-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/06/2017] [Indexed: 02/08/2023] Open
Abstract
RIG-I and MDA5 are cytoplasmic viral RNA sensors that belong to the RIG-I-like receptors (RLRs), which induce antiviral innate immune responses, including the production of type I interferon and other pro-inflammatory cytokines. After recognition of viral RNA, the N-terminal caspase activation and recruitment domains (CARDs) of RIG-I and MDA5 bind to a CARD in the MAVS adaptor molecule, resulting in MAVS oligomerization and downstream signaling. To reveal the molecular mechanism of MAVS-dependent signaling, we performed a yeast two-hybrid screening and identified zyxin as a protein that binds to MAVS. Zyxin co-immunoprecipitated with MAVS in human cells. A proximity ligation assay showed that zyxin and MAVS partly co-localized on mitochondria. Ectopic expression of zyxin augmented MAVS-mediated IFN-β promoter activation, and knockdown of zyxin (ZYX) attenuated the IFN-β promoter activation. Moreover, ZYX knockdown reduced the expression of type I IFN and an interferon-inducible gene after stimulation with polyI:C or influenza A virus RNA. Interestingly, physical interactions between RLRs and MAVS were abrogated by ZYX knockdown. These observations indicate that zyxin serves as a scaffold for the interactions between RLRs and MAVS.
Collapse
Affiliation(s)
- Takahisa Kouwaki
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Masaaki Okamoto
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Yoshimi Fukushima
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan
| | - Misako Matsumoto
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Kita-Ku, Sapporo, 060-8556, Japan
| | - Tsukasa Seya
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Kita-Ku, Sapporo, 060-8556, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1, Honjo, Chuo-ku, Kumamoto, 860-8556, Japan.
- JST, PRESTO, 1-1-1- Honjo, Chuo-ku, Kumamoto, 060-8556, Japan.
| |
Collapse
|
20
|
Wang J, Li J, Yang J, Zhang L, Gao S, Jiao F, Yi M, Xu J. MicroRNA‑138‑5p regulates neural stem cell proliferation and differentiation in vitro by targeting TRIP6 expression. Mol Med Rep 2017; 16:7261-7266. [PMID: 28944841 PMCID: PMC5865854 DOI: 10.3892/mmr.2017.7504] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Research on neural stem cells (NSCs) has recently focused on microRNAs (miRNAs), a class of small non-coding RNAs that have crucial roles in regulating NSC proliferation and differentiation. In the present study, a quantitative-polymerase chain reaction assay revealed that the expression of miRNA (miR)-138-5p was significantly decreased during neural differentiation of NSCs in vitro. Overexpression of miR-138-5p reduced NSC proliferation and increased NSC differentiation. Furthermore, suppression of miR-138-5p via transfection with a miRNA inhibitor enhanced NSC proliferation and attenuated NSC differentiation. Additionally, expression of thyroid hormone receptor interacting protein 6 (TRIP6), a critical regulator of NSCs, was negatively correlated with the miR-138-5p level. A luciferase assay demonstrated that miR-138-5p regulate TRIP6 by directly binding the 3′-untranslated region of the mRNA. Additionally, upregulation of TRIP6 rescued the NSC proliferation deficiency induced by miR-138-5p and abolished miR-138-5p-promoted NSCs differentiation. By contrast, downregulation of TRIP6 produced the opposite effect on proliferation and differentiation of NSCs transfected with anti-miR-138-5p. Taken together, the data suggest that miR-138-5p regulates NSCs proliferation and differentiation, and may be useful in developing novel treatments for neurological disorders via manipulation of miR-138-5p in NSCs.
Collapse
Affiliation(s)
- Juan Wang
- Stem Cell Center, East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Jixia Li
- Clinical Laboratory, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Jian Yang
- Administration Office, Yantai Blood Station, Yantai, Shandong 264000, P.R. China
| | - Lianguo Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| | - Shane Gao
- Stem Cell Center, East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Fei Jiao
- Department of Biotechnology and Molecular, Binzhou Medical College, Yantai, Shandong 264003, P.R. China
| | - Maoli Yi
- Laboratory of Yantai Yuhuangding Hospital, Yantai, Shandong 264003, P.R. China
| | - Jun Xu
- Stem Cell Center, East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
21
|
Hoffman L, Jensen CC, Yoshigi M, Beckerle M. Mechanical signals activate p38 MAPK pathway-dependent reinforcement of actin via mechanosensitive HspB1. Mol Biol Cell 2017; 28:2661-2675. [PMID: 28768826 PMCID: PMC5620374 DOI: 10.1091/mbc.e17-02-0087] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/24/2017] [Accepted: 07/28/2017] [Indexed: 01/12/2023] Open
Abstract
Mechanical force induces protein phosphorylations, subcellular redistributions, and actin remodeling. We show that mechanical activation of the p38 MAPK pathway leads to phosphorylation of HspB1 (hsp25/27), which redistributes to cytoskeletal structures, and contributes to the actin cytoskeletal remodeling induced by mechanical stimulation. Despite the importance of a cell’s ability to sense and respond to mechanical force, the molecular mechanisms by which physical cues are converted to cell-instructive chemical information to influence cell behaviors remain to be elucidated. Exposure of cultured fibroblasts to uniaxial cyclic stretch results in an actin stress fiber reinforcement response that stabilizes the actin cytoskeleton. p38 MAPK signaling is activated in response to stretch, and inhibition of p38 MAPK abrogates stretch-induced cytoskeletal reorganization. Here we show that the small heat shock protein HspB1 (hsp25/27) is phosphorylated in stretch-stimulated mouse fibroblasts via a p38 MAPK-dependent mechanism. Phosphorylated HspB1 is recruited to the actin cytoskeleton, displaying prominent accumulation on actin “comet tails” that emanate from focal adhesions in stretch-stimulated cells. Site-directed mutagenesis to block HspB1 phosphorylation inhibits the protein’s cytoskeletal recruitment in response to mechanical stimulation. HspB1-null cells, generated by CRISPR/Cas9 nuclease genome editing, display an abrogated stretch-stimulated actin reinforcement response and increased cell migration. HspB1 is recruited to sites of increased traction force in cells geometrically constrained on micropatterned substrates. Our findings elucidate a molecular pathway by which a mechanical signal is transduced via activation of p38 MAPK to influence actin remodeling and cell migration via a zyxin-independent process.
Collapse
Affiliation(s)
- Laura Hoffman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Biology, University of Utah, Salt Lake City, UT 84112
| | | | - Masaaki Yoshigi
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112
| | - Mary Beckerle
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 .,Department of Biology, University of Utah, Salt Lake City, UT 84112.,Department of Pediatrics, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
22
|
Stretch-induced actomyosin contraction in epithelial tubes: Mechanotransduction pathways for tubular homeostasis. Semin Cell Dev Biol 2017; 71:146-152. [PMID: 28610943 DOI: 10.1016/j.semcdb.2017.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 05/24/2017] [Indexed: 02/08/2023]
Abstract
Many tissues in our body have a tubular shape and are constantly exposed to various stresses. Luminal pressure imposes tension on the epithelial and myoepithelial or smooth muscle cells surrounding the lumen of the tubes. Contractile forces generated by actomyosin assemblies within these cells oppose the luminal pressure and must be calibrated to maintain tube diameter homeostasis and tissue integrity. In this review, we discuss mechanotransduction pathways that can lead from sensation of cell stretch to activation of actomyosin contractility, providing rapid mechanochemical feedback for proper tubular tissue function.
Collapse
|
23
|
Rosner SR, Pascoe CD, Blankman E, Jensen CC, Krishnan R, James AL, Elliot JG, Green FH, Liu JC, Seow CY, Park JA, Beckerle MC, Paré PD, Fredberg JJ, Smith MA. The actin regulator zyxin reinforces airway smooth muscle and accumulates in airways of fatal asthmatics. PLoS One 2017; 12:e0171728. [PMID: 28278518 PMCID: PMC5344679 DOI: 10.1371/journal.pone.0171728] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/24/2017] [Indexed: 01/21/2023] Open
Abstract
Bronchospasm induced in non-asthmatic human subjects can be easily reversed by a deep inspiration (DI) whereas bronchospasm that occurs spontaneously in asthmatic subjects cannot. This physiological effect of a DI has been attributed to the manner in which a DI causes airway smooth muscle (ASM) cells to stretch, but underlying molecular mechanisms-and their failure in asthma-remain obscure. Using cells and tissues from wild type and zyxin-/- mice we report responses to a transient stretch of physiologic magnitude and duration. At the level of the cytoskeleton, zyxin facilitated repair at sites of stress fiber fragmentation. At the level of the isolated ASM cell, zyxin facilitated recovery of contractile force. Finally, at the level of the small airway embedded with a precision cut lung slice, zyxin slowed airway dilation. Thus, at each level zyxin stabilized ASM structure and contractile properties at current muscle length. Furthermore, when we examined tissue samples from humans who died as the result of an asthma attack, we found increased accumulation of zyxin compared with non-asthmatics and asthmatics who died of other causes. Together, these data suggest a biophysical role for zyxin in fatal asthma.
Collapse
Affiliation(s)
- Sonia R. Rosner
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Christopher D. Pascoe
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Elizabeth Blankman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Christopher C. Jensen
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Alan L. James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, West Australia, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - John G. Elliot
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, West Australia, Australia
| | - Francis H. Green
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeffrey C. Liu
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Chun Y. Seow
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Jin-Ah Park
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Mary C. Beckerle
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
| | - Peter D. Paré
- University of British Columbia Center for Heart Lung Innovation, St Paul Hospital, Vancouver, British Columbia, Canada
| | - Jeffrey J. Fredberg
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Mark A. Smith
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
24
|
Modjeski KL, Ture SK, Field DJ, Cameron SJ, Morrell CN. Glutamate Receptor Interacting Protein 1 Mediates Platelet Adhesion and Thrombus Formation. PLoS One 2016; 11:e0160638. [PMID: 27631377 PMCID: PMC5025166 DOI: 10.1371/journal.pone.0160638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/23/2016] [Indexed: 01/08/2023] Open
Abstract
Thrombosis-associated pathologies, such as myocardial infarction and stroke, are major causes of morbidity and mortality worldwide. Because platelets are necessary for hemostasis and thrombosis, platelet directed therapies must balance inhibiting platelet function with bleeding risk. Glutamate receptor interacting protein 1 (GRIP1) is a large scaffolding protein that localizes and organizes interacting proteins in other cells, such as neurons. We have investigated the role of GRIP1 in platelet function to determine its role as a molecular scaffold in thrombus formation. Platelet-specific GRIP1-/- mice were used to determine the role of GRIP1 in platelets. GRIP1-/- mice had normal platelet counts, but a prolonged bleeding time and delayed thrombus formation in a FeCl3-induced vessel injury model. In vitro stimulation of WT and GRIP1-/- platelets with multiple agonists showed no difference in platelet activation. However, in vivo platelet rolling velocity after endothelial stimulation was significantly greater in GRIP1-/- platelets compared to WT platelets, indicating a potential platelet adhesion defect. Mass spectrometry analysis of GRIP1 platelet immunoprecipitation revealed enrichment of GRIP1 binding to GPIb-IX complex proteins. Western blots confirmed the mass spectrometry findings that GRIP1 interacts with GPIbα, GPIbβ, and 14-3-3. Additionally, in resting GRIP1-/- platelets, GPIbα and 14-3-3 have increased interaction compared to WT platelets. GRIP1 interactions with the GPIb-IX binding complex are necessary for normal platelet adhesion to a stimulated endothelium.
Collapse
Affiliation(s)
- Kristina L. Modjeski
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, United States of America
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Sara K. Ture
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, United States of America
| | - David J. Field
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Scott J. Cameron
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, United States of America
- * E-mail:
| |
Collapse
|
25
|
Haubner BJ, Moik D, Schuetz T, Reiner MF, Voelkl JG, Streil K, Bader K, Zhao L, Scheu C, Mair J, Pachinger O, Metzler B. In vivo cardiac role of migfilin during experimental pressure overload. Cardiovasc Res 2015; 106:398-407. [DOI: 10.1093/cvr/cvv125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/14/2015] [Indexed: 11/14/2022] Open
|
26
|
Young JS, Vogl AW. Focal adhesion proteins Zyxin and Vinculin are co-distributed at tubulobulbar complexes. SPERMATOGENESIS 2014; 2:63-68. [PMID: 22553491 PMCID: PMC3341248 DOI: 10.4161/spmg.19391] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Tubulobulbar complexes (TBCs) are actin-related double-membrane invaginations formed at intercellular junctions in the seminiferous epithelium of mammalian testis. They occur at basal junction complexes between neighboring Sertoli cells and at apical junctions between Sertoli cells and spermatids. They are proposed to internalize intercellular junctions during the translocation of spermatocytes from basal to adluminal compartments of the seminiferous epithelium, and during sperm release from Sertoli cells. Although TBCs are specific to the seminiferous epithelium, they morphologically resemble podosomes in osteoclasts. Previously, we have reported that a key group of proteins consisting of N-WASp, Arp2/3, cortactin and dynamin that occur at podosomes also is present at TBCs. Here we explore the prediction that zyxin, a focal adhesion protein known to be present at podosomes, also is present at apical TBCs. A rabbit polyclonal anti-zyxin antibody (B71) was used to label fixed fragments and frozen sections of testis. In both fragments and sections, B71 labeled tubular regions of TBCs at apical sites of attachment between Sertoli cells and spermatids, in addition to being localized at actin related intercellular adhesion junctions termed ectoplasmic specializations. Although the function of zyxin at TBCs has yet to be determined, the protein is known to interact with the cytoplasmic domain of integrins at focal adhesions, and integrins are known to be present in TBCs.
Collapse
Affiliation(s)
- J'nelle S Young
- Department of Cellular and Physiological Sciences; Faculty of Medicine; Life Sciences Centre; The University of British Columbia; Vancouver, BC Canada
| | | |
Collapse
|
27
|
Luo S, Schaefer AM, Dour S, Nonet ML. The conserved LIM domain-containing focal adhesion protein ZYX-1 regulates synapse maintenance in Caenorhabditis elegans. Development 2014; 141:3922-33. [PMID: 25252943 DOI: 10.1242/dev.108217] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We describe the identification of zyxin as a regulator of synapse maintenance in mechanosensory neurons in C. elegans. zyx-1 mutants lacked PLM mechanosensory synapses as adult animals. However, most PLM synapses initially formed during development but were subsequently lost as the animals developed. Vertebrate zyxin regulates cytoskeletal responses to mechanical stress in culture. Our work provides in vivo evidence in support of such a role for zyxin. In particular, zyx-1 mutant synaptogenesis phenotypes were suppressed by disrupting locomotion of the mutant animals, suggesting that zyx-1 protects mechanosensory synapses from locomotion-induced forces. In cultured cells, zyxin is recruited to focal adhesions and stress fibers via C-terminal LIM domains and modulates cytoskeletal organization via the N-terminal domain. The synapse-stabilizing activity was mediated by a short isoform of ZYX-1 containing only the LIM domains. Consistent with this notion, PLM synaptogenesis was independent of α-actinin and ENA-VASP, both of which bind to the N-terminal domain of zyxin. Our results demonstrate that the LIM domain moiety of zyxin functions autonomously to mediate responses to mechanical stress and provide in vivo evidence for a role of zyxin in neuronal development.
Collapse
Affiliation(s)
- Shuo Luo
- Department of Anatomy and Neurobiology, Washington University Medical School, 660 S Euclid Ave, St Louis, MO 63110, USA
| | - Anneliese M Schaefer
- Department of Anatomy and Neurobiology, Washington University Medical School, 660 S Euclid Ave, St Louis, MO 63110, USA Department of Neurology, Washington University Medical School, 660 S Euclid Ave, St Louis, MO 63110, USA
| | - Scott Dour
- Department of Anatomy and Neurobiology, Washington University Medical School, 660 S Euclid Ave, St Louis, MO 63110, USA
| | - Michael L Nonet
- Department of Anatomy and Neurobiology, Washington University Medical School, 660 S Euclid Ave, St Louis, MO 63110, USA
| |
Collapse
|
28
|
Miyazaki Y, Yusa T, Matsuo S, Terauchi Y, Miyazaki S. Zyxin modulates the transmigration of Haemophilus influenzae to the central nervous system. Virulence 2014; 5:665-72. [PMID: 25025691 DOI: 10.4161/viru.29786] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The mechanism by which Haemophilus influenzae causes meningitis is unclear. Previously, we established murine meningitis by intranasal instillation of H. influenzae as a cell-bound organism (CBO). In this study, we aimed to identify the molecules associated with inhibiting the transmigration of cells across the blood-brain barrier (BBB). Two-dimensional difference gel electrophoresis and protein identification by mass spectrometry were used for proteomic analysis. Analysis of the membranous extract from a tumor necrosis factor (TNF)-α-treated human brain microvascular endothelial cell (HBMEC) monolayer revealed 41 differentially expressed proteins. Zyxin, which is thought to be essential for tight cell-to-cell junctions, decreased 1.8-fold in TNF-α-treated HBMECs. In addition, zyxin transcript levels decreased 1.5-fold in cells derived from TNF-α-treated HBMECs. Intranasal instillation of CBOs in zyxin-deficient mice resulted in a significant higher mortality rate than in wild-type mice. Transmigration of CBOs across a HBMEC monolayer pretreated with TNF-α (1 ng/mL), interleukin (IL)-1β (10 ng/mL), or lipopolysaccharide (LPS; 10 ng/mL) was assayed by counting CBOs that migrated from an upper chamber into a lower chamber. HBMEC pretreated with TNF-α exhibited significantly greater migration (P<0.01) than did control cells or cells treated with IL-1β or LPS. Our findings highlight that zyxin is an important protein protecting the tight junction of the BBB against cell transmigration across the BBB. Finally, TNF-α produced in respiratory infection when the primary infection reached the BBB caused decreased zyxin levels in BBB cell membranes. Furthermore, H. influenzae reaching the BBB as CBOs could transmigrate into cerebrospinal fluid across the zyxin-decreased BBB.
Collapse
Affiliation(s)
- Yuko Miyazaki
- Department of Endocrinology and Metabolism; Yokohama City University School of Medicine; Yokohama, Kanagawa, Japan
| | - Takashi Yusa
- Division of Microbiology and Immunology; Center for Advanced Research; Graduate School of Medical Sciences; Toho University; Ota, Tokyo, Japan
| | - Saburo Matsuo
- Laboratory of Toxicology; Course of Veterinary Science; Graduate School of Life and Environmental Biosciences; Osaka Prefecture University; Izumisano, Osaka, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism; Yokohama City University School of Medicine; Yokohama, Kanagawa, Japan
| | - Shuichi Miyazaki
- Division of Microbiology and Immunology; Center for Advanced Research; Graduate School of Medical Sciences; Toho University; Ota, Tokyo, Japan
| |
Collapse
|
29
|
Smith MA, Hoffman LM, Beckerle MC. LIM proteins in actin cytoskeleton mechanoresponse. Trends Cell Biol 2014; 24:575-83. [PMID: 24933506 DOI: 10.1016/j.tcb.2014.04.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 04/23/2014] [Accepted: 04/30/2014] [Indexed: 01/21/2023]
Abstract
The actin cytoskeleton assembles into branched networks or bundles to generate mechanical force for critical cellular processes such as establishment of polarity, adhesion, and migration. Stress fibers (SFs) are contractile actomyosin structures that physically couple to the extracellular matrix through integrin-based focal adhesions (FAs), thereby transmitting force into and across the cell. Recently, LIN-11, Isl1, and MEC-3 (LIM) domain proteins have been implicated in mediating this cytoskeletal mechanotransduction. Among the more well-studied LIM domain adapter proteins is zyxin, a dynamic component of both FAs and SFs. Here we discuss recent research detailing the mechanisms by which SFs adjust their structure and composition to balance mechanical forces and suggest ways that zyxin and other LIM domain proteins mediate mechanoresponse.
Collapse
Affiliation(s)
- M A Smith
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - L M Hoffman
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - M C Beckerle
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
30
|
Van Itallie CM, Tietgens AJ, Aponte A, Fredriksson K, Fanning AS, Gucek M, Anderson JM. Biotin ligase tagging identifies proteins proximal to E-cadherin, including lipoma preferred partner, a regulator of epithelial cell-cell and cell-substrate adhesion. J Cell Sci 2013; 127:885-95. [PMID: 24338363 DOI: 10.1242/jcs.140475] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Known proteins associated with the cell-adhesion protein E-cadherin include catenins and proteins involved in signaling, trafficking and actin organization. However, the list of identified adherens junction proteins is likely to be incomplete, limiting investigation into this essential cell structure. To expand the inventory of potentially relevant proteins, we expressed E-cadherin fused to biotin ligase in MDCK epithelial cells, and identified by mass spectrometry neighboring proteins that were biotinylated. The most abundant of the 303 proteins identified were catenins and nearly 40 others that had been previously reported to influence cadherin function. Many others could be rationalized as novel candidates for regulating the adherens junction, cytoskeleton, trafficking or signaling. We further characterized lipoma preferred partner (LPP), which is present at both cell contacts and focal adhesions. Knockdown of LPP demonstrated its requirement for E-cadherin-dependent adhesion and suggested that it plays a role in coordination of the cell-cell and cell-substrate cytoskeletal interactions. The analysis of LPP function demonstrates proof of principle that the proteomic analysis of E-cadherin proximal proteins expands the inventory of components and tools for understanding the function of E-cadherin.
Collapse
Affiliation(s)
- Christina M Van Itallie
- Laboratory of Tight Junction Structure and Function, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Duperret EK, Ridky TW. Focal adhesion complex proteins in epidermis and squamous cell carcinoma. Cell Cycle 2013; 12:3272-85. [PMID: 24036537 DOI: 10.4161/cc.26385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Focal adhesions (FAs) are large, integrin-containing, multi-protein assemblies spanning the plasma membrane that link the cellular cytoskeleton to surrounding extracellular matrix. They play critical roles in adhesion and cell signaling and are major regulators of epithelial homeostasis, tissue response to injury, and tumorigenesis. Most integrin subunits and their associated FA proteins are expressed in skin, and murine genetic models have provided insight into the functional roles of FAs in normal and neoplastic epidermis. Here, we discuss the roles of these proteins in normal epidermal proliferation, adhesion, wound healing, and cancer. While many downstream signaling mechanisms remain unclear, the critically important roles of FAs are highlighted by the development of therapeutics targeting FAs for human cancer.
Collapse
|
32
|
Li N, Goodwin RL, Potts JD. Zyxin regulates cell migration and differentiation in EMT during chicken AV valve morphogenesis. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2013; 19:842-854. [PMID: 23742986 DOI: 10.1017/s1431927613001633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
During heart valve development, epithelial-mesenchymal transformation (EMT) is a key process for valve formation. EMT leads to the generation of mesenchymal cells that will eventually become the interstitial cells (fibroblasts) of the mature valve. During EMT, cell architecture and motility change markedly; significant changes are also observed in various signaling pathways. Here we systematically examined the expression, localization, and function of zyxin, a focal adhesion protein, in EMT during atrioventricular (AV) valve morphogenesis. Expression and localization studies showed that zyxin was expressed in the AV canal region during crucial stages of valve development. An in vitro 3D collagen gel culture system was used to determine zyxin function either after siRNA gene knockdown or after overexpression. Our studies revealed that zyxin overexpression inhibited endocardial cell migration and cell differentiation and also led to a decrease in the number of migrating mesenchymal cells. Moreover, correlative cytoskeletal changes were apparent in response to both overexpression and knockdown treatments. Thus, zyxin appears to play a role as a regulator of cell migration and differentiation during EMT in chicken AV valve formation.
Collapse
Affiliation(s)
- Na Li
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | | | | |
Collapse
|
33
|
Lecroisey C, Brouilly N, Qadota H, Mariol MC, Rochette NC, Martin E, Benian GM, Ségalat L, Mounier N, Gieseler K. ZYX-1, the unique zyxin protein of Caenorhabditis elegans, is involved in dystrophin-dependent muscle degeneration. Mol Biol Cell 2013; 24:1232-49. [PMID: 23427270 PMCID: PMC3623643 DOI: 10.1091/mbc.e12-09-0679] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In vertebrates, zyxin is a LIM-domain protein belonging to a family composed of seven members. We show that the nematode Caenorhabditis elegans has a unique zyxin-like protein, ZYX-1, which is the orthologue of the vertebrate zyxin subfamily composed of zyxin, migfilin, TRIP6, and LPP. The ZYX-1 protein is expressed in the striated body-wall muscles and localizes at dense bodies/Z-discs and M-lines, as well as in the nucleus. In yeast two-hybrid assays ZYX-1 interacts with several known dense body and M-line proteins, including DEB-1 (vinculin) and ATN-1 (α-actinin). ZYX-1 is mainly localized in the middle region of the dense body/Z-disk, overlapping the apical and basal regions containing, respectively, ATN-1 and DEB-1. The localization and dynamics of ZYX-1 at dense bodies depend on the presence of ATN-1. Fluorescence recovery after photobleaching experiments revealed a high mobility of the ZYX-1 protein within muscle cells, in particular at dense bodies and M-lines, indicating a peripheral and dynamic association of ZYX-1 at these muscle adhesion structures. A portion of the ZYX-1 protein shuttles from the cytoplasm into the nucleus, suggesting a role for ZYX-1 in signal transduction. We provide evidence that the zyx-1 gene encodes two different isoforms, ZYX-1a and ZYX-1b, which exhibit different roles in dystrophin-dependent muscle degeneration occurring in a C. elegans model of Duchenne muscular dystrophy.
Collapse
|
34
|
Suresh Babu S, Wojtowicz A, Freichel M, Birnbaumer L, Hecker M, Cattaruzza M. Mechanism of stretch-induced activation of the mechanotransducer zyxin in vascular cells. Sci Signal 2012; 5:ra91. [PMID: 23233529 DOI: 10.1126/scisignal.2003173] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular cells respond to supraphysiological amounts of stretch with a characteristic phenotypic change that results in dysfunctional remodeling of the affected arteries. Although the pathophysiological consequences of stretch-induced signaling are well characterized, the mechanism of mechanotransduction is unclear. We focused on the mechanotransducer zyxin, which translocates to the nucleus to drive gene expression in response to stretch. In cultured human endothelial cells and perfused femoral arteries isolated from wild-type and several knockout mouse strains, we characterized a multistep signaling pathway whereby stretch led to a transient receptor potential channel 3-mediated release of the endothelial vasoconstrictor peptide endothelin-1 (ET-1). ET-1, through autocrine activation of its B-type receptor, elicited the release of pro-atrial natriuretic peptide (ANP), which caused the autocrine activation of the ANP receptor guanylyl cyclase A (GC-A). Activation of GC-A, in turn, led to protein kinase G-mediated phosphorylation of zyxin at serine 142, thereby triggering the translocation of zyxin to the nucleus, where it was required for stretch-induced gene expression. Thus, we have identified a stretch-induced signaling pathway in vascular cells that leads to the activation of zyxin, a cytoskeletal protein specifically involved in transducing mechanical stimuli.
Collapse
Affiliation(s)
- Sahana Suresh Babu
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Rajput R, Khanna M, Kumar P, Kumar B, Sharma S, Gupta N, Saxena L. Small interfering RNA targeting the nonstructural gene 1 transcript inhibits influenza A virus replication in experimental mice. Nucleic Acid Ther 2012; 22:414-22. [PMID: 23062009 DOI: 10.1089/nat.2012.0359] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Nonstructural protein 1 (NS1) of influenza A viruses counteracts the host immune response against the influenza viruses by not only inhibiting the nuclear export and maturation of host cell messenger RNA (mRNA), but by also blocking the double-stranded RNA-activated protein kinase-mediated inhibition of viral RNA translation. Reduction of NS1 gene product in the host cell may be a potent antiviral strategy to provide protection against the influenza virus infection. We used small interfering RNAs (siRNAs) synthesized against the viral mRNA to down regulate the NS1 gene and observed its effect on inhibition of virus replication. When NS1 gene-specific siRNA were transfected in Madin Darby canine kidney (MDCK) cells followed by influenza A virus infection, approximately 60% inhibition in intracellular levels of NS1 RNA was observed. When siRNA was administered in BALB/c mice, 92% reduction in the levels of NS1 gene expression in mice lungs was observed. A significant reduction in the lung virus titers and cytokine levels was also detected in the presence of siRNAs as compared with the untreated control. The study was validated by the use of selectively disabled mutants of each set of siRNA. Our findings suggest that siRNA targeted against NS1 gene of influenza A virus can provide considerable protection to the virus-infected host cells and may be used as potential candidates for nucleic acid-based antiviral therapy for prevention of influenza A virus infection.
Collapse
Affiliation(s)
- Roopali Rajput
- Department of Respiratory Virology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | | | | | | | | | | | | |
Collapse
|
36
|
Hirata H, Tatsumi H, Sokabe M. Zyxin emerges as a key player in the mechanotransduction at cell adhesive structures. Commun Integr Biol 2012; 1:192-5. [PMID: 19513257 DOI: 10.4161/cib.1.2.7001] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 09/17/2008] [Indexed: 01/09/2023] Open
Abstract
Actin stress fiber (SF), focal adhesion (FA) and adherens junction (AJ) are known structures whose formation and development are mechanical force-dependent. At these structures, actin is actively polymerized, which in turn contributes the development of these structures. Recently, we reported that actin polymerization at FAs is facilitated by mechanical forces, which was critically dependent on the force-induced recruitment of the LIM protein zyxin to FAs. Zyxin enhances actin polymerization with the aid of Ena/VASP proteins. Both zyxin and Ena/VASP proteins are localized not only to FAs but also to AJs and SFs, facilitating actin polymerization at these structures. We discuss here the possibility that zyxin is a common mechanotransducer element regulating actin polymerization at FAs, AJs and SFs.
Collapse
Affiliation(s)
- Hiroaki Hirata
- Cell Mechanosensing Project; ICORP/SORST; Japan Science and Technology Agency; Nagoya Japan; Department of Molecular Physiology; National Institute for Physiological Sciences; National Institutes of Natural Sciences; Okazaki Japan; Department of Physiology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | | | | |
Collapse
|
37
|
Mise N, Savai R, Yu H, Schwarz J, Kaminski N, Eickelberg O. Zyxin is a transforming growth factor-β (TGF-β)/Smad3 target gene that regulates lung cancer cell motility via integrin α5β1. J Biol Chem 2012; 287:31393-405. [PMID: 22778267 DOI: 10.1074/jbc.m112.357624] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although TGF-β acts as a tumor suppressor in normal tissues and in early carcinogenesis, these tumor suppressor effects are lost in advanced malignancies. Single cell migration and epithelial-mesenchymal transition (EMT), both of which are regulated by TGF-β, are critical steps in mediating cancer progression. Here, we sought to identify novel direct targets of TGF-β signaling in lung cancer cells and have indentified the zyxin gene as a target of Smad3-mediated TGF-β1 signaling. Zyxin concentrates at focal adhesions and along the actin cytoskeleton; as such, we hypothesized that cytoskeletal organization, motility, and EMT in response to TGF-β1 might be regulated by zyxin expression. We show that TGF-β1 treatment of lung cancer cells caused rapid phospho-Smad3-dependent expression of zyxin. Zyxin expression was critical for the formation and integrity of cell adherens junctions. Silencing of zyxin decreased expression of the focal adhesion protein vasodilator-activated phospho-protein (VASP), although the formation and morphology of focal adhesions remained unchanged. Zyxin-depleted cells displayed significantly increased integrin α5β1 levels, accompanied by enhanced adhesion to fibronectin and acquisition of a mesenchymal phenotype in response to TGF-β1. Zyxin silencing led to elevated integrin α5β1-dependent single cell motility. Importantly, these features are mirrored in the K-ras-driven mouse model of lung cancer. Here, lung tumors revealed decreased levels of both zyxin and phospho-Smad3 when compared with normal tissues. Our data thus demonstrate that zyxin is a novel functional target and effector of TGF-β signaling in lung cancer. By regulating cell-cell junctions, integrin α5β1 expression, and cell-extracellular matrix adhesion, zyxin may regulate cancer cell motility and EMT during lung cancer development and progression.
Collapse
Affiliation(s)
- Nikica Mise
- Comprehensive Pneumology Center, University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, 81377 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
38
|
Hoffman LM, Jensen CC, Chaturvedi A, Yoshigi M, Beckerle MC. Stretch-induced actin remodeling requires targeting of zyxin to stress fibers and recruitment of actin regulators. Mol Biol Cell 2012; 23:1846-59. [PMID: 22456508 PMCID: PMC3350550 DOI: 10.1091/mbc.e11-12-1057] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Mechanical stimulation induces zyxin-dependent actin cytoskeletal reinforcement. Stretch induces MAPK activation, zyxin phosphorylation, and recruitment to actin stress fibers, independent of p130Cas. Zyxin's C-terminal LIM domains are required for stretch-induced targeting to stress fibers, and zyxin's N-terminus is necessary for actin remodeling. Reinforcement of actin stress fibers in response to mechanical stimulation depends on a posttranslational mechanism that requires the LIM protein zyxin. The C-terminal LIM region of zyxin directs the force-sensitive accumulation of zyxin on actin stress fibers. The N-terminal region of zyxin promotes actin reinforcement even when Rho kinase is inhibited. The mechanosensitive integrin effector p130Cas binds zyxin but is not required for mitogen-activated protein kinase–dependent zyxin phosphorylation or stress fiber remodeling in cells exposed to uniaxial cyclic stretch. α-Actinin and Ena/VASP proteins bind to the stress fiber reinforcement domain of zyxin. Mutation of their docking sites reveals that zyxin is required for recruitment of both groups of proteins to regions of stress fiber remodeling. Zyxin-null cells reconstituted with zyxin variants that lack either α-actinin or Ena/VASP-binding capacity display compromised response to mechanical stimulation. Our findings define a bipartite mechanism for stretch-induced actin remodeling that involves mechanosensitive targeting of zyxin to actin stress fibers and localized recruitment of actin regulatory machinery.
Collapse
Affiliation(s)
- Laura M Hoffman
- Departments of Biology and Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Integrating signals from the ECM (extracellular matrix) via the cell surface into the nucleus is an essential feature of multicellular life and often malfunctions in cancer. To date many signal transducers known as shuttle proteins have been identified that act as both: a cytoskeletal and a signalling protein. Here, we highlight the interesting member of the Zyxin family TRIP6 [thyroid receptor interactor protein 6; also designated ZRP-1 (zyxin-related protein 1)] and review current literature to define its role in cell physiology and cancer. TRIP6 is a versatile scaffolding protein at FAs (focal adhesions) involved in cytoskeletal organization, coordinated cell migration and tissue invasion. Via its LIM and TDC domains TRIP6 interacts with different components of the LPA (lysophosphatidic acid), NF-κB (nuclear factor κB), glucocorticoid and AMPK (AMP-activated protein kinase) signalling pathway and thereby modulates their activity. Within the nucleus TRIP6 acts as a transcriptional cofactor regulating the transcriptional responses of these pathways. Moreover, intranuclear TRIP6 associates with proteins ensuring telomere protection and hence may contribute to genome stability. Accordingly, TRIP6 is engaged in key cellular processes such as cell proliferation, differentiation and survival. These diverse functions of TRIP6 are found to be dysregulated in various cancers and may have pleiotropic roles in tumour initiation, tumour growth and metastasis, which turn TRIP6 into an attractive candidate for cancer diagnosis and targeted therapy.
Collapse
|
40
|
He H, Ding F, Li Y, Luo A, Chen H, Wu C, Liu Z. Migfilin Regulates Esophageal Cancer Cell Motility through Promoting GSK-3β–Mediated Degradation of β-Catenin. Mol Cancer Res 2012; 10:273-81. [DOI: 10.1158/1541-7786.mcr-11-0419] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Das M, Ithychanda SS, Qin J, Plow EF. Migfilin and filamin as regulators of integrin activation in endothelial cells and neutrophils. PLoS One 2011; 6:e26355. [PMID: 22043318 PMCID: PMC3197140 DOI: 10.1371/journal.pone.0026355] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/25/2011] [Indexed: 12/18/2022] Open
Abstract
Cell adhesion and migration depend on engagement of extracellular matrix ligands by integrins. Integrin activation is dynamically regulated by interactions of various cytoplasmic proteins, such as filamin and integrin activators, talin and kindlin, with the cytoplasmic tail of the integrin β subunit. Although filamin has been suggested to be an inhibitor of integrin activation, direct functional evidence for the inhibitory role of filamin is limited. Migfilin, a filamin-binding protein enriched at cell-cell and cell-extracellular matrix contact sites, can displace filamin from β1 and β3 integrins and promote integrin activation. However, its role in activation and functions of different β integrins in human vascular cells is unknown. In this study, using flow cytometry, we demonstrate that filamin inhibits β1 and αIIbβ3 integrin activation, and migfilin can overcome its inhibitory effect. Migfilin protein is widely expressed in different adherent and circulating blood cells and can regulate integrin activation in naturally-occurring vascular cells, endothelial cells and neutrophils. Migfilin can activate β1, β2 and β3 integrins and promote integrin mediated responses while migfilin depletion impairs the spreading and migration of endothelial cells. Thus, filamin can act broadly as an inhibitor and migfilin is a promoter of integrin activation.
Collapse
Affiliation(s)
- Mitali Das
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Sujay Subbayya Ithychanda
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jun Qin
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Edward F. Plow
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
42
|
Cell Adhesion and Transcriptional Activity - Defining the Role of the Novel Protooncogene LPP. Transl Oncol 2011; 2:107-16. [PMID: 19701494 DOI: 10.1593/tlo.09112] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 02/20/2009] [Accepted: 02/25/2009] [Indexed: 12/13/2022] Open
Abstract
Integrating signals from the extracellular matrix through the cell surface into the nucleus is an essential feature of metazoan life. To date, many signal transducers known as shuttle proteins have been identified to act as both a cytoskeletal and a signaling protein. Among them, the most prominent representatives are zyxin and lipoma preferred (translocation) partner (LPP). These proteins belong to the LIM domain protein family and are associated with cell migration, proliferation, and transcription. LPP was first identified in benign human lipomas and was subsequently found to be overexpressed in human malignancies such as lung carcinoma, soft tissue sarcoma, and leukemia. This review portrays LPP in the context of human neoplasia based on a study of the literature to define its important role as a novel protooncogene in carcinogenesis.
Collapse
|
43
|
Rauskolb C, Pan G, Reddy BVVG, Oh H, Irvine KD. Zyxin links fat signaling to the hippo pathway. PLoS Biol 2011; 9:e1000624. [PMID: 21666802 PMCID: PMC3110180 DOI: 10.1371/journal.pbio.1000624] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 04/27/2011] [Indexed: 01/15/2023] Open
Abstract
Using genetic and molecular analyses, the authors identify Zyx as a positive regulator of Hippo signaling and characterize its role within the pathway. The Hippo signaling pathway has a conserved role in growth control and is of fundamental importance during both normal development and oncogenesis. Despite rapid progress in recent years, key steps in the pathway remain poorly understood, in part due to the incomplete identification of components. Through a genetic screen, we identified the Drosophila Zyxin family gene, Zyx102 (Zyx), as a component of the Hippo pathway. Zyx positively regulates the Hippo pathway transcriptional co-activator Yorkie, as its loss reduces Yorkie activity and organ growth. Through epistasis tests, we position the requirement for Zyx within the Fat branch of Hippo signaling, downstream of Fat and Dco, and upstream of the Yorkie kinase Warts, and we find that Zyx is required for the influence of Fat on Warts protein levels. Zyx localizes to the sub-apical membrane, with distinctive peaks of accumulation at intercellular vertices. This partially overlaps the membrane localization of the myosin Dachs, which has similar effects on Fat-Hippo signaling. Co-immunoprecipitation experiments show that Zyx can bind to Dachs and that Dachs stimulates binding of Zyx to Warts. We also extend characterization of the Ajuba LIM protein Jub and determine that although Jub and Zyx share C-terminal LIM domains, they regulate Hippo signaling in distinct ways. Our results identify a role for Zyx in the Hippo pathway and suggest a mechanism for the role of Dachs: because Fat regulates the localization of Dachs to the membrane, where it can overlap with Zyx, we propose that the regulated localization of Dachs influences downstream signaling by modulating Zyx-Warts binding. Mammalian Zyxin proteins have been implicated in linking effects of mechanical strain to cell behavior. Our identification of Zyx as a regulator of Hippo signaling thus also raises the possibility that mechanical strain could be linked to the regulation of gene expression and growth through Hippo signaling. Processes that control cell numbers are essential during normal development, when they are required to generate organs of the correct size, and during cancinogenesis, when they influence tumor growth. The Hippo pathway is an intercellular signaling pathway that relays information about cell-cell contact and cell polarity to a signal transduction pathway that regulates the transcription of genes controlling cell numbers. The role of Hippo signaling in controlling growth is conserved from fruit flies to humans, but many aspects of the Hippo signal transduction pathway remain poorly understood. In this article, we identify Zyx as a previously unknown component of the Hippo pathway in Drosophila, and characterize its role within the pathway. We show that Zyx plays an essential role in a branch of Hippo signaling that involves the transmembrane receptor protein Fat and its target Dachs, which is a myosin family protein. Our results suggest a model in which Fat regulates the localization of Dachs, Dachs subsequently binds Zyx, stimulating its binding with the kinase Warts/Lats, and thereby regulates downstream signaling events. Zyx is conserved in vertebrates and we suggest that vertebrate Zyx proteins might also be involved in the regulation of Hippo signaling and, thereby, organ growth.
Collapse
Affiliation(s)
- Cordelia Rauskolb
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Guohui Pan
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - B. V. V. G. Reddy
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Hyangyee Oh
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Kenneth D. Irvine
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
44
|
Moik DV, Janbandhu VC, Fässler R. Loss of migfilin expression has no overt consequences on murine development and homeostasis. J Cell Sci 2011; 124:414-21. [PMID: 21224394 DOI: 10.1242/jcs.075960] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Migfilin is a LIM-domain-containing protein of the zyxin family of adaptor proteins and is found at cell-matrix and cell-cell adhesion sites and in the nucleus. In vitro studies have suggested that migfilin promotes β1 integrin activity, regulates cell spreading and migration and induces cardiomyocyte differentiation. To test directly the function of migfilin in vivo, we generated a migfilin-null mouse strain. Here, we report that loss of migfilin expression permits normal development and normal postnatal aging. Fibroblasts and keratinocytes from migfilin-null mice display normal spreading and adhesion, and normal integrin expression and activation. The migration velocity and directionality of migfilin-null embryonic fibroblasts were normal, whereas the velocity of migfilin-null keratinocytes in wound scratch assays was slightly but significantly reduced. Our findings indicate that the roles of migfilin are functionally redundant during mouse development and tissue homeostasis.
Collapse
Affiliation(s)
- Daniel V Moik
- Max-Planck-Institute of Biochemistry, Department of Molecular Medicine, 82152 Martinsried, Germany
| | | | | |
Collapse
|
45
|
Renfranz PJ, Blankman E, Beckerle MC. The cytoskeletal regulator zyxin is required for viability in Drosophila melanogaster. Anat Rec (Hoboken) 2010; 293:1455-69. [PMID: 20648572 DOI: 10.1002/ar.21193] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The zyxin family of proteins function as cytoskeletal regulators in adhesion, actin assembly, and cell motility. Though fibroblasts derived from zyxin-null mice show striking defects in motility and response to mechanical stimuli, the mice are viable and fertile. In Drosophila melanogaster, the family is represented by a single homologue, Zyx102. To study the role of zyxin during development, we generated a zyx102 RNA-interference transgenic line that allows for the conditional knockdown of Zyx102. When UAST-zyx102-dsRNAi expression is driven broadly by Actin5C-GAL4, loss of Zyx102 results in lethality during the pharate adult stage, a narrow developmental window during which the fly must molt, resorb molting fluid, fill adult trachea with air, and execute a behavioral program to eclose. Zyx102 knockdown animals attempt to emerge, but their adult trachea do not fill with air. If dissected from the pupal case, knockdown individuals appear morphologically normal, but remain inviable.
Collapse
Affiliation(s)
- Patricia J Renfranz
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112-5550, USA
| | | | | |
Collapse
|
46
|
Nguyen TN, Uemura A, Shih W, Yamada S. Zyxin-mediated actin assembly is required for efficient wound closure. J Biol Chem 2010; 285:35439-45. [PMID: 20801875 DOI: 10.1074/jbc.m110.119487] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cytoskeletal regulation of cell adhesion is vital to the organization of multicellular structures. The focal adhesion protein zyxin emerged as a key regulator of actin assembly because zyxin recruits Enabled/vasodilator-stimulated phospho-proteins (Ena/VASP) to promote actin assembly. Zyxin also localizes to the sites of cell-cell adhesion and is thought to promote actin assembly with Ena/VASP. Using shRNA targeted to zyxin, we analyzed the roles of zyxin at adhesive contacts. In zyxin-deficient cells, the actin assembly at both focal adhesion and cell-cell adhesion was limited, but their migration rate was unchanged. Cell spreading on E-cadherin-coated surfaces and the formation of cell clusters were slower for zyxin-deficient cells than wild type cells. By ablating a single cell within a cell monolayer, we quantified the rate of wound closure driven by a contractile circumferential actin ring. Zyxin-deficient cells failed to recruit VASP to cell-cell junctions at the wound edge and had a slower wound closure rate than wild type cells. Our results suggest that, by recruiting VASP, zyxin regulates actin assembly at the sites of force-bearing cell-cell adhesion.
Collapse
Affiliation(s)
- Thuc Nghi Nguyen
- Biomedical Engineering Department, University of California, Davis, California 95616, USA
| | | | | | | |
Collapse
|
47
|
siRNA for Influenza Therapy. Viruses 2010; 2:1448-1457. [PMID: 21994689 PMCID: PMC3185718 DOI: 10.3390/v2071448] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 07/05/2010] [Accepted: 07/07/2010] [Indexed: 11/17/2022] Open
Abstract
Influenza virus is one of the most prevalent and ancient infections in humans. About a fifth of world’s population is infected by influenza virus annually, leading to high morbidity and mortality, particularly in infants, the elderly and the immunocompromised. In the US alone, influenza outbreaks lead to roughly 30,000 deaths each year. Current vaccines and anti-influenza drugs are of limited use due to high mutation rate of the virus and side effects. In recent years, RNA interference, triggered by synthetic short interfering RNA (siRNA), has rapidly evolved as a potent antiviral regimen. Properly designed siRNAs have been shown to function as potent inhibitors of influenza virus replication. The siRNAs outperform traditional small molecule antivirals in a number of areas, such as ease of design, modest cost, and fast turnaround. Although specificity and tissue delivery remain major bottlenecks in the clinical applications of RNAi in general, intranasal application of siRNA against respiratory viruses including, but not limited to influenza virus, has experienced significant success and optimism, which is reviewed here.
Collapse
|
48
|
Hervy M, Hoffman LM, Jensen CC, Smith M, Beckerle MC. The LIM Protein Zyxin Binds CARP-1 and Promotes Apoptosis. Genes Cancer 2010; 1:506-515. [PMID: 20852740 DOI: 10.1177/1947601910376192] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Zyxin is a dual-function LIM domain protein that regulates actin dynamics in response to mechanical stress and shuttles between focal adhesions and the cell nucleus. Here we show that zyxin contributes to UV-induced apoptosis. Exposure of wild-type fibroblasts to UV-C irradiation results in apoptotic cell death, whereas cells harboring a homozygous disruption of the zyxin gene display a statistically significant survival advantage. To gain insight into the molecular mechanism by which zyxin promotes apoptotic signaling, we expressed an affinity-tagged zyxin variant in zyxin-null cells and isolated zyxin-associated proteins from cell lysates under physiological conditions. A 130-kDa protein that was co-isolated with zyxin was identified by microsequence analysis as the Cell Cycle and Apoptosis Regulator Protein-1 (CARP-1). CARP-1 associates with the LIM region of zyxin. Zyxin lacking the CARP-1 binding region shows reduced proapoptotic activity in response to UV-C irradiation. We demonstrate that CARP-1 is a nuclear protein. Zyxin is modified by phosphorylation in cells exposed to UV-C irradiation, and nuclear accumulation of zyxin is induced by UV-C exposure. These findings highlight a novel mechanism for modulating the apoptotic response to UV irradiation.
Collapse
Affiliation(s)
- Martial Hervy
- Huntsman Cancer Institute, Departments of Biology and Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | | | | | | | | |
Collapse
|
49
|
Sperry RB, Bishop NH, Bramwell JJ, Brodeur MN, Carter MJ, Fowler BT, Lewis ZB, Maxfield SD, Staley DM, Vellinga RM, Hansen MDH. Zyxin controls migration in epithelial-mesenchymal transition by mediating actin-membrane linkages at cell-cell junctions. J Cell Physiol 2010; 222:612-24. [PMID: 19927303 DOI: 10.1002/jcp.21977] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Development is punctuated by morphogenetic rearrangements of epithelial tissues, including detachment of motile cells during epithelial-mesenchymal transition (EMT). Dramatic actin rearrangements occur as cell-cell junctions are dismantled and cells become independently motile during EMT. Characterizing dynamic actin rearrangements and identifying actin machinery driving these rearrangements is essential for understanding basic mechanisms of cell-cell junction remodeling. Using immunofluorescence and live cell imaging of scattering MDCK cells we examine dynamic actin rearrangement events during EMT and demonstrate that zyxin-VASP complexes mediate linkage of dynamic medial actin networks to adherens junction (AJ) membranes. A functional analysis of zyxin in EMT reveals its role in regulating disruption of actin membrane linkages at cell-cell junctions, altering cells' ability to fully detach and migrate independently during EMT. Expression of a constitutively active zyxin mutant results in persistent actin-membrane linkages and cell migration without loss of cell-cell adhesion. We propose zyxin functions in morphogenetic rearrangements, maintaining collective migration by transducing individual cells' movements through AJs, thus preventing the dissociation of individual migratory cells.
Collapse
Affiliation(s)
- Rebecca Bakkevig Sperry
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah 84602, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Srikanchai T, Murani E, Phatsara C, Schwerin M, Schellander K, Ponsuksili S, Wimmers K. Association of ZYX polymorphisms with carcass and meat quality traits in commercial pigs. Meat Sci 2009; 84:159-64. [PMID: 20374769 DOI: 10.1016/j.meatsci.2009.08.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Revised: 03/25/2009] [Accepted: 08/21/2009] [Indexed: 10/20/2022]
Abstract
Zyxin (ZYX) is one of the proteins in focal adhesions along the actin fibers playing a role in actin organization and signal transduction. By radiation hybrid and genetic mapping we assigned ZYX to porcine chromosome 18 in the area of quantitative traits loci for carcass and meat quality and muscle fiber traits and hence considered ZYX a functional positional candidate gene. Analysis of a newly detected SNPs (c.+279 C>T, c.+399 A>G, c.+522 A>G) in pigs from different commercial breeds (Pietrain [Pi], German Landrace [LR], German Large White x German Landrace [F1] and PiF1) revealed a significant association with carcass traits (including: side- and backfat thickness, loin weight and carcass lean content) and meat quality traits (including: pH, color and drip loss). However, the lack of consistent association across all pig populations in this study indicates that the association of the SNPs may be depending on causal mutations in linkage disequilibrium and/or interactions with other loci.
Collapse
Affiliation(s)
- T Srikanchai
- Research Institute for the Biology of Farm Animals, Dummerstorf, Germany
| | | | | | | | | | | | | |
Collapse
|