1
|
Parise A, Magistrato A. Assessing the mechanism of fast-cycling cancer-associated mutations of Rac1 small Rho GTPase. Protein Sci 2024; 33:e4939. [PMID: 38501467 PMCID: PMC10949326 DOI: 10.1002/pro.4939] [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: 09/07/2023] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 03/20/2024]
Abstract
Rho-GTPases proteins function as molecular switches alternating from an active to an inactive state upon Guanosine triphosphate (GTP) binding and hydrolysis to Guanosine diphosphate (GDP). Among them, Rac subfamily regulates cell dynamics, being overexpressed in distinct cancer types. Notably, these proteins are object of frequent cancer-associated mutations at Pro29 (P29S, P29L, and P29Q). To assess the impact of these mutations on Rac1 structure and function, we performed extensive all-atom molecular dynamics simulations on wild-type (wt) and oncogenic isoforms of this protein in GDP- and GTP-bound states. Our results unprecedentedly elucidate that P29Q/S-induced structural and dynamical perturbations of Rac1 core domain weaken the binding of the catalytic site Mg2+ ion, and reduce the GDP residence time within protein, enhancing the GDP/GTP exchange rate and Rac1 activity. This broadens our knowledge of the role of cancer-associated mutations on small GTPases mechanism supplying valuable information for future drug discovery efforts targeting specific Rac1 isoforms.
Collapse
Affiliation(s)
- Angela Parise
- Consiglio Nazionale delle ricerche (CNR)‐IOM c/o International School for Advanced Studies (SISSA/ISAS)TriesteItaly
| | - Alessandra Magistrato
- Consiglio Nazionale delle ricerche (CNR)‐IOM c/o International School for Advanced Studies (SISSA/ISAS)TriesteItaly
| |
Collapse
|
2
|
Karkali K, Pastor-Pareja JC, Martin-Blanco E. JNK signaling and integrins cooperate to maintain cell adhesion during epithelial fusion in Drosophila. Front Cell Dev Biol 2024; 11:1034484. [PMID: 38264353 PMCID: PMC10803605 DOI: 10.3389/fcell.2023.1034484] [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: 09/01/2022] [Accepted: 12/13/2023] [Indexed: 01/25/2024] Open
Abstract
The fusion of epithelial sheets is an essential and conserved morphogenetic event that requires the maintenance of tissue continuity. This is secured by membrane-bound or diffusible signals that instruct the epithelial cells, in a coordinated fashion, to change shapes and adhesive properties and when, how and where to move. Here we show that during Dorsal Closure (DC) in Drosophila, the Jun kinase (JNK) signaling pathway modulates integrins expression and ensures tissue endurance. An excess of JNK activity, as an outcome of a failure in the negative feedback implemented by the dual-specificity phosphatase Puckered (Puc), promotes the loss of integrins [the ß-subunit Myospheroid (Mys)] and amnioserosa detachment. Likewise, integrins signal back to the pathway to regulate the duration and strength of JNK activity. Mys is necessary for the regulation of JNK activity levels and in its absence, puc expression is downregulated and JNK activity increases.
Collapse
Affiliation(s)
- Katerina Karkali
- Instituto de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| | - Jose Carlos Pastor-Pareja
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (IN-CSIC), Alicante, Spain
| | - Enrique Martin-Blanco
- Instituto de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| |
Collapse
|
3
|
Faralli JA, Filla MS, Peters DM. Role of integrins in the development of fibrosis in the trabecular meshwork. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1274797. [PMID: 38983065 PMCID: PMC11182094 DOI: 10.3389/fopht.2023.1274797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/09/2023] [Indexed: 07/11/2024]
Abstract
Primary open angle glaucoma (POAG) is a progressive and chronic disease exhibiting many of the features of fibrosis. The extracellular matrix (ECM) in the trabecular meshwork (TM) undergoes extensive remodeling and enhanced rigidity, resembling fibrotic changes. In addition, there are changes associated with myofibroblast activation and cell contractility that further drives tissue fibrosis and stiffening. This review discusses what is known about the integrins in the TM and their involvement in fibrotic processes.
Collapse
Affiliation(s)
- Jennifer A. Faralli
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Mark S. Filla
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Donna M. Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Ophthalmology & Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
4
|
Li S, Sampson C, Liu C, Piao HL, Liu HX. Integrin signaling in cancer: bidirectional mechanisms and therapeutic opportunities. Cell Commun Signal 2023; 21:266. [PMID: 37770930 PMCID: PMC10537162 DOI: 10.1186/s12964-023-01264-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
Integrins are transmembrane receptors that possess distinct ligand-binding specificities in the extracellular domain and signaling properties in the cytoplasmic domain. While most integrins have a short cytoplasmic tail, integrin β4 has a long cytoplasmic tail that can indirectly interact with the actin cytoskeleton. Additionally, 'inside-out' signals can induce integrins to adopt a high-affinity extended conformation for their appropriate ligands. These properties enable integrins to transmit bidirectional cellular signals, making it a critical regulator of various biological processes.Integrin expression and function are tightly linked to various aspects of tumor progression, including initiation, angiogenesis, cell motility, invasion, and metastasis. Certain integrins have been shown to drive tumorigenesis or amplify oncogenic signals by interacting with corresponding receptors, while others have marginal or even suppressive effects. Additionally, different α/β subtypes of integrins can exhibit opposite effects. Integrin-mediated signaling pathways including Ras- and Rho-GTPase, TGFβ, Hippo, Wnt, Notch, and sonic hedgehog (Shh) are involved in various stages of tumorigenesis. Therefore, understanding the complex regulatory mechanisms and molecular specificities of integrins are crucial to delaying cancer progression and suppressing tumorigenesis. Furthermore, the development of integrin-based therapeutics for cancer are of great importance.This review provides an overview of integrin-dependent bidirectional signaling mechanisms in cancer that can either support or oppose tumorigenesis by interacting with various signaling pathways. Finally, we focus on the future opportunities for emergent therapeutics based on integrin agonists. Video Abstract.
Collapse
Affiliation(s)
- Siyi Li
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Chibuzo Sampson
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Changhao Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Hai-Long Piao
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
5
|
Maldonado H, Leyton L. CSK-mediated signalling by integrins in cancer. Front Cell Dev Biol 2023; 11:1214787. [PMID: 37519303 PMCID: PMC10382208 DOI: 10.3389/fcell.2023.1214787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Cancer progression and metastasis are processes heavily controlled by the integrin receptor family. Integrins are cell adhesion molecules that constitute the central components of mechanosensing complexes called focal adhesions, which connect the extracellular environment with the cell interior. Focal adhesions act as key players in cancer progression by regulating biological processes, such as cell migration, invasion, proliferation, and survival. Src family kinases (SFKs) can interplay with integrins and their downstream effectors. SFKs also integrate extracellular cues sensed by integrins and growth factor receptors (GFR), transducing them to coordinate metastasis and cell survival in cancer. The non-receptor tyrosine kinase CSK is a well-known SFK member that suppresses SFK activity by phosphorylating its specific negative regulatory loop (C-terminal Y527 residue). Consequently, CSK may play a pivotal role in tumour progression and suppression by inhibiting SFK oncogenic effects in several cancer types. Remarkably, CSK can localise near focal adhesions when SFKs are activated and even interact with focal adhesion components, such as phosphorylated FAK and Paxillin, among others, suggesting that CSK may regulate focal adhesion dynamics and structure. Even though SFK oncogenic signalling has been extensively described before, the specific role of CSK and its crosstalk with integrins in cancer progression, for example, in mechanosensing, remain veiled. Here, we review how CSK, by regulating SFKs, can regulate integrin signalling, and focus on recent discoveries of mechanotransduction. We additionally examine the cross talk of integrins and GFR as well as the membrane availability of these receptors in cancer. We also explore new pharmaceutical approaches to these signalling pathways and analyse them as future therapeutic targets.
Collapse
Affiliation(s)
- Horacio Maldonado
- Receptor Dynamics in Cancer Laboratory, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Xie N, Xiao C, Shu Q, Cheng B, Wang Z, Xue R, Wen Z, Wang J, Shi H, Fan D, Liu N, Xu F. Cell response to mechanical microenvironment cues via Rho signaling: From mechanobiology to mechanomedicine. Acta Biomater 2023; 159:1-20. [PMID: 36717048 DOI: 10.1016/j.actbio.2023.01.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/30/2023]
Abstract
Mechanical cues in the cell microenvironment such as those from extracellular matrix properties, stretching, compression and shear stress, play a critical role in maintaining homeostasis. Upon sensing mechanical stimuli, cells can translate these external forces into intracellular biochemical signals to regulate their cellular behaviors, but the specific mechanisms of mechanotransduction at the molecular level remain elusive. As a subfamily of the Ras superfamily, Rho GTPases have been recognized as key intracellular mechanotransduction mediators that can regulate multiple cell activities such as proliferation, migration and differentiation as well as biological processes such as cytoskeletal dynamics, metabolism, and organ development. However, the upstream mechanosensors for Rho proteins and downstream effectors that respond to Rho signal activation have not been well illustrated. Moreover, Rho-mediated mechanical signals in previous studies are highly context-dependent. In this review, we systematically summarize the types of mechanical cues in the cell microenvironment and provide recent advances on the roles of the Rho-based mechanotransduction in various cell activities, physiological processes and diseases. Comprehensive insights into the mechanical roles of Rho GTPase partners would open a new paradigm of mechanomedicine for a variety of diseases. STATEMENT OF SIGNIFICANCE: In this review, we highlight the critical role of Rho GTPases as signal mediators to respond to physical cues in microenvironment. This article will add a distinct contribution to this set of knowledge by intensively addressing the relationship between Rho signaling and mechanobiology/mechanotransduction/mechanomedcine. This topic has not been discussed by the journal, nor has it yet been developed by the field. The comprehensive picture that will develop, from molecular mechanisms and engineering methods to disease treatment strategies, represents an important and distinct contribution to the field. We hope that this review would help researchers in various fields, especially clinicians, oncologists and bioengineers, who study Rho signal pathway and mechanobiology/mechanotransduction, understand the critical role of Rho GTPase in mechanotransduction.
Collapse
Affiliation(s)
- Ning Xie
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Cailan Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Qiuai Shu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ziwei Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Runxin Xue
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Zhang Wen
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Jinhai Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Haitao Shi
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an Shaanxi 710049, China.
| | - Na Liu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
7
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
8
|
McCourt JL, Stearns-Reider KM, Mamsa H, Kannan P, Afsharinia MH, Shu C, Gibbs EM, Shin KM, Kurmangaliyev YZ, Schmitt LR, Hansen KC, Crosbie RH. Multi-omics analysis of sarcospan overexpression in mdx skeletal muscle reveals compensatory remodeling of cytoskeleton-matrix interactions that promote mechanotransduction pathways. Skelet Muscle 2023; 13:1. [PMID: 36609344 PMCID: PMC9817407 DOI: 10.1186/s13395-022-00311-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The dystrophin-glycoprotein complex (DGC) is a critical adhesion complex of the muscle cell membrane, providing a mechanical link between the extracellular matrix (ECM) and the cortical cytoskeleton that stabilizes the sarcolemma during repeated muscle contractions. One integral component of the DGC is the transmembrane protein, sarcospan (SSPN). Overexpression of SSPN in the skeletal muscle of mdx mice (murine model of DMD) restores muscle fiber attachment to the ECM in part through an associated increase in utrophin and integrin adhesion complexes at the cell membrane, protecting the muscle from contraction-induced injury. In this study, we utilized transcriptomic and ECM protein-optimized proteomics data sets from wild-type, mdx, and mdx transgenic (mdxTG) skeletal muscle tissues to identify pathways and proteins driving the compensatory action of SSPN overexpression. METHODS The tibialis anterior and quadriceps muscles were isolated from wild-type, mdx, and mdxTG mice and subjected to bulk RNA-Seq and global proteomics analysis using methods to enhance capture of ECM proteins. Data sets were further analyzed through the ingenuity pathway analysis (QIAGEN) and integrative gene set enrichment to identify candidate networks, signaling pathways, and upstream regulators. RESULTS Through our multi-omics approach, we identified 3 classes of differentially expressed genes and proteins in mdxTG muscle, including those that were (1) unrestored (significantly different from wild type, but not from mdx), (2) restored (significantly different from mdx, but not from wild type), and (3) compensatory (significantly different from both wild type and mdx). We identified signaling pathways that may contribute to the rescue phenotype, most notably cytoskeleton and ECM organization pathways. ECM-optimized proteomics revealed an increased abundance of collagens II, V, and XI, along with β-spectrin in mdxTG samples. Using ingenuity pathway analysis, we identified upstream regulators that are computationally predicted to drive compensatory changes, revealing a possible mechanism of SSPN rescue through a rewiring of cell-ECM bidirectional communication. We found that SSPN overexpression results in upregulation of key signaling molecules associated with regulation of cytoskeleton organization and mechanotransduction, including Yap1, Sox9, Rho, RAC, and Wnt. CONCLUSIONS Our findings indicate that SSPN overexpression rescues dystrophin deficiency partially through mechanotransduction signaling cascades mediated through components of the ECM and the cortical cytoskeleton.
Collapse
Affiliation(s)
- Jackie L McCourt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Kristen M Stearns-Reider
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
- Department of Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Hafsa Mamsa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Pranav Kannan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | | | - Cynthia Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Elizabeth M Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Kara M Shin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, Howard Hughes Medical Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, CO, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, CO, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Alshehri S, Pavlovič T, Farsinejad S, Behboodi P, Quan L, Centeno D, Kung D, Rezler M, Lee W, Jasiński P, Dziabaszewska E, Nowak-Markwitz E, Kalyon D, Zaborowski MP, Iwanicki M. Extracellular Matrix Modulates Outgrowth Dynamics in Ovarian Cancer. Adv Biol (Weinh) 2022; 6:e2200197. [PMID: 36084257 PMCID: PMC9772079 DOI: 10.1002/adbi.202200197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/18/2022] [Indexed: 01/28/2023]
Abstract
Ovarian carcinoma (OC) forms outgrowths that extend from the outer surface of an afflicted organ into the peritoneum. OC outgrowth formation is poorly understood due to the limited availability of cell culture models examining the behavior of cells that form outgrowths. Prompted by immunochemical evaluation of extracellular matrix (ECM) components in human tissues, laminin and collagen-rich ECM-reconstituted cell culture models amenable to studies of cell clusters that can form outgrowths are developed. It is demonstrated that ECM promotes outgrowth formation in fallopian tube non-ciliated epithelial cells (FNE) expressing mutant p53 and various OC cell lines. Outgrowths are initiated by cells that underwent outward translocation and retained the ability to intercalate into mesothelial cell monolayers. Electron microscopy, optical coherence tomography, and small amplitude oscillatory shear experiments reveal that increased ECM levels led to increased fibrous network thickness and high shear elasticity of the microenvironment. These physical characteristics are associated with outgrowth suppression. The low ECM microenvironment mimicks the viscoelasticity of malignant peritoneal fluid (ascites) and supports cell proliferation, cell translocation, and outgrowth formation. These results highlight the importance of the ECM microenvironment in modulating OC growth and can provide additional insights into the mode of dissemination of primary and recurrent ovarian tumors.
Collapse
Affiliation(s)
- Sarah Alshehri
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Tonja Pavlovič
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Sadaf Farsinejad
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Panteha Behboodi
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Li Quan
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Daniel Centeno
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Douglas Kung
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Marta Rezler
- Poznań University of Medical Sciences, Collegium Maius, Fredry 10, Poznań, 61-701, Poland
| | - Woo Lee
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Piotr Jasiński
- Department of Pathology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Elżbieta Dziabaszewska
- Department of Pathology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Ewa Nowak-Markwitz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Dilhan Kalyon
- Department of Chemical Engineering, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| | - Mikołaj P Zaborowski
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Poznań University of Medical Sciences, Polna 33, Poznań, 60-535, Poland
| | - Marcin Iwanicki
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, 1 Castle Point Terrace, Hoboken, NJ, 07030, USA
| |
Collapse
|
10
|
Chen PC, Feng XQ, Li B. Unified multiscale theory of cellular mechanical adaptations to substrate stiffness. Biophys J 2022; 121:3474-3485. [PMID: 35978549 PMCID: PMC9515123 DOI: 10.1016/j.bpj.2022.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 11/19/2022] Open
Abstract
Rigidity of the extracellular matrix markedly regulates many cellular processes. However, how cells detect and respond to matrix rigidity remains incompletely understood. Here, we propose a unified two-dimensional multiscale framework accounting for the chemomechanical feedback to explore the interrelated cellular mechanosensing, polarization, and migration, which constitute the dynamic cascade in cellular response to matrix stiffness but are often modeled separately in previous theories. By combining integrin dynamics and intracellular force transduction, we show that substrate stiffness can act as a switch to activate or deactivate cell polarization. Our theory quantitatively reproduces rich stiffness-dependent cellular dynamics, including spreading, polarity selection, migration pattern, durotaxis, and even negative durotaxis, reported in a wide spectrum of cell types, and reconciles some inconsistent experimental observations. We find that a specific bipolarized mode can determine the optimal substrate stiffness, which enables the fastest cell migration rather than the largest traction forces that cells apply on the substrate. We identify that such a mechanical adaptation stems from the force balance across the whole cell. These findings could yield universal insights into various stiffness-mediated cellular processes within the context of tissue morphogenesis, wound healing, and cancer invasion.
Collapse
Affiliation(s)
- Peng-Cheng Chen
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
11
|
Ahangar P, Strudwick XL, Cowin AJ. Wound Healing from an Actin Cytoskeletal Perspective. Cold Spring Harb Perspect Biol 2022; 14:a041235. [PMID: 35074864 PMCID: PMC9341468 DOI: 10.1101/cshperspect.a041235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing requires a complex cascade of highly controlled and conserved cellular and molecular processes. These involve numerous cell types and extracellular matrix molecules regulated by the actin cytoskeleton. This microscopic network of filaments is present within the cytoplasm of all cells and provides the shape and mechanical support required for cell movement and proliferation. Here, an overview of the processes of wound healing are described from the perspective of the cell in relation to the actin cytoskeleton. Key points of discussion include the role of actin, its binding proteins, signaling pathways, and events that play significant roles in the phases of wound healing. The identification of cytoskeletal targets that can be used to manipulate and improve wound healing is included as an emerging area of focus that may inform future therapeutic approaches to improve healing of complex wounds.
Collapse
Affiliation(s)
- Parinaz Ahangar
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Allison J Cowin
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| |
Collapse
|
12
|
Sharma V, Letson J, Furuta S. Fibrous stroma: Driver and passenger in cancer development. Sci Signal 2022; 15:eabg3449. [PMID: 35258999 DOI: 10.1126/scisignal.abg3449] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cumulative evidence shows that fibrogenic stroma and stiff extracellular matrix (ECM) not only result from tumor growth but also play pivotal roles in cellular transformation and tumor initiation. This emerging concept may largely account for the increased cancer risk associated with environmental fibrogenic agents, such as asbestos and silica, and with chronic conditions that are fibrogenic, such as obesity and diabetes. It may also contribute to poor outcomes in patients treated with certain chemotherapeutics that can promote fibrosis, such as bleomycin and methotrexate. Although the mechanistic details of this phenomenon are still being unraveled, we provide an overview of the experimental evidence linking fibrogenic stroma and tumor initiation. In this Review, we will summarize the causes and consequences of fibrous stroma and how this stromal cue is transmitted to the nuclei of parenchymal cells through a physical continuum from the ECM to chromatin, as well as ECM-dependent biochemical signaling that contributes to cellular transformation.
Collapse
Affiliation(s)
- Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| |
Collapse
|
13
|
Pan L, Bai P, Weng X, Liu J, Chen Y, Chen S, Ma X, Hu K, Sun A, Ge J. Legumain Is an Endogenous Modulator of Integrin αvβ3 Triggering Vascular Degeneration, Dissection, and Rupture. Circulation 2022; 145:659-674. [PMID: 35100526 DOI: 10.1161/circulationaha.121.056640] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND The development of thoracic aortic dissection (TAD) is closely related to extracellular matrix degradation and vascular smooth muscle cell (VSMC) transformation from contractile to synthetic type. LGMN (legumain) degrades extracellular matrix components directly or by activating downstream signals. The role of LGMN in VSMC differentiation and the occurrence of TAD remains elusive. METHODS Microarray datasets concerning vascular dissection or aneurysm were downloaded from the Gene Expression Omnibus database to screen differentially expressed genes. Four-week-old male Lgmn knockout mice (Lgmn-/-), macrophage-specific Lgmn knockout mice (LgmnF/F;LysMCre), and RR-11a-treated C57BL/6 mice were given BAPN (β-aminopropionitrile monofumarate; 1 g/kg/d) in drinking water for 4 weeks for TAD modeling. RNA sequencing analysis was performed to recapitulate transcriptome profile changes. Cell interaction was examined in macrophage and VSMC coculture system. The reciprocity of macrophage-derived LGMN with integrin αvβ3 in VSMCs was tested by coimmunoprecipitation assay and colocalization analyses. RESULTS Microarray datasets from the Gene Expression Omnibus database indicated upregulated LGMN in aorta from patients with TAD and mice with angiotensin II-induced AAA. Elevated LGMN was evidenced in aorta and sera from patients with TAD and mice with BAPN-induced TAD. BAPN-induced TAD progression was significantly ameliorated in Lgmn-deficient or inhibited mice. Macrophage-specific deletion of Lgmn alleviated BAPN-induced extracellular matrix degradation. Unbiased profiler polymerase chain reaction array and Gene Ontology analysis displayed that LGMN regulated VSMC phenotype transformation. Macrophage-specific deletion of Lgmn ameliorated VSMC phenotypic switch in BAPN-treated mice. Macrophage-derived LGMN inhibited VSMC differentiation in vitro as assessed by macrophages and the VSMC coculture system. Macrophage-derived LGMN bound to integrin αvβ3 in VSMCs and blocked integrin αvβ3, thereby attenuating Rho GTPase activation, downregulating VSMC differentiation markers and eventually exacerbating TAD development. ROCK (Rho kinase) inhibitor Y-27632 reversed the protective role of LGMN depletion in vascular dissection. CONCLUSIONS LGMN signaling may be a novel target for the prevention and treatment of TAD.
Collapse
Affiliation(s)
- Lihong Pan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Peiyuan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Xinyu Weng
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Jin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Yingjie Chen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson (Y.C.)
| | - Siqin Chen
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Xiurui Ma
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.)
| | - Aijun Sun
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China (L.P., S.C., A.S., J.G.).,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China (P.B., X.W., J.L., X.M., K.H., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.).,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China (L.P., P.B., X.W., J.L., S.C., X.M., A.S., J.G.)
| |
Collapse
|
14
|
Mierke CT. Viscoelasticity, Like Forces, Plays a Role in Mechanotransduction. Front Cell Dev Biol 2022; 10:789841. [PMID: 35223831 PMCID: PMC8864183 DOI: 10.3389/fcell.2022.789841] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Viscoelasticity and its alteration in time and space has turned out to act as a key element in fundamental biological processes in living systems, such as morphogenesis and motility. Based on experimental and theoretical findings it can be proposed that viscoelasticity of cells, spheroids and tissues seems to be a collective characteristic that demands macromolecular, intracellular component and intercellular interactions. A major challenge is to couple the alterations in the macroscopic structural or material characteristics of cells, spheroids and tissues, such as cell and tissue phase transitions, to the microscopic interferences of their elements. Therefore, the biophysical technologies need to be improved, advanced and connected to classical biological assays. In this review, the viscoelastic nature of cytoskeletal, extracellular and cellular networks is presented and discussed. Viscoelasticity is conceptualized as a major contributor to cell migration and invasion and it is discussed whether it can serve as a biomarker for the cells' migratory capacity in several biological contexts. It can be hypothesized that the statistical mechanics of intra- and extracellular networks may be applied in the future as a powerful tool to explore quantitatively the biomechanical foundation of viscoelasticity over a broad range of time and length scales. Finally, the importance of the cellular viscoelasticity is illustrated in identifying and characterizing multiple disorders, such as cancer, tissue injuries, acute or chronic inflammations or fibrotic diseases.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
15
|
Wirth F, Huck K, Lubosch A, Zoeller C, Ghura H, Porubsky S, Nakchbandi IA. Cdc42 in osterix-expressing cells alters osteoblast behavior and myeloid lineage commitment. Bone 2021; 153:116150. [PMID: 34400384 DOI: 10.1016/j.bone.2021.116150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023]
Abstract
Osteoblasts are not only responsible for bone formation. They also support hematopoiesis. This requires responding to cues originating from several signaling pathways, a task performed by Rho GTPases. We therefore examined several transgenic mouse models and used inhibitors of Cdc42 in vitro. Deletion of Cdc42 in vivo using the Osterix promoter suppressed osteoblast function, while its deletion in differentiating osteoblasts using the Collagen-α1(I) promoter decreased osteoblast numbers. In both cases, bone mineral density diminished confirming the importance of Cdc42. Evaluation of hematopoiesis revealed that deletion of Cdc42 using the Osterix, but not the Collagen-α1(I) promoter increased the common myeloid progenitors (CMPs) in the bone marrow as well as the erythrocytes and the thrombocytes/platelets in peripheral blood. Causality between Cdc42 loss in early osteoblasts and increased myelopoiesis was confirmed in vitro. Work in vitro supported the conclusion that interleukin-4 mediated the increase in myelopoiesis. Thus, Cdc42 is required for healthy bone through regulation of bone formation in Osterix-expressing osteoblasts and the number of osteoblasts in differentiating osteoblasts. In addition, its expression in early osteoblasts/stromal cells modulates myelopoiesis. This highlights the importance of osteoblasts in regulating hematopoiesis.
Collapse
Affiliation(s)
- Franziska Wirth
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Katrin Huck
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Alexander Lubosch
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Caren Zoeller
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Hiba Ghura
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Stefan Porubsky
- Institute of Pathology, University of Mainz, 55131 Mainz, Germany
| | - Inaam A Nakchbandi
- Institute of Immunology, University of Heidelberg, 69120 Heidelberg, Germany; Max-Planck Institute for Medical Research, 69120 Heidelberg, Germany; Max-Planck Institute for Biochemistry, 82152 Martinsried, Germany.
| |
Collapse
|
16
|
Kang N, Matsui TS, Liu S, Deguchi S. ARHGAP4-SEPT2-SEPT9 complex enables both up- and down-modulation of integrin-mediated focal adhesions, cell migration, and invasion. Mol Biol Cell 2021; 32:ar28. [PMID: 34524873 PMCID: PMC8693978 DOI: 10.1091/mbc.e21-01-0010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The Rho family of GTPases are inactivated in a cell context–dependent manner by Rho-GTPase-activating proteins (Rho-GAPs), but their signaling mechanisms are poorly understood. Here we demonstrate that ARHGAP4, one of the Rho-GAPs, forms a complex with SEPT2 and SEPT9 via its Rho-GAP domain and SH3 domain to enable both up- and down-modulation of integrin-mediated focal adhesions (FAs). We show that silencing ARHGAP4 and overexpressing its two mutually independent upstream regulators, SEPT2 and SEPT9, all induce reorganization of FAs to newly express Integrin Beta 1 and also enhance both cell migration and invasion. Interestingly, even if these cell migration/invasion–associated phenotypic changes are induced upon perturbations to the complex, it does not necessarily cause enhanced clustering of FAs. Instead, its extent depends on whether the microenvironment contains ligands suitable for the up-regulated Integrin Beta 1. These results provide novel insights into cell migration, invasion, and microenvironment-dependent phenotypic changes regulated by the newly identified complex.
Collapse
Affiliation(s)
- Na Kang
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka 560-8531, Japan
| | - Tsubasa S Matsui
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka 560-8531, Japan
| | - Shiyou Liu
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka 560-8531, Japan
| | - Shinji Deguchi
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka 560-8531, Japan
| |
Collapse
|
17
|
Cytoskeleton Response to Ionizing Radiation: A Brief Review on Adhesion and Migration Effects. Biomedicines 2021; 9:biomedicines9091102. [PMID: 34572287 PMCID: PMC8465203 DOI: 10.3390/biomedicines9091102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
The cytoskeleton is involved in several biological processes, including adhesion, motility, and intracellular transport. Alterations in the cytoskeletal components (actin filaments, intermediate filaments, and microtubules) are strictly correlated to several diseases, such as cancer. Furthermore, alterations in the cytoskeletal structure can lead to anomalies in cells’ properties and increase their invasiveness. This review aims to analyse several studies which have examined the alteration of the cell cytoskeleton induced by ionizing radiations. In particular, the radiation effects on the actin cytoskeleton, cell adhesion, and migration have been considered to gain a deeper knowledge of the biophysical properties of the cell. In fact, the results found in the analysed works can not only aid in developing new diagnostic tools but also improve the current cancer treatments.
Collapse
|
18
|
Li HL, Li QY, Jin MJ, Lu CF, Mu ZY, Xu WY, Song J, Zhang Y, Zhang SY. A review: hippo signaling pathway promotes tumor invasion and metastasis by regulating target gene expression. J Cancer Res Clin Oncol 2021; 147:1569-1585. [PMID: 33864521 DOI: 10.1007/s00432-021-03604-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/16/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The Hippo pathway is widely considered to inhibit cell growth and play an important role in regulating the size of organs. However, recent studies have shown that abnormal regulation of the Hippo pathway can also affect tumor invasion and metastasis. Therefore, finding out how the Hippo pathway promotes tumor development by regulating the expression of target genes provides new ideas for future research on targeted drugs that inhibit tumor progression. METHODS PubMed, Embase, Web of Science, and the Cochrane Library were systematically searched. RESULTS The search strategy identified 1892 hits and 196 publications were finally included in this review. As the core molecule of the Hippo pathway, YAP/TAZ are usually highly expressed in tumors that undergo invasion and migration and are accompanied by abnormally strong nuclear metastasis. Through its interaction with nuclear transcription factors TEADs, it directly or indirectly regulates and the expressions of target genes related to tumor metastasis and invasion. These target genes can induce the formation of invasive pseudopodia in tumor cells, reduce intercellular adhesion, degrade extracellular matrix (ECM), and cause epithelial-mesenchymal transition (EMT), or indirectly promote through other signaling pathways, such as mitogen-activated protein kinases (MAPK), TGF/Smad, etc, which facilitate the invasion and metastasis of tumors. CONCLUSION This article mainly introduces the research progress of YAP/TAZ which are the core molecules of the Hippo pathway regulating related target genes to promote tumor invasion and metastasis. Focus on the target genes that affect tumor invasion and metastasis, providing the possibility for the selection of clinical drug treatment targets, to provide some help for a more in-depth study of tumor invasion and migration mechanism and the development of clinical drugs.
Collapse
Affiliation(s)
- Hong-Li Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Qian-Yu Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Min-Jie Jin
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chao-Fan Lu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Zhao-Yang Mu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wei-Yi Xu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China. .,School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Institute of Drug Discovery and Development, Zhengzhou, 450001, China.
| | - Yan Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China. .,School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Institute of Drug Discovery and Development, Zhengzhou, 450001, China. .,Zhengzhou University, Henan Institute of Advanced Technology, Zhengzhou, 450001, China.
| |
Collapse
|
19
|
Safka Brozkova D, Stojkovic T, Haberlová J, Mazanec R, Windhager R, Fernandes Rosenegger P, Hacker S, Züchner S, Kochański A, Leonard-Louis S, Francou B, Latour P, Senderek J, Seeman P, Auer-Grumbach M. Demyelinating Charcot-Marie-Tooth neuropathy associated with FBLN5 mutations. Eur J Neurol 2020; 27:2568-2574. [PMID: 32757322 DOI: 10.1111/ene.14463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/29/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Charcot-Marie-Tooth disease type 1 (CMT1) is a group of autosomal dominantly inherited demyelinating sensorimotor neuropathies. Symptoms usually start in the first to second decade and include distal muscle weakness and wasting, sensory disturbances and foot deformities. The most frequent cause is a duplication of PMP22 whilst point mutations in PMP22 and other genes are rare causes. Recently, FBLN5 mutations have been reported in CMT1 families. METHODS Individuals with FBLN5-associated CMT1 were compiled from clinical and research genetic testing laboratories. Clinical data were extracted from medical records or obtained during patients' visits at our centres or primary care sites. RESULTS Nineteen CMT1 families containing 38 carriers of three different FBLN5 missense variants were identified and a mutational hotspot at c.1117C>T (p.Arg373Cys) was confirmed. Compared to patients with the common PMP22 duplication, individuals with FBLN5 variants had a later age of diagnosis (third to fifth decade) and less severely reduced motor median nerve conduction velocities (around 31 m/s). The most frequent clinical presentations were prominent sensory disturbances and painful sensations, often as initial symptom and pronounced in the upper limbs, contrasting with rather mild to moderate motor deficits. CONCLUSIONS Our study confirms the relevance of FBLN5 mutations in CMT1. It is proposed to include FBLN5 in the genetic work-up of individuals suspected with CMT1, particularly when diagnosis is established beyond the first and second decade and comparably moderate motor deficits contrast with early and marked sensory involvement. FBLN5-associated CMT1 has a recognizable clinical phenotype and should be referred to as CMT1H according to the current classification scheme.
Collapse
Affiliation(s)
- D Safka Brozkova
- DNA Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - T Stojkovic
- Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Institut de Myologie, APHP, G-H Pitié-Salpêtrière, Paris, France
| | - J Haberlová
- DNA Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - R Mazanec
- Department of Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - R Windhager
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - P Fernandes Rosenegger
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - S Hacker
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - S Züchner
- Dr John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A Kochański
- Neuromuscular Unit, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - S Leonard-Louis
- Unité de Pathologie Neuromusculaire, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, APHP, G-H Pitié-Salpêtrière, Paris, France
| | - B Francou
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, APHP, Hôpital Kremlin-Bicêtre, Paris, France
| | - P Latour
- Service de Biochimie et Biologie Moléculaire Grand Est, CHU de Lyon, GH Est, Bron, France
| | - J Senderek
- Department of Neurology, Friedrich-Baur-Institute, LMU Munich, Munich, Germany
| | - P Seeman
- DNA Laboratory, Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - M Auer-Grumbach
- Department of Orthopaedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Afrin S, Islam MS, Patzkowsky K, Malik M, Catherino WH, Segars JH, Borahay MA. Simvastatin ameliorates altered mechanotransduction in uterine leiomyoma cells. Am J Obstet Gynecol 2020; 223:733.e1-733.e14. [PMID: 32417359 DOI: 10.1016/j.ajog.2020.05.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 04/18/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress β1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies. OBJECTIVE This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells. STUDY DESIGN This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 μM, or control) for 48 hours. Protein and mRNA levels of β1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction. RESULTS We found that simvastatin significantly reduced the protein expression of β1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in β1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1. CONCLUSION The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials.
Collapse
|
21
|
Reply to Liu: The disassembly of the actin cytoskeleton is an early event during NETosis. Proc Natl Acad Sci U S A 2020; 117:22655-22656. [PMID: 32943583 PMCID: PMC7502743 DOI: 10.1073/pnas.2015951117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
22
|
Nakano T, Uchiyama K, Ushiroda C, Kashiwagi S, Toyokawa Y, Mizushima K, Inoue K, Dohi O, Okayama T, Yoshida N, Katada K, Kamada K, Handa O, Ishikawa T, Takagi T, Konishi H, Naito Y, Itoh Y. Promotion of wound healing by acetate in murine colonic epithelial cell via c-Jun N-terminal kinase activation. J Gastroenterol Hepatol 2020; 35:1171-1179. [PMID: 31961456 DOI: 10.1111/jgh.14987] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 12/20/2019] [Accepted: 01/16/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND AIM Mucosal healing is an important clinical goal in patients with inflammatory bowel disease. Recently, short-chain fatty acids (SCFAs) have been reported to have multifaceted effects to host. However, the effects of SCFAs on wound healing in intestinal epithelial cells are unclear. In the present study, we investigated the effects of acetate, one of the major SCFAs, on the wound healing of murine colonic epithelial cells. METHODS Young adult mouse colonic epithelial cells were used to determine the effect of acetate using wound healing assay. Mitogen-activated protein kinase and Rho kinase inhibitor were used to elucidate intracellular signal of wound healing treated with acetate. Meanwhile, Rho activation assays were utilized to measure Rho activation levels. To assess in vivo effects, C57B6 mice with dextran sodium sulfate for 7 days were treated with enema administration of acetate for 7 days. Body weight, disease activity index, colon length, and mucosal break ratio in histology were examined. RESULTS Acetate enhanced wound healing and fluorescence intensity of actin stress fiber compared with control. These effects were canceled with pretreatment of c-Jun N-terminal kinase (JNK) inhibitor or Rho kinase inhibitor. Furthermore, JNK inhibitor reduced the activation of Rho induced by acetate. In the dextran sodium sulfate-induced colitis model, the mice with enema treatment of acetate significantly exhibited recovery. CONCLUSIONS In this study, we demonstrated that acetate promoted murine colonic epithelial cell wound healing via activation of JNK and Rho signaling pathways. These findings suggested that acetate could have applications as a therapeutic agent for patients with intestinal mucosal damage, such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Takahiro Nakano
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chihiro Ushiroda
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Saori Kashiwagi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuki Toyokawa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsura Mizushima
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Inoue
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Dohi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Okayama
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, North Center of Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naohisa Yoshida
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Katada
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Gastroenterology and Hepatology, North Center of Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Kamada
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Handa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Division of Gastroenterology, Department of Internal Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Takeshi Ishikawa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohisa Takagi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideyuki Konishi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
23
|
Li X, Tao Y, Wang X, Wang T, Liu J. Advanced glycosylation end products (AGEs) controls proliferation, invasion and permeability through orchestrating ARHGAP18/RhoA pathway in human umbilical vein endothelial cells. Glycoconj J 2020; 37:209-219. [PMID: 32016689 DOI: 10.1007/s10719-020-09908-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022]
Abstract
Diabetic vascular complications caused by endothelial dysfunction play an important role in the pathogenesis of diabetic foot. A well understanding of the role of endothelial dysfunction in diabetic foot vasculopathy will help to further reveal the pathogenesis of diabetic foot. This study aimed to assess whether the RhoA/ROCK signaling pathway is controlled by Rho GTPase-activating proteins (RhoGAP, ARHGAP) and advanced glycosylation end products (AGEs), and to clarify the roles of ARHGAP and AGEs in the RhoA/ROCK signaling pathway or the mechanism by which AGEs regulated RhoA. Real-time PCR was applied to detect gene expression. Manipulation of endothelial biological functions by ARHGAP18 and AGEs were studied via cell counting kit-8 (CCK-8), Western blot, transwell, FITC-Dextran and TEER permeability experiments. RhoA-specific inhibitor Y-27632 was used to silence the activity of RhoA. Dual Luciferase Reporter Assay, Western blot and ELISA assays were used to detect molecular mechanism of endothelial biological functions. In this study, we found that ARHGAP18 was negatively correlated with RhoA, and the expression of ARHGAP18 in human umbilical vein endothelial cells (HUVECs) was decreased with gradient-increased AGEs. Furthermore, AGEs and ARHGAP18 could orchestrate RhoA activity, then activate NF-κB signaling pathway, affect the structural and morphological of VE-cadherin and tight junction protein, and cause endothelial cell contraction, thereby increasing permeability of endothelial cells. In conclusion, AGEs and ARHGAP18 orchestrate cell proliferation, invasion and permeability by controlling the RhoA/ROCK signaling pathway, affecting NF-κB signaling pathway as well as the structure and morphology of VE-cadherin and tight junction protein, and regulating endothelial cell contraction.
Collapse
Affiliation(s)
- Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Yue Tao
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Xiaojun Wang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Tao Wang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China
| | - Jianjun Liu
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 9/F, Building 7, East Park Road No.1158, Qingpu District, Shanghai, 201700, People's Republic of China.
| |
Collapse
|
24
|
Thüroff F, Goychuk A, Reiter M, Frey E. Bridging the gap between single-cell migration and collective dynamics. eLife 2019; 8:e46842. [PMID: 31808744 PMCID: PMC6992385 DOI: 10.7554/elife.46842] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 12/06/2019] [Indexed: 11/13/2022] Open
Abstract
Motivated by the wealth of experimental data recently available, we present a cellular-automaton-based modeling framework focussing on high-level cell functions and their concerted effect on cellular migration patterns. Specifically, we formulate a coarse-grained description of cell polarity through self-regulated actin organization and its response to mechanical cues. Furthermore, we address the impact of cell adhesion on collective migration in cell cohorts. The model faithfully reproduces typical cell shapes and movements down to the level of single cells, yet allows for the efficient simulation of confluent tissues. In confined circular geometries, we find that specific properties of individual cells (polarizability; contractility) influence the emerging collective motion of small cell cohorts. Finally, we study the properties of expanding cellular monolayers (front morphology; stress and velocity distributions) at the level of extended tissues.
Collapse
Affiliation(s)
- Florian Thüroff
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Andriy Goychuk
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Matthias Reiter
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Erwin Frey
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| |
Collapse
|
25
|
Ayad NME, Kaushik S, Weaver VM. Tissue mechanics, an important regulator of development and disease. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180215. [PMID: 31431174 DOI: 10.1098/rstb.2018.0215] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A growing body of work describes how physical forces in and around cells affect their growth, proliferation, migration, function and differentiation into specialized types. How cells receive and respond biochemically to mechanical signals is a process termed mechanotransduction. Disease may arise if a disruption occurs within this mechanism of sensing and interpreting mechanics. Cancer, cardiovascular diseases and developmental defects, such as during the process of neural tube formation, are linked to changes in cell and tissue mechanics. A breakdown in normal tissue and cellular forces activates mechanosignalling pathways that affect their function and can promote disease progression. The recent advent of high-resolution techniques enables quantitative measurements of mechanical properties of the cell and its extracellular matrix, providing insight into how mechanotransduction is regulated. In this review, we will address the standard methods and new technologies available to properly measure mechanical properties, highlighting the challenges and limitations of probing different length-scales. We will focus on the unique environment present throughout the development and maintenance of the central nervous system and discuss cases where disease, such as brain cancer, arises in response to changes in the mechanical properties of the microenvironment that disrupt homeostasis. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Nadia M E Ayad
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Shelly Kaushik
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.,UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.,Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
Leiphart RJ, Chen D, Peredo AP, Loneker AE, Janmey PA. Mechanosensing at Cellular Interfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7509-7519. [PMID: 30346180 DOI: 10.1021/acs.langmuir.8b02841] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
At the plasma membrane interface, cells use various adhesions to sense their extracellular environment. These adhesions facilitate the transmission of mechanical signals that dictate cell behavior. This review discusses the mechanisms by which these mechanical signals are transduced through cell-matrix and cell-cell adhesions and how this mechanotransduction influences cell processes. Cell-matrix adhesions require the activation of and communication between various transmembrane protein complexes such as integrins. These links at the plasma membrane affect how a cell senses and responds to its matrix environment. Cells also communicate with each other through cell-cell adhesions, which further regulate cell behavior on a single- and multicellular scale. Coordination and competition between cell-cell and cell-matrix adhesions in multicellular aggregates can, to a significant extent, be modeled by differential adhesion analyses between the different interfaces even without knowing the details of cellular signaling. In addition, cell-matrix and cell-cell adhesions are connected by an intracellular cytoskeletal network that allows for direct communication between these distinct adhesions and activation of specific signaling pathways. Other membrane-embedded protein complexes, such as growth factor receptors and ion channels, play additional roles in mechanotransduction. Overall, these mechanoactive elements show the dynamic interplay between the cell, its matrix, and neighboring cells and how these relationships affect cellular function.
Collapse
Affiliation(s)
- Ryan J Leiphart
- Department of Bioengineering , University of Pennsylvania , 210 S 33rd St , Philadelphia , Pennsylvania 19104 , United States
- McKay Orthopedic Research Laboratory , University of Pennsylvania , Philadelphia , Pennsylvania , United States
| | - Dongning Chen
- Department of Bioengineering , University of Pennsylvania , 210 S 33rd St , Philadelphia , Pennsylvania 19104 , United States
- Center for Engineering Mechanobiology , University of Pennsylvania , Philadelphia , Pennsylvania , United States
| | - Ana P Peredo
- Department of Bioengineering , University of Pennsylvania , 210 S 33rd St , Philadelphia , Pennsylvania 19104 , United States
- McKay Orthopedic Research Laboratory , University of Pennsylvania , Philadelphia , Pennsylvania , United States
| | - Abigail E Loneker
- Department of Bioengineering , University of Pennsylvania , 210 S 33rd St , Philadelphia , Pennsylvania 19104 , United States
- Center for Engineering Mechanobiology , University of Pennsylvania , Philadelphia , Pennsylvania , United States
| | - Paul A Janmey
- Department of Bioengineering , University of Pennsylvania , 210 S 33rd St , Philadelphia , Pennsylvania 19104 , United States
- Institute for Medicine and Engineering, Department of Physiology , University of Pennsylvania , 3340 Smith Walk , Philadelphia , Pennsylvania 19104 , United States
- Center for Engineering Mechanobiology , University of Pennsylvania , Philadelphia , Pennsylvania , United States
| |
Collapse
|
27
|
Qu L, Pan C, He SM, Lang B, Gao GD, Wang XL, Wang Y. The Ras Superfamily of Small GTPases in Non-neoplastic Cerebral Diseases. Front Mol Neurosci 2019; 12:121. [PMID: 31213978 PMCID: PMC6555388 DOI: 10.3389/fnmol.2019.00121] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
The small GTPases from the Ras superfamily play crucial roles in basic cellular processes during practically the entire process of neurodevelopment, including neurogenesis, differentiation, gene expression, membrane and protein traffic, vesicular trafficking, and synaptic plasticity. Small GTPases are key signal transducing enzymes that link extracellular cues to the neuronal responses required for the construction of neuronal networks, as well as for synaptic function and plasticity. Different subfamilies of small GTPases have been linked to a number of non-neoplastic cerebral diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), intellectual disability, epilepsy, drug addiction, Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and a large number of idiopathic cerebral diseases. Here, we attempted to make a clearer illustration of the relationship between Ras superfamily GTPases and non-neoplastic cerebral diseases, as well as their roles in the neural system. In future studies, potential treatments for non-neoplastic cerebral diseases which are based on small GTPase related signaling pathways should be explored further. In this paper, we review all the available literature in support of this possibility.
Collapse
Affiliation(s)
- Liang Qu
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Chao Pan
- Beijing Institute of Biotechnology, Beijing, China
| | - Shi-Ming He
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China.,Department of Neurosurgery, Xi'an International Medical Center, Xi'an, China
| | - Bing Lang
- The School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
28
|
Barnawi R, Al-Khaldi S, Colak D, Tulbah A, Al-Tweigeri T, Fallatah M, Monies D, Ghebeh H, Al-Alwan M. β1 Integrin is essential for fascin-mediated breast cancer stem cell function and disease progression. Int J Cancer 2019; 145:830-841. [PMID: 30719702 PMCID: PMC6593770 DOI: 10.1002/ijc.32183] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 12/19/2018] [Accepted: 01/28/2019] [Indexed: 12/19/2022]
Abstract
Breast cancer remains the second cause of tumor‐related mortality in women worldwide mainly due to chemoresistance and metastasis. The chemoresistance and metastasis are attributed to a rare subpopulation with enriched stem‐like characteristics, thus called Cancer Stem Cells (CSCs). We have previously reported aberrant expression of the actin‐bundling protein (fascin) in breast cancer cells, which enhances their chemoresistance, metastasis and enriches CSC population. The intracellular mechanisms that link fascin with its downstream effectors are not fully elucidated. Here, loss and gain of function approaches in two different breast cancer models were used to understand how fascin promotes disease progression. Importantly, findings were aligned with expression data from actual breast cancer patients. Expression profiling of a large breast cancer dataset (TCGA, 530 patients) showed statistically significant correlation between fascin expression and a key adherence molecule, β1 integrin (ITGB1). In vitro manipulation of fascin expression in breast cancer cells exhibited its direct effect on ITGB1 expression. Fascin‐mediated regulation of ITGB1 was critical for several breast cancer cell functions including adhesion to different extracellular matrix, self‐renewability and chemoresistance. Importantly, there was a significant relationship between fascin and ITGB1 co‐expression and short disease‐free as well as overall survival in chemo‐treated breast cancer patients. This novel role of fascin effect on ITGB1 expression and its outcome on cell self‐renewability and chemoresistance strongly encourages for dual targeting of fascin‐ITGB1 axis as a therapeutic approach to halt breast cancer progression and eradicate it from the root. What's new? Residual cancer stem cells (CSCs) have the ability to regrow tumors and to metastasize to distant organs, resulting in disease relapse and increased cancer mortality. In breast cancer, CSC populations are enriched by aberrant expression of the actin‐bundling protein fascin, induction of which is also associated with chemoresistance and metastasis. In this study, fascin was found to upregulate β1 integrin (ITGB1) expression, an effect that proved critical to breast cancer cell adhesion and self‐renewal. Coexpression of fascin and ITGB1 was associated with decreased survival in chemotherapy‐treated breast cancer patients. The findings identify the fascin‐ITGB1 axis as a potential therapeutic target.
Collapse
Affiliation(s)
- Rayanah Barnawi
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Samiyah Al-Khaldi
- National Center for Stem Cells, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia
| | - Dilek Colak
- Department of Biostatistics, Epidemiology and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Asma Tulbah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Taher Al-Tweigeri
- Department of Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohannad Fallatah
- National Center for Stem Cells, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia
| | - Dorota Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hazem Ghebeh
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Bell S, Redmann AL, Terentjev EM. Universal Kinetics of the Onset of Cell Spreading on Substrates of Different Stiffness. Biophys J 2019; 116:551-559. [PMID: 30665696 PMCID: PMC6369430 DOI: 10.1016/j.bpj.2018.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 12/17/2018] [Accepted: 12/28/2018] [Indexed: 12/19/2022] Open
Abstract
When plated onto substrates, cell morphology and even stem-cell differentiation are influenced by the stiffness of their environment. Stiffer substrates give strongly spread (eventually polarized) cells with strong focal adhesions and stress fibers; very soft substrates give a less developed cytoskeleton and much lower cell spreading. The kinetics of this process of cell spreading is studied extensively, and important universal relationships are established on how the cell area grows with time. Here, we study the population dynamics of spreading cells, investigating the characteristic processes involved in the cell response to the substrate. We show that unlike the individual cell morphology, this population dynamics does not depend on the substrate stiffness. Instead, a strong activation temperature dependence is observed. Different cell lines on different substrates all have long-time statistics controlled by the thermal activation over a single energy barrier ΔG ≈ 18 kcal/mol, whereas the early-time kinetics follows a power law ∼t5. This implies that the rate of spreading depends on an internal process of adhesion complex assembly and activation; the operational complex must have five component proteins, and the last process in the sequence (which we believe is the activation of focal adhesion kinase) is controlled by the binding energy ΔG.
Collapse
Affiliation(s)
- Samuel Bell
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Anna-Lena Redmann
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Eugene M Terentjev
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
30
|
Park SH, Lee CW, Lee JH, Park JY, Roshandell M, Brennan CA, Choe KM. Requirement for and polarized localization of integrin proteins during Drosophila wound closure. Mol Biol Cell 2018; 29:2137-2147. [PMID: 29995573 PMCID: PMC6249799 DOI: 10.1091/mbc.e17-11-0635] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 06/19/2018] [Accepted: 07/05/2018] [Indexed: 01/06/2023] Open
Abstract
Wound reepithelialization is an evolutionarily conserved process in which skin cells migrate as sheets to heal the breach and is critical to prevent infection but impaired in chronic wounds. Integrin heterodimers mediate attachment between epithelia and underlying extracellular matrix and also act in large signaling complexes. The complexity of the mammalian wound environment and evident redundancy among integrins has impeded determination of their specific contributions to reepithelialization. Taking advantage of the genetic tools and smaller number of integrins in Drosophila, we undertook a systematic in vivo analysis of integrin requirements in the reepithelialization of skin wounds in the larva. We identify αPS2-βPS and αPS3-βPS as the crucial integrin dimers and talin as the only integrin adhesion component required for reepithelialization. The integrins rapidly accumulate in a JNK-dependent manner in a few rows of cells surrounding a wound. Intriguingly, the integrins localize to the distal margin in these cells, instead of the frontal or lamellipodial distribution expected for proteins providing traction and recruit nonmuscle myosin II to the same location. These findings indicate that signaling roles of integrins may be important for epithelial polarization around wounds and lay the groundwork for using Drosophila to better understand integrin contributions to reepithelialization.
Collapse
Affiliation(s)
- Si-Hyoung Park
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Chan-wool Lee
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Ji-Hyun Lee
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Jin Young Park
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| | - Mobina Roshandell
- Department of Biological Science, California State University, Fullerton, Fullerton, CA 92831
| | - Catherine A. Brennan
- Department of Biological Science, California State University, Fullerton, Fullerton, CA 92831
| | - Kwang-Min Choe
- Department of Systems Biology, Yonsei University, Seodaemun-gu, Seoul 03722, South Korea
| |
Collapse
|
31
|
Song S, Cong W, Zhou S, Shi Y, Dai W, Zhang H, Wang X, He B, Zhang Q. Small GTPases: Structure, biological function and its interaction with nanoparticles. Asian J Pharm Sci 2018; 14:30-39. [PMID: 32104436 PMCID: PMC7032109 DOI: 10.1016/j.ajps.2018.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022] Open
Abstract
Small GTPase is a kind of GTP-binding protein commonly found in eukaryotic cells. It plays an important role in cytoskeletal reorganization, cell polarity, cell cycle progression, gene expression and many other significant events in cells, such as the interaction with foreign particles. Therefore, it is of great scientific significance to understand the biological properties of small GTPases as well as the GTPase-nano interplay, since more and more nanomedicine are supposed to be used in biomedical field. However, there is no review in this aspect. This review summarizes the small GTPases in terms of the structure, biological function and its interaction with nanoparticles. We briefly introduced the various nanoparticles such as gold/silver nanoparticles, SWCNT, polymeric micelles and other nano delivery systems that interacted with different GTPases. These current nanoparticles exhibited different pharmacological effect modes and various target design concepts in the small GTPases study. This will help to elucidate the conclusion that the therapeutic strategy targeting small GTPases might be a new research direction. It is believed that the in-depth study on the functional mechanism of GTPases can provide insights for the design and study of nanomedicines.
Collapse
Affiliation(s)
- Siyang Song
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.,Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Wenshu Cong
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Shurong Zhou
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Yujie Shi
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Wenbing Dai
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Hua Zhang
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Xueqing Wang
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Bing He
- Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| | - Qiang Zhang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.,Peking University, No. 38, Xueyuan Road, Beijing 100191, China
| |
Collapse
|
32
|
Wang Z, Lv Z, Li C, Shao Y, Zhang W, Zhao X. An invertebrate β-integrin mediates coelomocyte phagocytosis via activation of septin2 and 7 but not septin10. Int J Biol Macromol 2018. [DOI: 10.1016/j.ijbiomac.2018.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Qiu Q, Zhang F, Wu J, Xu N, Liang M. Gingipains disrupt F-actin and cause osteoblast apoptosis via integrin β1. J Periodontal Res 2018; 53:762-776. [PMID: 29777544 DOI: 10.1111/jre.12563] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVE The aim of this study was to explore the cellular mechanisms underlying gingipain-caused changes in cell morphology and apoptosis of osteoblasts. MATERIAL AND METHODS Human calvarial osteoblasts and mouse osteoblasts MC3T3-E1 were treated with gingipain extracts from Porphyromonas gingivalis stain W83. Apoptosis was detected with annexin V and propidium iodide flow cytometry analysis or terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling staining. F-actin was determined by immunostaining. Western blotting was used to detect protein expression. Knocking down and overexpressing approaches were used to determine the role of integrin β1. RESULTS Osteoblasts exposed to gingipain extracts displayed increased apoptosis, accompanied by loss of F-actin integrity and cell shrinkage. The effects of gingipain extracts were abolished by the cysteine protease inhibitor N-tosyl-l-lysyl chloromethyl-ketone. Notably, gingipain extracts resulted in reduction of integrin β1, accompanied by diminished active RhoA whereas without effect on the total RhoA. Knockdown of integrin β1 resembled those seen in gingipain-treated osteoblasts. By contrast, the effects of gingipain extracts were abrogated by either overexpression of integrin β1 or presence of RhoA agonist CN03. CONCLUSION Gingipain-induced F-actin disruption and apoptosis are mediated by the degradation of integrin β1 and inhibition of RhoA activity, which account for osteoblast apoptosis.
Collapse
Affiliation(s)
- Q Qiu
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - F Zhang
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - J Wu
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - N Xu
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - M Liang
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
34
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
35
|
Abstract
Reactive astrogliosis occurs after central nervous system (CNS) injuries whereby resident astrocytes form rapid responses along a graded continuum. Following CNS lesions, naïve astrocytes are converted into reactive astrocytes and eventually into scar-forming astrocytes that block axon regeneration and neural repair. It has been known for decades that scarring development and its related extracellular matrix molecules interfere with regeneration of injured axons after CNS injury, but the cellular and molecular mechanisms for controlling astrocytic scar formation and maintenance are not well known. Recent use of various genetic tools has made tremendous progress in better understanding genesis of reactive astrogliosis. Especially, the latest experiments demonstrate environment-dependent plasticity of reactive astrogliosis because reactive astrocytes isolated from injured spinal cord form scarring astrocytes when transplanted into injured spinal cord, but revert in retrograde to naive astrocytes when transplanted into naive spinal cord. The interactions between upregulated type I collagen and its receptor integrin β1 and the N-cadherin-mediated cell adhesion appear to play major roles for local astrogliosis around the lesion. This review centers on the environment-dependent plasticity of reactive astrogliosis after spinal cord injury and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Fatima M Nathan
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
36
|
Castelnovo LF, Bonalume V, Melfi S, Ballabio M, Colleoni D, Magnaghi V. Schwann cell development, maturation and regeneration: a focus on classic and emerging intracellular signaling pathways. Neural Regen Res 2017; 12:1013-1023. [PMID: 28852375 PMCID: PMC5558472 DOI: 10.4103/1673-5374.211172] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development, maturation and regeneration of Schwann cells (SCs), the main glial cells of the peripheral nervous system, require the coordinate and complementary interaction among several factors, signals and intracellular pathways. These regulatory molecules consist of integrins, neuregulins, growth factors, hormones, neurotransmitters, as well as entire intracellular pathways including protein-kinase A, C, Akt, Erk/MAPK, Hippo, mTOR, etc. For instance, Hippo pathway is overall involved in proliferation, apoptosis, regeneration and organ size control, being crucial in cancer proliferation process. In SCs, Hippo is linked to merlin and YAP/TAZ signaling and it seems to respond to mechanic/physical challenges. Recently, among factors regulating SCs, also the signaling intermediates Src tyrosine kinase and focal adhesion kinase (FAK) proved relevant for SC fate, participating in the regulation of adhesion, motility, migration and in vitro myelination. In SCs, the factors Src and FAK are regulated by the neuroactive steroid allopregnanolone, thus corroborating the importance of this steroid in the control of SC maturation. In this review, we illustrate some old and novel signaling pathways modulating SC biology and functions during the different developmental, mature and regenerative states.
Collapse
Affiliation(s)
- Luca Franco Castelnovo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bonalume
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Simona Melfi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marinella Ballabio
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Deborah Colleoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
37
|
Kutys ML, Chen CS. Forces and mechanotransduction in 3D vascular biology. Curr Opin Cell Biol 2016; 42:73-79. [PMID: 27209346 DOI: 10.1016/j.ceb.2016.04.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 04/27/2016] [Indexed: 01/03/2023]
Abstract
The effects of hemodynamic and interstitial mechanical forces on endothelial biology in vivo have been appreciated for over half a century, regulating vessel network development, homeostatic function, and progression of vascular disease. Investigations using cultures of endothelial cells on two-dimensional (2D) substrates have elucidated important mechanisms by which microenvironmental stresses are sensed and transduced into chemical signaling responses. However recent studies in vivo and in three-dimensional (3D) in vitro models of vascular beds have enabled the investigation of forces and cellular behaviors previously not possible in traditional 2D culture systems. These studies support a developing paradigm that the 3D chemo-mechanical architecture of the vascular niche impacts how endothelial cells both sense and respond to microenvironmental forces. We present evolving concepts in endothelial force sensing and mechanical signaling and highlight recent insights gained from in vivo and 3D in vitro vascular models.
Collapse
Affiliation(s)
- Matthew L Kutys
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States; The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States.
| |
Collapse
|
38
|
Circulating Fibroblast Growth Factor-2, HIV-Tat, and Vascular Endothelial Cell Growth Factor-A in HIV-Infected Children with Renal Disease Activate Rho-A and Src in Cultured Renal Endothelial Cells. PLoS One 2016; 11:e0153837. [PMID: 27097314 PMCID: PMC4838216 DOI: 10.1371/journal.pone.0153837] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 04/05/2016] [Indexed: 01/14/2023] Open
Abstract
Renal endothelial cells (REc) are the first target of HIV-1 in the kidney. The integrity of REc is maintained at least partially by heparin binding growth factors that bind to heparan sulfate proteoglycans located on their cell surface. However, previous studies showed that the accumulation of two heparin-binding growth factors, Vascular Endothelial Cell Growth Factor-A (VEGF-A) and Fibroblast Growth Factor-2 (FGF-2), in combination with the viral protein Tat, can precipitate the progression of HIV-renal diseases. Nonetheless, very little is known about how these factors affect the behavior of REc in HIV+ children. We carried out this study to determine how VEGF-A, FGF-2, and HIV-Tat, modulate the cytoskeletal structure and permeability of cultured REc, identify key signaling pathways involved in this process, and develop a functional REc assay to detect HIV+ children affected by these changes. We found that VEGF-A and FGF-2, acting in synergy with HIV-Tat and heparin, affected the cytoskeletal structure and permeability of REc through changes in Rho-A, Src, and Rac-1 activity. Furthermore, urine samples from HIV+ children with renal diseases, showed high levels of VEGF-A and FGF-2, and induced similar changes in cultured REc and podocytes. These findings suggest that FGF-2, VEGF-A, and HIV-Tat, may affect the glomerular filtration barrier in HIV+ children through the induction of synergistic changes in Rho-A and Src activity. Further studies are needed to define the clinical value of the REc assay described in this study to identify HIV+ children exposed to circulating factors that may induce glomerular injury through similar mechanisms.
Collapse
|
39
|
Alteration of Pituitary Tumor Transforming Gene-1 Regulates Trophoblast Invasion via the Integrin/Rho-Family Signaling Pathway. PLoS One 2016; 11:e0149371. [PMID: 26900962 PMCID: PMC4764760 DOI: 10.1371/journal.pone.0149371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/01/2016] [Indexed: 11/23/2022] Open
Abstract
Trophoblast invasion ability is an important factor in early implantation and placental development. Recently, pituitary tumor transforming gene 1 (PTTG1) was shown to be involved in invasion and proliferation of cancer. However, the role of PTTG1 in trophoblast invasion remains unknown. Thus, in this study we analyzed PTTG1 expression in trophoblasts and its effect on trophoblast invasion activity and determined the mechanism through which PTTG1 regulates trophoblast invasion. Trophoblast proliferation and invasion abilities, regardless of PTTG1 expression, were analyzed by quantitative real-time polymerase chain reaction, fluorescence-activated cell sorting analysis, invasion assay, western blot, and zymography after treatment with small interfering RNA against PTTG1 (siPTTG1). Additionally, integrin/Rho-family signaling in trophoblasts by PTTG1 alteration was analyzed. Furthermore, the effect of PTTG1 on trophoblast invasion was evaluated by microRNA (miRNA) mimic and inhibitor treatment. Trophoblast invasion was significantly reduced through decreased matrix metalloproteinase (MMP)-2 and MMP-9 expression when PTTG1 expression was inhibited by siPTTG1 (p < 0.05). Furthermore, knockdown of PTTG1 increased expression of integrin alpha 4 (ITGA4), ITGA5, and integrin beta 1 (ITGB1); otherwise, RhoA expression was significantly decreased (p < 0.05). Treatment of miRNA-186-5p mimic and inhibitor controlled trophoblast invasion ability by altering PTTG1 and MMP expression. PTTG1 can control trophoblast invasion ability via regulation of MMP expression through integrin/Rho-family signaling. In addition, PTTG1 expression and its function were regulated by miRNA-186-5p. These results help in understanding the mechanism through which PTTG1 regulates trophoblast invasion and thereby implantation and placental development.
Collapse
|
40
|
Koo PK, Weitzman M, Sabanaygam CR, van Golen KL, Mochrie SGJ. Extracting Diffusive States of Rho GTPase in Live Cells: Towards In Vivo Biochemistry. PLoS Comput Biol 2015; 11:e1004297. [PMID: 26512894 PMCID: PMC4626024 DOI: 10.1371/journal.pcbi.1004297] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/26/2015] [Indexed: 11/19/2022] Open
Abstract
Resolving distinct biochemical interaction states when analyzing the trajectories of diffusing proteins in live cells on an individual basis remains challenging because of the limited statistics provided by the relatively short trajectories available experimentally. Here, we introduce a novel, machine-learning based classification methodology, which we call perturbation expectation-maximization (pEM), that simultaneously analyzes a population of protein trajectories to uncover the system of diffusive behaviors which collectively result from distinct biochemical interactions. We validate the performance of pEM in silico and demonstrate that pEM is capable of uncovering the proper number of underlying diffusive states with an accurate characterization of their diffusion properties. We then apply pEM to experimental protein trajectories of Rho GTPases, an integral regulator of cytoskeletal dynamics and cellular homeostasis, in vivo via single particle tracking photo-activated localization microcopy. Remarkably, pEM uncovers 6 distinct diffusive states conserved across various Rho GTPase family members. The variability across family members in the propensities for each diffusive state reveals non-redundant roles in the activation states of RhoA and RhoC. In a resting cell, our results support a model where RhoA is constantly cycling between activation states, with an imbalance of rates favoring an inactive state. RhoC, on the other hand, remains predominantly inactive. Single particle tracking is a powerful tool that captures the diffusive dynamics of proteins as they undergo various interactions in living cells. Uncovering different biochemical interactions by analyzing the diffusive behaviors of individual protein trajectories, however, is challenging due to the limited statistics provided by short trajectories and experimental noise sources which are intimately coupled into each protein’s localization. Here, we introduce a novel, unsupervised, machine-learning based classification methodology, which we call perturbation expectation-maximization (pEM), that simultaneously analyzes a population of protein trajectories to uncover the system of diffusive behaviors which collectively result from distinct biochemical interactions. We validate the performance of pEM in silico and in vivo on the biological system of Rho GTPase, a signal transduction protein responsible for regulating cytoskeletal dynamics. We envision that the presented methodology will be applicable to a wide range of single protein tracking data where different biochemical interactions result in distinct diffusive behaviors. More generally, this study brings us an important step closer to the possibility of monitoring the endogenous biochemistry of diffusing proteins within live cells with single molecule resolution.
Collapse
Affiliation(s)
- Peter K. Koo
- Department of Physics, Yale University, New Haven, Connecticut, United States of America
| | - Matthew Weitzman
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Chandran R. Sabanaygam
- Delaware Biotechnology Institute, Bioimaging Center, Newark, Delaware, United States of America
| | - Kenneth L. van Golen
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Simon G. J. Mochrie
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States of America
- Department of Applied Physics, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
41
|
Neves-Carvalho A, Logarinho E, Freitas A, Duarte-Silva S, Costa MDC, Silva-Fernandes A, Martins M, Serra SC, Lopes AT, Paulson HL, Heutink P, Relvas JB, Maciel P. Dominant negative effect of polyglutamine expansion perturbs normal function of ataxin-3 in neuronal cells. Hum Mol Genet 2014; 24:100-17. [PMID: 25143392 DOI: 10.1093/hmg/ddu422] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The physiological function of Ataxin-3 (ATXN3), a deubiquitylase (DUB) involved in Machado-Joseph Disease (MJD), remains elusive. In this study, we demonstrate that ATXN3 is required for neuronal differentiation and for normal cell morphology, cytoskeletal organization, proliferation and survival of SH-SY5Y and PC12 cells. This cellular phenotype is associated with increased proteasomal degradation of α5 integrin subunit (ITGA5) and reduced activation of integrin signalling and is rescued by ITGA5 overexpression. Interestingly, silencing of ATXN3, overexpression of mutant versions of ATXN3 lacking catalytic activity or bearing an expanded polyglutamine (polyQ) tract led to partially overlapping phenotypes. In vivo analysis showed that both Atxn3 knockout and MJD transgenic mice had decreased levels of ITGA5 in the brain. Furthermore, abnormal morphology and reduced branching were observed both in cultured neurons expressing shRNA for ATXN3 and in those obtained from MJD mice. Our results show that ATXN3 rescues ITGA5 from proteasomal degradation in neurons and that polyQ expansion causes a partial loss of this cellular function, resulting in reduced integrin signalling and neuronal cytoskeleton modifications, which may be contributing to neurodegeneration.
Collapse
Affiliation(s)
- Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | - Elsa Logarinho
- Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal
| | - Ana Freitas
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | | | - Anabela Silva-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | - Margarida Martins
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | - Sofia Cravino Serra
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | - André T Lopes
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA and
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - João B Relvas
- Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães and
| |
Collapse
|
42
|
El-Sayed FG, Camden JM, Woods LT, Khalafalla MG, Petris MJ, Erb L, Weisman GA. P2Y2 nucleotide receptor activation enhances the aggregation and self-organization of dispersed salivary epithelial cells. Am J Physiol Cell Physiol 2014; 307:C83-96. [PMID: 24760984 DOI: 10.1152/ajpcell.00380.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hyposalivation resulting from salivary gland dysfunction leads to poor oral health and greatly reduces the quality of life of patients. Current treatments for hyposalivation are limited. However, regenerative medicine to replace dysfunctional salivary glands represents a revolutionary approach. The ability of dispersed salivary epithelial cells or salivary gland-derived progenitor cells to self-organize into acinar-like spheres or branching structures that mimic the native tissue holds promise for cell-based reconstitution of a functional salivary gland. However, the mechanisms involved in salivary epithelial cell aggregation and tissue reconstitution are not fully understood. This study investigated the role of the P2Y2 nucleotide receptor (P2Y2R), a G protein-coupled receptor that is upregulated following salivary gland damage and disease, in salivary gland reconstitution. In vitro results with the rat parotid acinar Par-C10 cell line indicate that P2Y2R activation with the selective agonist UTP enhances the self-organization of dispersed salivary epithelial cells into acinar-like spheres. Other results indicate that the P2Y2R-mediated response is dependent on epidermal growth factor receptor activation via the metalloproteases ADAM10/ADAM17 or the α5β1 integrin/Cdc42 signaling pathway, which leads to activation of the MAPKs JNK and ERK1/2. Ex vivo data using primary submandibular gland cells from wild-type and P2Y2R(-/-) mice confirmed that UTP-induced migratory responses required for acinar cell self-organization are mediated by the P2Y2R. Overall, this study suggests that the P2Y2R is a promising target for salivary gland reconstitution and identifies the involvement of two novel components of the P2Y2R signaling cascade in salivary epithelial cells, the α5β1 integrin and the Rho GTPase Cdc42.
Collapse
Affiliation(s)
- Farid G El-Sayed
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Jean M Camden
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Lucas T Woods
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Mahmoud G Khalafalla
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Michael J Petris
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Department of Nutritional Sciences and Exercise Physiology, University of Missouri, Columbia, Missouri; and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Laurie Erb
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| | - Gary A Weisman
- Department of Biochemistry, University of Missouri, Columbia, Missouri; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
43
|
Wang HY, Chen Z, Wang ZH, Wang H, Huang LM. Prognostic significance of α5β1-integrin expression in cervical cancer. Asian Pac J Cancer Prev 2014; 14:3891-5. [PMID: 23886203 DOI: 10.7314/apjcp.2013.14.6.3891] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The purpose of this study was to evaluate the association of expression of α5β1-integrin with clinicopathologic features and prognosis in cervical cancer. Levels of α5β1-integrin in normal cervical mucosa and cervical cancer tissue were detected with immunohistochemistry. Survival analysis by the Kaplan-Meier method was performed to assess prognostic significance. α5β1-integrin expression was detected in 84.6% (143/169) cervical cancer samples, significantly different from that in normal cervical mucosa (P < 0.05). Positive expression rates of α5β1-integrin in patients with poor histologic differentiation, lymph node metastasis, and recurrence were elevated. Using Kaplan-Meier analysis, a comparison of survival curves of low versus high expression of α5β1-integrin revealed a highly significant difference in human cervical cancer cases (P < 0.05), suggesting that overexpression of α5β1-integrin is associated with a worse prognosis.The α5β1-integrin promotes angiogenesis and associates with lymph node metastasis, vascular invasion and poor prognosis of cervical cancer. The current study indicated that α5β1-integrin may be an independent prognostic factor for cervical cancer patients.
Collapse
Affiliation(s)
- Hua-Yi Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | | | | | | | | |
Collapse
|
44
|
Fu X, Xu M, Liu J, Qi Y, Li S, Wang H. Regulation of migratory activity of human keratinocytes by topography of multiscale collagen-containing nanofibrous matrices. Biomaterials 2014; 35:1496-506. [PMID: 24268197 PMCID: PMC5682108 DOI: 10.1016/j.biomaterials.2013.11.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/02/2013] [Indexed: 10/26/2022]
Abstract
Nanofibrous matrices hold great promise in skin wound repair partially due to their capability of recapturing the essential attributes of native extracellular matrix (ECM). With regard to limited studies on the effect of nanofibrous matrices on keratinocytes, the present study was aimed to understand how the topographical feature of nanofibrous matrices regulates keratinocyte motility by culturing keratinocytes on polycaprolactone (PCL)/collagen nanofibrous matrices (rough surface with fiber diameters of 331 ± 112 nm) or the matrices coated with a thin layer of collagen gel to form a secondary ultrafine fibrous network (smooth surface with ultrafine fiber diameters of 55 ± 26 nm). It was found that the PCL/collagen nanofibrous matrices alone did not stimulate cell migration, while collagen gel coating could significantly increase cell motility. Further studies demonstrated that the ultrafine fibrous network of collagen gel coating significantly activated integrin β1, Rac1 and Cdc42, facilitated the deposition of laminin-332 (formerly called laminin-5), and promoted the expression of active matrix metalloproteinases (MMPs) (i.e., MMP-2 and 9). Neutralization of integrin β1 activity abrogated the gel coating-induced keratinocyte migration. These findings provide important evidence on the role of topographical features of nanofibrous matrices in regulating the phenotypic alteration of keratinocytes and suggest the possible utility of collagen-containing nanofibrous matrices for skin regeneration especially in re-epithelialization.
Collapse
Affiliation(s)
- Xiaoling Fu
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA; The National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510640, People's Republic of China; The School of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Meng Xu
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Jie Liu
- Department of Surgery, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | - Yanmei Qi
- Department of Surgery, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | - Shaohua Li
- Department of Surgery, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | - Hongjun Wang
- Department of Chemistry, Chemical Biology and Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA.
| |
Collapse
|
45
|
Azizi G, Boghozian R, Mirshafiey A. The potential role of angiogenic factors in rheumatoid arthritis. Int J Rheum Dis 2014; 17:369-83. [PMID: 24467605 DOI: 10.1111/1756-185x.12280] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Angiogenesis is an important phenomenon in the pathogenesis of some diseases, such as numerous types of tumors and autoimmunity, and also a number of soluble and cell-bound factors may stimulate neovascularization in inflammatory reaction processes. Here, by highlighting the significance of angiogenesis reaction in rheumatoid arthritis (RA), we will mainly focus on the role of various growth factors, cytokines, enzymes, cells, hypoxic conditions and transcription factors in the angiogenic process and we will then explain some therapeutic strategies based on blockage of angiogenesis and modification of the vascular pathology in RA.
Collapse
Affiliation(s)
- Gholamreza Azizi
- Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | | | | |
Collapse
|
46
|
Kim SJ, Wan Q, Cho E, Han B, Yoder MC, Voytik-Harbin SL, Na S. Matrix rigidity regulates spatiotemporal dynamics of Cdc42 activity and vacuole formation kinetics of endothelial colony forming cells. Biochem Biophys Res Commun 2014; 443:1280-5. [PMID: 24393843 DOI: 10.1016/j.bbrc.2013.12.135] [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] [Received: 12/09/2013] [Accepted: 12/26/2013] [Indexed: 12/15/2022]
Abstract
Recent evidence has shown that endothelial colony forming cells (ECFCs) may serve as a cell therapy for improving blood vessel formation in subjects with vascular injury, largely due to their robust vasculogenic potential. The Rho family GTPase Cdc42 is known to play a primary role in this vasculogenesis process, but little is known about how extracellular matrix (ECM) rigidity affects Cdc42 activity during the process. In this study, we addressed two questions: Does matrix rigidity affect Cdc42 activity in ECFC undergoing early vacuole formation? How is the spatiotemporal activation of Cdc42 related to ECFC vacuole formation? A fluorescence resonance energy transfer (FRET)-based Cdc42 biosensor was used to examine the effects of the rigidity of three-dimensional (3D) collagen matrices on spatiotemporal activity of Cdc42 in ECFCs. Collagen matrix stiffness was modulated by varying the collagen concentration and therefore fibril density. The results showed that soft (150 Pa) matrices induced an increased level of Cdc42 activity compared to stiff (1 kPa) matrices. Time-course imaging and colocalization analysis of Cdc42 activity and vacuole formation revealed that Cdc42 activity was colocalized to the periphery of cytoplasmic vacuoles. Moreover, soft matrices generated faster and larger vacuoles than stiff matrices. The matrix-driven vacuole formation was enhanced by a constitutively active Cdc42 mutant, but significantly inhibited by a dominant-negative Cdc42 mutant. Collectively, the results suggest that matrix rigidity is a strong regulator of Cdc42 activity and vacuole formation kinetics, and that enhanced activity of Cdc42 is an important step in early vacuole formation in ECFCs.
Collapse
Affiliation(s)
- Seung Joon Kim
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Qiaoqiao Wan
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Eunhye Cho
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
47
|
Ren J, Xu S, Guo D, Zhang J, Liu S. Increased expression of α5β1-integrin is a prognostic marker for patients with gastric cancer. Clin Transl Oncol 2013; 16:668-74. [PMID: 24248895 DOI: 10.1007/s12094-013-1133-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/01/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The study was to evaluate the association of expression level of α5β1-integrin with clinicopathologic features and prognosis in gastric cancer (GC). METHODS The expression of α5β1-integrin in normal gastric mucosa and GC tissue was detected with immunohistochemistry. The level of α5 and β1 mRNA in GC tissues and non-neoplastic tissues was evaluated in 48 paired cases by quantitative real-time polymerase chain reaction (qRT-PCR). Survival analysis by the Kaplan-Meier method was performed to assess prognostic significance. RESULTS The α5β1-integrin expression was detected in 68.3 % (127/186) GC samples, and there was a significant difference on their positive expression rate between GC tissue and normal gastric mucosa (P < 0.001). The positive expression rate of α5β1-integrin in patients with poor histologic differentiation (P = 0.001), lymph node metastasis (P < 0.001), and recurrence (P < 0.001) group was heightened. Using Kaplan-Meier analysis, a comparison of survival curves of low versus high expresser of α5β1-integrin revealed a highly significant difference in human GC tissue (P = 0.002), which suggested that overexpression of α5β1-integrin is associated with a worse prognosis. Multivariate analyses showed that α5β1-integrin expression was independent risk factor predicting overall survival [Hazard ratio (HR) 1.594, 95 % confidence interval (CI) 1.236-2.408, P = 0.006] and disease-free survival [HR 3.952, 95 % CI 1.676-9.861, P = 0.003] in GC. CONCLUSIONS The α5β1-integrin promotes angiogenesis and associates with lymph node metastasis, vascular invasion and poor prognosis of GC. The current study shows that α5β1-integrin may be an independent prognostic factor for GC patients.
Collapse
Affiliation(s)
- J Ren
- Department of Pathology, Shenzhen Futian Hospital Affiliated to Guangdong Medical College, 3025 Shennan Middle Road, Shenzhen, 518029, Guangdong, People's Republic of China
| | | | | | | | | |
Collapse
|
48
|
p120 catenin: an essential regulator of cadherin stability, adhesion-induced signaling, and cancer progression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:409-32. [PMID: 23481205 DOI: 10.1016/b978-0-12-394311-8.00018-2] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
p120 catenin is the best studied member of a subfamily of proteins that associate with the cadherin juxtamembrane domain to suppress cadherin endocytosis. p120 also recruits the minus ends of microtubules to the cadherin complex, leading to junction maturation. In addition, p120 regulates the activity of Rho family GTPases through multiple interactions with Rho GEFs, GAPs, Rho GTPases, and their effectors. Nuclear signaling is affected by the interaction of p120 with Kaiso, a transcription factor regulating Wnt-responsive genes as well as transcriptionally repressing methylated promoters. Multiple alternatively spliced p120 isoforms and complex phosphorylation events affect these p120 functions. In cancer, reduced p120 expression correlates with reduced E-cadherin function and with tumor progression. In contrast, in tumor cells that have lost E-cadherin expression, p120 promotes cell invasion and anchorage-independent growth. Furthermore, p120 is required for Src-induced oncogenic transformation and provides a potential target for future therapeutic interventions.
Collapse
|
49
|
Wang J, Li X, Cheng H, Wang K, Lu W, Wen T. Overexpression of Rho-GDP-dissociation inhibitor-γ inhibits migration of neural stem cells. J Neurosci Res 2013; 91:1394-401. [PMID: 23996536 DOI: 10.1002/jnr.23261] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/03/2013] [Accepted: 05/21/2013] [Indexed: 12/11/2022]
Abstract
Neural stem cell (NSC) migration relies heavily on the regulation of actin and microtubule cytoskeletons by Rho GTPases, which are critical regulators of key steps during NSC migration. However, the migration mechanism remains unclear. Rho-GDP-dissociation inhibitor-γ (Rho-GDIγ) was identified as an important downregulator of the Rho family of GTPases, because of its ability to prevent nucleotide exchange and thus membrane association. This study investigates the role of Rho-GDIγ in neural stem cells migration. Our results indicate that the overexpression of Rho-GDIγ maintains NSCs in the stem cell state, meanwhile preventing NSC migration through inhibition of Rac1 expression, one of the Rho-family GTPases. This study provides the basis for further study of the molecular mechanism of NSC migration.
Collapse
Affiliation(s)
- Jiao Wang
- Laboratory of Molecular Neural Biology, Institute of Systems Biology, School of Life Sciences, Shanghai University, Shanghai, China; School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
50
|
Azzi S, Hebda JK, Gavard J. Vascular permeability and drug delivery in cancers. Front Oncol 2013; 3:211. [PMID: 23967403 PMCID: PMC3744053 DOI: 10.3389/fonc.2013.00211] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/01/2013] [Indexed: 01/22/2023] Open
Abstract
The endothelial barrier strictly maintains vascular and tissue homeostasis, and therefore modulates many physiological processes such as angiogenesis, immune responses, and dynamic exchanges throughout organs. Consequently, alteration of this finely tuned function may have devastating consequences for the organism. This is particularly obvious in cancers, where a disorganized and leaky blood vessel network irrigates solid tumors. In this context, vascular permeability drives tumor-induced angiogenesis, blood flow disturbances, inflammatory cell infiltration, and tumor cell extravasation. This can directly restrain the efficacy of conventional therapies by limiting intravenous drug delivery. Indeed, for more effective anti-angiogenic therapies, it is now accepted that not only should excessive angiogenesis be alleviated, but also that the tumor vasculature needs to be normalized. Recovery of normal state vasculature requires diminishing hyperpermeability, increasing pericyte coverage, and restoring the basement membrane, to subsequently reduce hypoxia, and interstitial fluid pressure. In this review, we will introduce how vascular permeability accompanies tumor progression and, as a collateral damage, impacts on efficient drug delivery. The molecular mechanisms involved in tumor-driven vascular permeability will next be detailed, with a particular focus on the main factors produced by tumor cells, especially the emblematic vascular endothelial growth factor. Finally, new perspectives in cancer therapy will be presented, centered on the use of anti-permeability factors and normalization agents.
Collapse
Affiliation(s)
- Sandy Azzi
- CNRS, UMR8104 , Paris , France ; INSERM, U1016 , Paris , France ; Sorbonne Paris Cite, Universite Paris Descartes , Paris , France
| | | | | |
Collapse
|