1
|
Wu C, Luo D, Shi B, Chen S, Sun C, He Z, Yu C. Sciellin inhibits senescence and promotes pancreatic cancer progress by activating the notch signaling pathway. Sci Rep 2025; 15:16133. [PMID: 40341648 PMCID: PMC12062315 DOI: 10.1038/s41598-025-88265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/28/2025] [Indexed: 05/10/2025] Open
Abstract
Pancreatic cancer (PC) incidence is increasing annually globally, and the five-year survival rate of patients with PC is approximately 10%. Cellular senescence is a regulatory mechanism against cancer that prevents tumor development by inhibiting the proliferation of damaged or abnormal cells. However, the mechanisms underlying cellular senescence in PC is unclear. Sciellin (SCEL) is a precursor protein of the cornified envelope predominantly enriched in epithelial cells. Previous studies have discovered potential links between SCEL and cellular senescence through bioinformatics analysis. Therefore, the specific role of SCEL in cellular senescence and the malignant features of PC are unclear. In vivo and in vitro assays were performed to investigate the role of SCEL in PC cell senescence, proliferation, invasion, and metastasis. Gene set enrichment analysis was used to identify the Notch signaling pathways activated by SCEL, and coimmunoprecipitation was used to detect proteins that interact with SCEL. The results revealed that SCEL was significantly upregulated in PC tissues and cell models and was correlated with poor clinical outcomes. Further investigation revealed that the interaction between SCEL and Jagged-1 promotes the activation of the Notch signaling pathway, effectively inhibiting the senescence of PC cells while enhancing their proliferation, invasion, and metastatic capabilities. Therefore, SCEL is a potential therapeutic target for PC.
Collapse
Affiliation(s)
- Changhao Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Dan Luo
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
- Department of Hepatobiliary Surgery, People's Hospital of the Guizhou Province, Guiyang, 550003, China
| | - Binbin Shi
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Shiyu Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, Medical School, South China Hospital, Shenzhen University, Shenzhen, 518116, China
| | - Chengyi Sun
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China.
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China.
| | - Chao Yu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China.
- College of Clinical Medicine, Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Institute of Hepatobiliary, Pancreatic and Splenic Diseases, Guiyang, 550001, China.
- Key Laboratory of Liver, Gallbladder, Pancreas and Spleen of Guizhou Medical University, Guiyang, 550001, China.
- Guizhou Provincial Clinical Medical Research Center of Hepatobiliary Surgery, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
2
|
Banerjee DS, Falk MJ, Gardel ML, Walczak AM, Mora T, Vaikuntanathan S. Learning via mechanosensitivity and activity in cytoskeletal networks. ARXIV 2025:arXiv:2504.15107v1. [PMID: 40313666 PMCID: PMC12045384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
In this work we show how a network inspired by a coarse-grained description of actomyosin cytoskeleton can learn - in a contrastive learning framework - from environmental perturbations if it is endowed with mechanosensitive proteins and motors. Our work is a proof of principle for how force-sensitive proteins and molecular motors can form the basis of a general strategy to learn in biological systems. Our work identifies a minimal biologically plausible learning mechanism and also explores its implications for commonly occuring phenomenolgy such as adaptation and homeostatis.
Collapse
Affiliation(s)
- Deb S. Banerjee
- James Franck Institute, University of Chicago, Chicago, IL 60637
| | - Martin J. Falk
- James Franck Institute, University of Chicago, Chicago, IL 60637
- Department of Physics, University of Chicago, Chicago, IL 60637
| | - Margaret L. Gardel
- James Franck Institute, University of Chicago, Chicago, IL 60637
- Department of Physics, University of Chicago, Chicago, IL 60637
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Pritzker School for Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Aleksandra M. Walczak
- James Franck Institute, University of Chicago, Chicago, IL 60637
- Department of Physics, University of Chicago, Chicago, IL 60637
- Laboratoire de physique de l’École normale supérieure, CNRS, Paris Sciences et Lettres University, Sorbonne Université, and Université Paris-Cité, Paris, 75005, France
| | - Thierry Mora
- James Franck Institute, University of Chicago, Chicago, IL 60637
- Department of Physics, University of Chicago, Chicago, IL 60637
- Laboratoire de physique de l’École normale supérieure, CNRS, Paris Sciences et Lettres University, Sorbonne Université, and Université Paris-Cité, Paris, 75005, France
| | - Suriyanarayanan Vaikuntanathan
- James Franck Institute, University of Chicago, Chicago, IL 60637
- Department of Chemistry, University of Chicago, Chicago, IL 60637
| |
Collapse
|
3
|
Yang S, Zhang X, Li X, Li H. Crip2 affects vascular development by fine-tuning endothelial cell aggregation and proliferation. Cell Mol Life Sci 2025; 82:110. [PMID: 40074973 PMCID: PMC11904032 DOI: 10.1007/s00018-025-05624-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025]
Abstract
Endothelial cell adhesion and migration are crucial to various biological processes, including vascular development. The identification of factors that modulate vascular development through these cell functions has emerged as a prominent focus in cardiovascular research. Crip2 is known to play a crucial role in cardiac development, yet its involvement in vascular development and the underlying mechanism remains elusive. In this study, we revealed that Crip2 is expressed predominantly in the vascular system, particularly in the posterior cardinal vein and caudal vein plexus intersegmental vein. Upon Crip2 loss, the posterior cardinal vein plexus and caudal vein plexus are hypoplastic, and endothelial cells exhibit aberrant aggregation. In human umbilical vein endothelial cells (HUVECs), CRIP2 interacts with the cytoskeleton proteins KRT8 and VIM. The absence of CRIP2 negatively regulates their expression, thereby fine-tuning cytoskeleton formation, resulting in a hyperadhesive phenotype. Moreover, CRIP2 deficiency perturbs the VEGFA/CDC42 signaling pathway, which in turn diminishes the migrating capacity of HUVECs. Furthermore, the loss of CRIP2 impairs cell proliferation by affecting its interaction with SRF through PDE10A/cAMP and PDGF/JAK/STAT/SRF signaling. Collectively, our findings delineate a crucial role for CRIP2 in controlling the migration, adhesion and proliferation of endothelial cells, thereby contributing to vascular development in zebrafish. These insights may provide a deeper understanding of the etiology of cardiovascular disorders.
Collapse
Affiliation(s)
- Shuaiqi Yang
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education), Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Xiangmin Zhang
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education), Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Xianpeng Li
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hongyan Li
- College of Marine Life Sciences, Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education), Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
- Ocean University of China, Room 301, Darwin Building, 5 Yushan Road, Qingdao, 266003, China.
| |
Collapse
|
4
|
Phua DYZ, Sun X, Alushin GM. Force-activated zyxin assemblies coordinate actin nucleation and crosslinking to orchestrate stress fiber repair. Curr Biol 2025; 35:854-870.e9. [PMID: 39952249 PMCID: PMC11867737 DOI: 10.1016/j.cub.2025.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
As the cytoskeleton sustains cell and tissue forces, it incurs physical damage that must be repaired to maintain mechanical homeostasis. The LIN-11, Isl-1, and Mec-3 (LIM)-domain protein zyxin detects force-induced ruptures in actin-myosin stress fibers, coordinating downstream repair factors to restore stress fiber integrity through unclear mechanisms. Here, we reconstitute stress fiber repair with purified proteins, uncovering detailed links between zyxin's force-regulated binding interactions and cytoskeletal dynamics. In addition to binding individual tensed actin filaments (F-actin), zyxin's LIM domains form force-dependent assemblies that bridge broken filament fragments. Zyxin assemblies engage repair factors through multivalent interactions, coordinating nucleation of new F-actin by VASP and its crosslinking into aligned bundles by ɑ-actinin. Through these combined activities, stress fiber repair initiates within the cores of micron-scale damage sites in cells, explaining how these F-actin-depleted regions are rapidly restored. Thus, zyxin's force-dependent organization of actin repair machinery inherently operates at the network scale to maintain cytoskeletal integrity.
Collapse
Affiliation(s)
- Donovan Y Z Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA.
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
5
|
Troyanovsky RB, Indra I, Troyanovsky SM. Actin-dependent α-catenin oligomerization contributes to adherens junction assembly. Nat Commun 2025; 16:1801. [PMID: 39979305 PMCID: PMC11842732 DOI: 10.1038/s41467-025-57079-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Classic cadherins, specifically E-cadherin in most epithelial cells, are transmembrane adhesion receptors, whose intracellular region interacts with proteins, termed catenins, forming the cadherin-catenin complex (CCC). The cadherin ectodomain generates 2D adhesive clusters (E-clusters) through cooperative trans and cis interactions, while catenins anchor the E-clusters to the actin cytoskeleton. How these two types of interactions are coordinated in the formation of specialized cell-cell adhesions, adherens junctions (AJ), remains unclear. Here, we focus on the role of the actin-binding domain of α-catenin (αABD) by showing that the interaction of the αABD with actin generates actin-bound linear CCC oligomers (CCC/actin strands) incorporating up to six CCCs. This actin-driven CCC oligomerization, which is cadherin ectodomain independent, preferentially occurs along the actin cortex enriched with key basolateral proteins, myosin-1c, scribble, and DLG1. In cell-cell contacts, the CCC/actin strands integrate with the E-clusters giving rise to the composite oligomers, E/actin clusters. Targeted inactivation of strand formation by point mutations emphasizes the importance of this oligomerization process for blocking intercellular protrusive membrane activity and for coupling AJs with the actomyosin-derived tensional forces.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Indrajyoti Indra
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Department of Cell & Developmental Biology, The Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Zhou R, Wang M, Li X, Liu Y, Yao Y, Wang A, Chen C, Zhang Q, Wu Q, Zhang Q, Neculai D, Xia B, Shao JZ, Feng XH, Liang T, Zou J, Wang X, Xu P. TBK1-Zyxin signaling controls tumor-associated macrophage recruitment to mitigate antitumor immunity. EMBO J 2024; 43:4984-5017. [PMID: 39304793 PMCID: PMC11535546 DOI: 10.1038/s44318-024-00244-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
Mechanical control is fundamental for cellular localization within a tissue, including for tumor-associated macrophages (TAMs). While the innate immune sensing pathways cGAS-STING and RLR-MAVS impact the pathogenesis and therapeutics of malignant diseases, their effects on cell residency and motility remain incompletely understood. Here, we uncovered that TBK1 kinase, activated by cGAS-STING or RLR-MAVS signaling in macrophages, directly phosphorylates and mobilizes Zyxin, a key regulator of actin dynamics. Under pathological conditions and in STING or MAVS signalosomes, TBK1-mediated Zyxin phosphorylation at S143 facilitates rapid recruitment of phospho-Zyxin to focal adhesions, leading to subsequent F-actin reorganization and reduced macrophage migration. Intratumoral STING-TBK1-Zyxin signaling was evident in TAMs and critical in antitumor immunity. Furthermore, myeloid-specific or global disruption of this signaling decreased the population of CD11b+ F4/80+ TAMs and promoted PD-1-mediated antitumor immunotherapy. Thus, our findings identify a new biological function of innate immune sensing pathways by regulating macrophage tissue localization, thus providing insights into context-dependent mitigation of antitumor immunity.
Collapse
Affiliation(s)
- Ruyuan Zhou
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mengqiu Wang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Xiao Li
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yutong Liu
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yihan Yao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ailian Wang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Chen Chen
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Qian Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Qirou Wu
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Qi Zhang
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
| | - Dante Neculai
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Bing Xia
- Department of Thoracic Cancer, Affiliated Hangzhou Cancer Hospital, Westlake University, Hangzhou, 310030, China
| | - Jian-Zhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Xin-Hua Feng
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Tingbo Liang
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Jian Zou
- Eye Center of the Second Affiliated Hospital School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiaojian Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Pinglong Xu
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 310058, China.
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Sala S, Caillier A, Oakes PW. Principles and regulation of mechanosensing. J Cell Sci 2024; 137:jcs261338. [PMID: 39297391 PMCID: PMC11423818 DOI: 10.1242/jcs.261338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
Research over the past two decades has highlighted that mechanical signaling is a crucial component in regulating biological processes. Although many processes and proteins are termed 'mechanosensitive', the underlying mechanisms involved in mechanosensing can vary greatly. Recent studies have also identified mechanosensing behaviors that can be regulated independently of applied force. This important finding has major implications for our understanding of downstream mechanotransduction, the process by which mechanical signals are converted into biochemical signals, as it offers another layer of biochemical regulatory control for these crucial signaling pathways. In this Review, we discuss the different molecular and cellular mechanisms of mechanosensing, how these processes are regulated and their effects on downstream mechanotransduction. Together, these discussions provide an important perspective on how cells and tissues control the ways in which they sense and interpret mechanical signals.
Collapse
Affiliation(s)
- Stefano Sala
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Alexia Caillier
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Patrick W. Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| |
Collapse
|
8
|
Katsuta H, Sokabe M, Hirata H. From stress fiber to focal adhesion: a role of actin crosslinkers in force transmission. Front Cell Dev Biol 2024; 12:1444827. [PMID: 39193363 PMCID: PMC11347286 DOI: 10.3389/fcell.2024.1444827] [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: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
The contractile apparatus, stress fiber (SF), is connected to the cell adhesion machinery, focal adhesion (FA), at the termini of SF. The SF-FA complex is essential for various mechanical activities of cells, including cell adhesion to the extracellular matrix (ECM), ECM rigidity sensing, and cell migration. This mini-review highlights the importance of SF mechanics in these cellular activities. Actin-crosslinking proteins solidify SFs by attenuating myosin-driven flows of actin and myosin filaments within the SF. In the solidified SFs, viscous slippage between actin filaments in SFs and between the filaments and the surrounding cytosol is reduced, leading to efficient transmission of myosin-generated contractile force along the SFs. Hence, SF solidification via actin crosslinking ensures exertion of a large force to FAs, enabling FA maturation, ECM rigidity sensing and cell migration. We further discuss intracellular mechanisms for tuning crosslinker-modulated SF mechanics and the potential relationship between the aberrance of SF mechanics and pathology including cancer.
Collapse
Affiliation(s)
- Hiroki Katsuta
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Human Information Systems Laboratories, Kanazawa Institute of Technology, Hakusan, Japan
| | - Hiroaki Hirata
- Department of Applied Bioscience, Kanazawa Institute of Technology, Hakusan, Japan
| |
Collapse
|
9
|
Kim T, Egesa A, Qin C, Mather H, Sandoya G, Begcy K. Global identification of LIM genes in response to different heat stress regimes in Lactuca sativa. BMC PLANT BIOLOGY 2024; 24:751. [PMID: 39103763 DOI: 10.1186/s12870-024-05466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND LIM (Lineage-11 (LIN-11), Insulin-1 (ISL-1), and Mechanotransduction-3 (MEC-3)) genes belong to a family that hold ubiquitous properties contributing to organ, seed, and pollen development as well as developmental and cellular responses to biotic and abiotic stresses. Lettuce (Lactuca sativa) is a highly consumed vegetable crop susceptible heat stress. High temperatures limit lettuce's overall yield, quality and marketability. Lettuce LIM genes have not been identified and their role in response to high temperatures is not known. Aiming to identify potential new targets for thermoresilience, we searched for LIM genes in lettuce and compared them with orthologous of several dicotyledons and monocotyledons plant species. RESULTS We identified fourteen lettuce LIM genes distributed into eight different subgroups using a genome-wide analysis strategy. Three belonging to DAR (DA means "large" in Chinese) class I, two DAR class II, one in the WLIM1, two in the WLIM2, one in the PLIM1, two in the PLIM2 class, one ßLIM and two δLIMs. No DAR-like were identified in any of the species analyzed including lettuce. Interestingly, unlike other gene families in lettuce which underwent large genome tandem duplications, LIM genes did not increase in number compared to other plant species. The response to heat stress induced a dynamic transcriptional response on LsLIM genes. All heat stress regimes, including night stress, day stress and day and night stress were largely responsible for changes in LIM transcriptional expression. CONCLUSIONS Our global analysis at the genome level provides a detailed identification of LIM genes in lettuce and other dicotyledonous and monocotyledonous plant species. Gene structure, physical and chemical properties as well as chromosomal location and Cis-regulatory element analysis together with our gene expression analysis under different temperature regimes identified LsWLIM1, LsWLIM2b, LsDAR3 and LsDAR5 as candidate genes that could be used by breeding programs aiming to produce lettuce varieties able to withstand high temperatures.
Collapse
Affiliation(s)
- Taehoon Kim
- Environmental Horticulture Department, University of Florida, Gainesville, FL, 32611, USA
| | - Andrew Egesa
- Environmental Horticulture Department, University of Florida, Gainesville, FL, 32611, USA
| | - Claire Qin
- Environmental Horticulture Department, University of Florida, Gainesville, FL, 32611, USA
- Student Science Training Program, University of Florida, Gainesville, FL, 32611, USA
| | - Hannah Mather
- Environmental Horticulture Department, University of Florida, Gainesville, FL, 32611, USA
- Horticultural Science Department, University of Florida, Gainesville, FL, 32611, USA
| | - Germán Sandoya
- Horticultural Science Department, University of Florida, Gainesville, FL, 32611, USA
- Everglades Research and Education Center, Horticultural Sciences Department, University of Florida IFAS, Belle Glade, FL, 33430, USA
| | - Kevin Begcy
- Environmental Horticulture Department, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
10
|
Sales Conniff A, Tur J, Kohena K, Zhang M, Gibbons J, Heller LC. DNA Electrotransfer Regulates Molecular Functions in Skeletal Muscle. Bioelectricity 2024; 6:80-90. [PMID: 39119567 PMCID: PMC11304878 DOI: 10.1089/bioe.2022.0041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Background Tissues, such as skeletal muscle, have been targeted for the delivery of plasmid DNA (pDNA) encoding vaccines and therapeutics. The application of electric pulses (electroporation or electrotransfer) increases cell membrane permeability to enhance plasmid delivery and expression. However, the molecular effects of DNA electrotransfer on the muscle tissue are poorly characterized. Materials and Methods Four hours after intramuscular plasmid electrotransfer, we evaluated gene expression changes by RNA sequencing. Differentially expressed genes were analyzed by gene ontology (GO) pathway enrichment analysis. Results GO analysis highlighted many enriched molecular functions. The terms regulated by pulse application were related to muscle stress, the cytoskeleton and inflammation. The terms regulated by pDNA injection were related to a DNA-directed response and its control. Several terms regulated by pDNA electrotransfer were similar to those regulated by pulse application. However, the terms related to pDNA injection differed, focusing on entry of the plasmid into the cells and intracellular trafficking. Conclusion Each muscle stimulus resulted in specific regulated molecular functions. Identifying the unique intrinsic molecular changes driven by intramuscular DNA electrotransfer will aid in the design of preventative and therapeutic gene therapies.
Collapse
Affiliation(s)
- Amanda Sales Conniff
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| | - Jared Tur
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| | - Kristopher Kohena
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| | - Min Zhang
- USF Genomics Core, University of South Florida, Tampa, Florida, USA
| | - Justin Gibbons
- USF Omics Hub, University of South Florida, Tampa, Florida, USA
| | - Loree C. Heller
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
11
|
Phua DY, Sun X, Alushin GM. Force-activated zyxin assemblies coordinate actin nucleation and crosslinking to orchestrate stress fiber repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594765. [PMID: 38798419 PMCID: PMC11118565 DOI: 10.1101/2024.05.17.594765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
As the cytoskeleton sustains cell and tissue forces, it incurs physical damage that must be repaired to maintain mechanical homeostasis. The LIM-domain protein zyxin detects force-induced ruptures in actin-myosin stress fibers, coordinating downstream repair factors to restore stress fiber integrity through unclear mechanisms. Here, we reconstitute stress fiber repair with purified proteins, uncovering detailed links between zyxin's force-regulated binding interactions and cytoskeletal dynamics. In addition to binding individual tensed actin filaments (F-actin), zyxin's LIM domains form force-dependent assemblies that bridge broken filament fragments. Zyxin assemblies engage repair factors through multi-valent interactions, coordinating nucleation of new F-actin by VASP and its crosslinking into aligned bundles by ɑ-actinin. Through these combined activities, stress fiber repair initiates within the cores of micron-scale damage sites in cells, explaining how these F-actin depleted regions are rapidly restored. Thus, zyxin's force-dependent organization of actin repair machinery inherently operates at the network scale to maintain cytoskeletal integrity.
Collapse
Affiliation(s)
- Donovan Y.Z. Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
12
|
Wu Z, Wu D, Zhong Q, Zou X, Liu Z, Long H, Wei J, Li X, Dai F. The role of zyxin in signal transduction and its relationship with diseases. Front Mol Biosci 2024; 11:1371549. [PMID: 38712343 PMCID: PMC11070705 DOI: 10.3389/fmolb.2024.1371549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
This review highlighted the pivotal role of zyxin, an essential cell focal adhesions protein, in cellular biology and various diseases. Zyxin can orchestrate the restructuring and dynamic alterations of the cellular cytoskeleton, which is involved in cell proliferation, adhesion, motility, and gene transcription. Aberrant zyxin expression is closely correlated with tumor cell activity and cardiac function in both tumorigenesis and cardiovascular diseases. Moreover, in fibrotic and inflammatory conditions, zyxin can modulate cellular functions and inflammatory responses. Therefore, a comprehensive understanding of zyxin is crucial for deciphering signal transduction networks and disease pathogenesis. Investigating its role in diseases holds promise for novel avenues in early diagnosis and therapeutic strategies. Nevertheless, targeting zyxin as a therapeutic focal point presents challenges in terms of specificity, safety, drug delivery, and resistance. Nonetheless, in-depth studies on zyxin and the application of precision medicine could offer new possibilities for personalized treatment modalities.
Collapse
Affiliation(s)
- Zelan Wu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Daiqin Wu
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qin Zhong
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xue Zou
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhongjing Liu
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hehua Long
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Jing Wei
- Department of Endocrinology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xia Li
- Guizhou Precision Medicine Institute, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fangjie Dai
- Department of Cardiovascular Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
13
|
Kliewe F, Siegerist F, Hammer E, Al-Hasani J, Amling TRJ, Hollemann JZE, Schindler M, Drenic V, Simm S, Amann K, Daniel C, Lindenmeyer M, Hecker M, Völker U, Endlich N. Zyxin is important for the stability and function of podocytes, especially during mechanical stretch. Commun Biol 2024; 7:446. [PMID: 38605154 PMCID: PMC11009394 DOI: 10.1038/s42003-024-06125-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Podocyte detachment due to mechanical stress is a common issue in hypertension-induced kidney disease. This study highlights the role of zyxin for podocyte stability and function. We have found that zyxin is significantly up-regulated in podocytes after mechanical stretch and relocalizes from focal adhesions to actin filaments. In zyxin knockout podocytes, we found that the loss of zyxin reduced the expression of vinculin and VASP as well as the expression of matrix proteins, such as fibronectin. This suggests that zyxin is a central player in the translation of mechanical forces in podocytes. In vivo, zyxin is highly up-regulated in patients suffering from diabetic nephropathy and in hypertensive DOCA-salt treated mice. Furthermore, zyxin loss in mice resulted in proteinuria and effacement of podocyte foot processes that was measured by super resolution microscopy. This highlights the essential role of zyxin for podocyte maintenance in vitro and in vivo, especially under mechanical stretch.
Collapse
Affiliation(s)
- Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany.
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jaafar Al-Hasani
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | | | | | - Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Vedran Drenic
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| | - Stefan Simm
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Kerstin Amann
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology; Friedrich-Alexander University (FAU) Erlangen-Nuremberg, Erlangen, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
- NIPOKA GmbH, Center of High-End Imaging, Greifswald, Germany
| |
Collapse
|
14
|
Troyanovsky RB, Indra I, Troyanovsky SM. Characterization of early and late events of adherens junction assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583373. [PMID: 38496678 PMCID: PMC10942379 DOI: 10.1101/2024.03.04.583373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Cadherins are transmembrane adhesion receptors. Cadherin ectodomains form adhesive 2D clusters through cooperative trans and cis interactions, whereas its intracellular region interacts with specific cytosolic proteins, termed catenins, to anchor the cadherin-catenin complex (CCC) to the actin cytoskeleton. How these two types of interactions are coordinated in the formation of specialized cell-cell adhesions, adherens junctions (AJ), remains unclear. We focus here on the role of the actin-binding domain of α-catenin (αABD) by showing that the interaction of αABD with actin generates actin-bound CCC oligomers (CCC/actin strands) incorporating up to six CCCs. The strands are primarily formed on the actin-rich cell protrusions. Once in cell-cell interface, the strands become involved in cadherin ectodomain clustering. Such combination of the extracellular and intracellular oligomerizations gives rise to the composite oligomers, trans CCC/actin clusters. To mature, these clusters then rearrange their actin filaments using several redundant pathways, two of which are characterized here: one depends on the α-catenin-associated protein, vinculin and the second one depends on the unstructured C-terminus of αABD. Thus, AJ assembly proceeds through spontaneous formation of trans CCC/actin clusters and their successive reorganization.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | - Indrajyoti Indra
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | - Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
- Department of Cell & Developmental Biology, The Feinberg School of Medicine, Chicago, IL 60614
| |
Collapse
|
15
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
16
|
Healy MD, Collins BM. The PDLIM family of actin-associated proteins and their emerging role in membrane trafficking. Biochem Soc Trans 2023; 51:2005-2016. [PMID: 38095060 PMCID: PMC10754285 DOI: 10.1042/bst20220804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
The PDZ and LIM domain (PDLIM) proteins are associated with the actin cytoskeleton and have conserved in roles in metazoan actin organisation and function. They primarily function as scaffolds linking various proteins to actin and its binding partner α-actinin via two conserved domains; an N-terminal postsynaptic density 95, discs large and zonula occludens-1 (PDZ) domain, and either single or multiple C-terminal LIN-11, Isl-1 and MEC-3 (LIM) domains in the actinin-associated LIM protein (ALP)- and Enigma-related proteins, respectively. While their role in actin organisation, such as in stress fibres or in the Z-disc of muscle fibres is well known, emerging evidence also suggests a role in actin-dependent membrane trafficking in the endosomal system. This is mediated by a recently identified interaction with the sorting nexin 17 (SNX17) protein, an adaptor for the trafficking complex Commander which is itself intimately linked to actin-directed formation of endosomal recycling domains. In this review we focus on the currently understood structural basis for PDLIM function. The PDZ domains mediate direct binding to distinct classes of PDZ-binding motifs (PDZbms), including α-actinin and other actin-associated proteins, and a highly specific interaction with the type III PDZbm such as the one found in the C-terminus of SNX17. The structures of the LIM domains are less well characterised and how they engage with their ligands is completely unknown. Despite the lack of experimental structural data, we find that recently developed machine learning-based structure prediction methods provide insights into their potential interactions and provide a template for further studies of their molecular functions.
Collapse
Affiliation(s)
- Michael D. Healy
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| | - Brett M. Collins
- The University of Queensland, Institute for Molecular Bioscience, St Lucia, Queensland 4072, Australia
| |
Collapse
|
17
|
He S, Zhang J, Liu Z, Wang Y, Hao X, Wang X, Zhou Z, Ye X, Zhao Y, Zhao Y, Wang R. Upregulated Cytoskeletal Proteins Promote Pathological Angiogenesis in Moyamoya Disease. Stroke 2023; 54:3153-3164. [PMID: 37886851 DOI: 10.1161/strokeaha.123.044476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Moyamoya disease (MMD) is a rare progressive vascular disease that leads to intracranial internal carotid artery stenosis and eventual occlusion. However, its pathogenesis remains unclear. The purpose of this study is to explore the role of abnormally expressed proteins in the pathogenesis of MMD. METHODS Data-independent acquisition mass spectrometry identifies the differentially expressed proteins in MMD serum by detecting the serum from 60 patients with MMD and 20 health controls. The differentially expressed proteins were validated using enzyme linked immunosorbent assays. Immunofluorescence for superficial temporal artery and middle cerebral artery specimens was used to explore the morphological changes of vascular wall in MMD. In vitro experiments were used to explore the changes and mechanisms of differentially expressed proteins on endothelial cells. RESULTS Proteomic analysis showed that a total of 14 726 peptides and 1555 proteins were quantified by mass spectrometry data. FLNA (filamin A) and ZYX (zyxin) proteins were significantly higher in MMD serum compared with those in health controls (Log2FC >2.9 and >2.8, respectively). Immunofluorescence revealed an intimal hyperplasia in superficial temporal artery and middle cerebral artery specimens of MMD. FLNA and ZYX proteins increased the proportion of endothelial cells in S phase and promoted their proliferation, angiogenesis, and cytoskeleton enlargement. Mechanistic studies revealed that AKT (serine/threonine kinase)/GSK-3β (glycogen synthase kinase 3β)/β-catenin signaling pathway plays a major role in these FLNA- and ZYX-induced changes in endothelial cells. CONCLUSIONS This study provides proteomic data on a large sample size of MMD. The differential expression of FLNA and ZYX in patient with MMD and following in vitro experiments suggest that these upregulated proteins are related to the pathology of cerebrovascular intimal hyperplasia in MMD and are involved in MMD pathogenesis, with diagnostic and therapeutic ramifications.
Collapse
Affiliation(s)
- Shihao He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.H., Yuanli Zhao, R.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (S.H., Yuanli Zhao)
| | - Junze Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Ziqi Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Yanru Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Xiaokuan Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Xilong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Zhenyu Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Yahui Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.H., Yuanli Zhao, R.W.)
- Center of Stroke, Beijing Institute for Brain Disorders, China (S.H., Yuanli Zhao)
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China (Yuanli Zhao, R.W.)
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China (S.H., J.Z., Z.L., Y.W., X.H., X.W., Z.Z., X.Y., Yahui Zhao, Yuanli Zhao, R.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing, China (S.H., Yuanli Zhao, R.W.)
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, China (Yuanli Zhao, R.W.)
| |
Collapse
|
18
|
CAI TIANYING, BAI JUNJIE, TAN PENG, HUANG ZHIWEI, LIU CHEN, WU ZIMING, CHENG YONGLANG, LI TONGXI, CHEN YIFAN, RUAN JIAN, GAO LIN, DU YICHAO, FU WENGUANG. Zyxin promotes hepatocellular carcinoma progression via the activation of AKT/mTOR signaling pathway. Oncol Res 2023; 31:805-817. [PMID: 37547758 PMCID: PMC10398406 DOI: 10.32604/or.2023.029549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/17/2023] [Indexed: 08/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy that is driven by multiple genes and pathways. The aim of this study was to investigate the role and specific mechanism of the actin-interacting protein zyxin (ZYX) in HCC. We found that the expression of ZYX was significantly higher in HCC tissues compared to that in normal liver tissues. In addition, overexpression of ZYX in hepatoma cell lines (PLC/PRF/5, HCCLM3) enhanced their proliferation, migration and invasion, whereas ZYX knockdown had the opposite effects (SK HEP-1, Huh-7). Furthermore, the change in the expression levels of ZYX also altered that of proteins related to cell cycle, migration and invasion. Similar results were obtained with xenograft models. The AKT/mTOR signaling pathway is one of the key mediators of cancer development. While ZYX overexpression upregulated the levels of phosphorylated AKT/mTOR proteins, its knockdown had the opposite effect. In addition, the AKT inhibitor MK2206 neutralized the pro-oncogenic effects of ZYX on the HCC cells, whereas the AKT activator SC79 restored the proliferation, migration and invasion of HCC cells with ZYX knockdown. Taken together, ZYX promotes the malignant progression of HCC by activating AKT/mTOR signaling pathway, and is a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- TIANYING CAI
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Biobank, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - JUNJIE BAI
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - PENG TAN
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - ZHIWEI HUANG
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - CHEN LIU
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - ZIMING WU
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - YONGLANG CHENG
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - TONGXI LI
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - YIFAN CHEN
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - JIAN RUAN
- Department of Medical Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - LIN GAO
- Department of Health Management, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - YICHAO DU
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - WENGUANG FU
- Department of Hepatobiliary Surgery, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
19
|
Wang X, Zhang C, Song H, Yuan J, Zhang X, Yuan Y, Zhang L, He J. Characterization of LIMA1 and its emerging roles and potential therapeutic prospects in cancers. Front Oncol 2023; 13:1115943. [PMID: 37274282 PMCID: PMC10235525 DOI: 10.3389/fonc.2023.1115943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Actin is the most abundant and highly conserved cytoskeletal protein present in all eukaryotic cells. Remodeling of the actin cytoskeleton is controlled by a variety of actin-binding proteins that are extensively involved in biological processes such as cell motility and maintenance of cell shape. LIM domain and actin-binding protein 1 (LIMA1), as an important actin cytoskeletal regulator, was initially thought to be a tumor suppressor frequently downregulated in epithelial tumors. Importantly, the deficiency of LIMA1 may be responsible for dysregulated cytoskeletal dynamics, altered cell motility and disrupted cell-cell adhesion, which promote tumor proliferation, invasion and migration. As research progresses, the roles of LIMA1 extend from cytoskeletal dynamics and cell motility to cell division, gene regulation, apical extrusion, angiogenesis, cellular metabolism and lipid metabolism. However, the expression of LIMA1 in malignant tumors and its mechanism of action have not yet been elucidated, and many problems and challenges remain to be addressed. Therefore, this review systematically describes the structure and biological functions of LIMA1 and explores its expression and regulatory mechanism in malignant tumors, and further discusses its clinical value and therapeutic prospects.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Chao Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Huangqin Song
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Junlong Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaomin Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yiran Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Lei Zhang
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Hepatic Surgery Center, Institute of Hepato-Pancreato-Biliary Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiefeng He
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
20
|
Sala S, Oakes PW. LIM domain proteins. Curr Biol 2023; 33:R339-R341. [PMID: 37160086 DOI: 10.1016/j.cub.2023.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Sala and Oakes introduce LIM domain proteins and discuss their roles in transcription, cytokinesis, adhesion, motility and mechanosignaling.
Collapse
Affiliation(s)
- Stefano Sala
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Patrick W Oakes
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA.
| |
Collapse
|
21
|
Troyanovsky SM. Adherens junction: the ensemble of specialized cadherin clusters. Trends Cell Biol 2023; 33:374-387. [PMID: 36127186 PMCID: PMC10020127 DOI: 10.1016/j.tcb.2022.08.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
The cell-cell connections in adherens junctions (AJs) are mediated by transmembrane receptors, type I cadherins (referred to here as cadherins). These cadherin-based connections (or trans bonds) are weak. To upregulate their strength, cadherins exploit avidity, the increased affinity of binding between cadherin clusters compared with isolated monomers. Formation of such clusters is a unique molecular process that is driven by a synergy of direct and indirect cis interactions between cadherins located at the same cell. In addition to their role in adhesion, cadherin clusters provide structural scaffolds for cytosolic proteins, which implicate cadherin into different cellular activities and signaling pathways. The cluster lifetime, which depends on the actin cytoskeleton, and on the mechanical forces it generates, determines the strength of AJs and their plasticity. The key aspects of cadherin adhesion, therefore, cannot be understood at the level of isolated cadherin molecules, but should be discussed in the context of cadherin clusters.
Collapse
Affiliation(s)
- Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Cell and Molecular Biology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
22
|
Actin crosslinking by α-actinin averts viscous dissipation of myosin force transmission in stress fibers. iScience 2023; 26:106090. [PMID: 36852278 PMCID: PMC9958379 DOI: 10.1016/j.isci.2023.106090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Contractile force generated in actomyosin stress fibers (SFs) is transmitted along SFs to the extracellular matrix (ECM), which contributes to cell migration and sensing of ECM rigidity. In this study, we show that efficient force transmission along SFs relies on actin crosslinking by α-actinin. Upon reduction of α-actinin-mediated crosslinks, the myosin II activity induced flows of actin filaments and myosin II along SFs, leading to a decrease in traction force exertion to ECM. The fluidized SFs maintained their cable integrity probably through enhanced actin polymerization throughout SFs. A computational modeling analysis suggested that lowering the density of actin crosslinks caused viscous slippage of actin filaments in SFs and, thereby, dissipated myosin-generated force transmitting along SFs. As a cellular scale outcome, α-actinin depletion attenuated the ECM-rigidity-dependent difference in cell migration speed, which suggested that α-actinin-modulated SF mechanics is involved in the cellular response to ECM rigidity.
Collapse
|
23
|
Martynova NY, Parshina EA, Zaraisky AG. Cytoskeletal protein Zyxin in embryonic development: from controlling cell movements and pluripotency to regulating embryonic patterning. FEBS J 2023; 290:66-72. [PMID: 34854244 DOI: 10.1111/febs.16308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/29/2021] [Accepted: 11/30/2021] [Indexed: 01/14/2023]
Abstract
The Lim-domain protein Zyxin was initially identified as a minor actin cytoskeleton protein that regulates the assembly and repair of actin filaments. At the same time, additional functions revealed for Zyxin in recent decades indicate that this protein can also play an important role in regulating gene expression and cell differentiation. In this review, we analysed the data in the literature pointing to Zyxin as one of the possible molecular hubs linking morphogenetic cell movements with gene expression, stem cell status regulation and pattern formation during the most complex processes in organism life, embryogenesis.
Collapse
Affiliation(s)
- Natalia Y Martynova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Elena A Parshina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Andrey G Zaraisky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
24
|
Lynch AM, Zhu Y, Lucas BG, Winkelman JD, Bai K, Martin SCT, Block S, Slabodnick MM, Audhya A, Goldstein B, Pettitt J, Gardel ML, Hardin J. TES-1/Tes and ZYX-1/Zyxin protect junctional actin networks under tension during epidermal morphogenesis in the C. elegans embryo. Curr Biol 2022; 32:5189-5199.e6. [PMID: 36384139 PMCID: PMC9729467 DOI: 10.1016/j.cub.2022.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/12/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022]
Abstract
LIM-domain-containing repeat (LCR) proteins are recruited to strained actin filaments within stress fibers in cultured cells,1,2,3 but their roles at cell-cell junctions in living organisms have not been extensively studied. Here, we show that the Caenorhabditis elegans LCR proteins TES-1/Tes and ZYX-1/Zyxin are recruited to apical junctions during embryonic elongation when junctions are under tension. In genetic backgrounds in which embryonic elongation fails, junctional recruitment is severely compromised. The two proteins display complementary patterns of expression: TES-1 is expressed in lateral (seam) epidermal cells, whereas ZYX-1 is expressed in dorsal and ventral epidermal cells. tes-1 and zyx-1 mutant embryos display junctional F-actin defects. The loss of either protein strongly enhances morphogenetic defects in hypomorphic mutant backgrounds for cadherin/catenin complex (CCC) components. The LCR regions of TES-1 and ZYX-1 are recruited to stress fiber strain sites (SFSSs) in cultured vertebrate cells. Together, these data establish TES-1 and ZYX-1 as components of a multicellular, tension-sensitive system that stabilizes the junctional actin cytoskeleton during embryonic morphogenesis.
Collapse
Affiliation(s)
- Allison M Lynch
- Program in Genetics, University of Wisconsin, Madison, WI 53706, USA
| | - Yuyun Zhu
- Program in Genetics, University of Wisconsin, Madison, WI 53706, USA
| | - Bethany G Lucas
- Department of Biology, Regis University, 3333 Regis Boulevard, Denver, CO 80221, USA
| | - Jonathan D Winkelman
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Keliya Bai
- University of Aberdeen, Institute of Medical Sciences, Aberdeen AB25 2ZD, UK
| | | | - Samuel Block
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53706, USA
| | - Mark M Slabodnick
- Department of Biology, Knox University, Galesburg, IL 61401, USA; Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53706, USA
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jonathan Pettitt
- University of Aberdeen, Institute of Medical Sciences, Aberdeen AB25 2ZD, UK
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA; Department of Physics, James Franck Institute and Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Jeff Hardin
- Program in Genetics, University of Wisconsin, Madison, WI 53706, USA; Biophysics Program, University of Wisconsin, Madison, WI 53706, USA; Department of Integrative Biology, University of Wisconsin, Madison, WI 53706, USA.
| |
Collapse
|
25
|
Liu S, Kanchanawong P. Emerging interplay of cytoskeletal architecture, cytomechanics and pluripotency. J Cell Sci 2022; 135:275761. [PMID: 35726598 DOI: 10.1242/jcs.259379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pluripotent stem cells (PSCs) are capable of differentiating into all three germ layers and trophoblasts, whereas tissue-specific adult stem cells have a more limited lineage potency. Although the importance of the cytoskeletal architecture and cytomechanical properties in adult stem cell differentiation have been widely appreciated, how they contribute to mechanotransduction in PSCs is less well understood. Here, we discuss recent insights into the interplay of cellular architecture, cell mechanics and the pluripotent states of PSCs. Notably, the distinctive cytomechanical and morphodynamic profiles of PSCs are accompanied by a number of unique molecular mechanisms. The extent to which such mechanobiological signatures are intertwined with pluripotency regulation remains an open question that may have important implications in developmental morphogenesis and regenerative medicine.
Collapse
Affiliation(s)
- Shiying Liu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore 117411, Republic of Singapore
| |
Collapse
|
26
|
Partynska A, Gomulkiewicz A, Piotrowska A, Grzegrzolka J, Rzechonek A, Ratajczak-Wielgomas K, Podhorska-Okolow M, Dziegiel P. Expression of Zyxin in Non-Small Cell Lung Cancer-A Preliminary Study. Biomolecules 2022; 12:biom12060827. [PMID: 35740950 PMCID: PMC9221212 DOI: 10.3390/biom12060827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/09/2022] Open
Abstract
Background: The potential involvement of zyxin (ZYX) in carcinogenesis has been investigated in many cancer types. However, there are a limited number of studies on the role of ZYX in the progression of non-small cell lung cancer (NSCLC). Since lung cancer is one of the most frequently diagnosed carcinomas, the aim of our study was to determine the localization and expression levels of ZYX in NSCLC and to correlate the results with the clinicopathological data. Materials and Methods: The expression of ZYX was assessed in NSCLC cases and in cell lines representing this tumor type. Levels of ZYX were determined in the clinical material using immunohistochemistry (IHC) and Western Blot. Real-time PCR was used to assess ZYX mRNA levels. The expression of ZYX was also checked in NSCLC cell lines using real-time PCR, Western Blot, and immunofluorescence/immunocytochemistry. Results: The results showed lower levels of ZYX in NSCLC cells compared with control tissues. This trend was observed at the protein and mRNA levels. The assays on the NSCLC model also demonstrated lower levels of ZYX in cancer cells compared with control cells. Conclusions: The decreased expression of ZYX in NSCLC may indicate a suppressor role of this protein in NSCLC.
Collapse
Affiliation(s)
- Aleksandra Partynska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.G.); (A.P.); (J.G.); (K.R.-W.); (P.D.)
- Correspondence:
| | - Agnieszka Gomulkiewicz
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.G.); (A.P.); (J.G.); (K.R.-W.); (P.D.)
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.G.); (A.P.); (J.G.); (K.R.-W.); (P.D.)
| | - Jedrzej Grzegrzolka
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.G.); (A.P.); (J.G.); (K.R.-W.); (P.D.)
| | - Adam Rzechonek
- Department of Thoracic Surgery, Wroclaw Medical University, 53-439 Wroclaw, Poland;
| | - Katarzyna Ratajczak-Wielgomas
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.G.); (A.P.); (J.G.); (K.R.-W.); (P.D.)
| | - Marzenna Podhorska-Okolow
- Division of Ultrastructural Research, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Piotr Dziegiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (A.G.); (A.P.); (J.G.); (K.R.-W.); (P.D.)
- Division of Human Biology, Faculty of Physiotherapy, University School of Physical Education in Wroclaw, 51-612 Wroclaw, Poland
| |
Collapse
|
27
|
Parshina EA, Orlov EE, Zaraisky AG, Martynova NY. The Cytoskeletal Protein Zyxin Inhibits Retinoic Acid Signaling by Destabilizing the Maternal mRNA of the RXRγ Nuclear Receptor. Int J Mol Sci 2022; 23:5627. [PMID: 35628438 PMCID: PMC9147113 DOI: 10.3390/ijms23105627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023] Open
Abstract
Zyxin is an LIM-domain-containing protein that regulates the assembly of F-actin filaments in cell contacts. Additionally, as a result of mechanical stress, Zyxin can enter nuclei and regulate gene expression. Previously, we found that Zyxin could affect mRNA stability of the maternally derived stemness factors of Pou5f3 family in Xenopus laevis embryos through binding to Y-box factor1. In the present work, we demonstrate that Zyxin can also affect mRNA stability of the maternally derived retinoid receptor Rxrγ through the same mechanism. Moreover, we confirmed the functional link between Zyxin and Rxrγ-dependent gene expression. As a result, Zyxin appears to play an essential role in the regulation of the retinoic acid signal pathway during early embryonic development. Besides, our research indicates that the mechanism based on the mRNA destabilization by Zyxin may take part in the control of the expression of a fairly wide range of maternal genes.
Collapse
Affiliation(s)
- Elena A. Parshina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (E.E.O.); (A.G.Z.)
| | - Eugeny E. Orlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (E.E.O.); (A.G.Z.)
| | - Andrey G. Zaraisky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (E.E.O.); (A.G.Z.)
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Natalia Y. Martynova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (E.E.O.); (A.G.Z.)
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
28
|
Impact of Treadmill Interval Running on the Appearance of Zinc Finger Protein FHL2 in Bone Marrow Cells in a Rat Model: A Pilot Study. Life (Basel) 2022; 12:life12040528. [PMID: 35455019 PMCID: PMC9029125 DOI: 10.3390/life12040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Although the benefits of physical exercise to preserve bone quality are now widely recognized, the intimate mechanisms leading to the underlying cell responses still require further investigations. Interval training running, for instance, appears as a generator of impacts on the skeleton, and particularly on the progenitor cells located in the bone marrow. Therefore, if this kind of stimulus initiates bone cell proliferation and differentiation, the activation of a devoted signaling pathway by mechano-transduction seems likely. This study aimed at investigating the effects of an interval running program on the appearance of the zinc finger protein FHL2 in bone cells and their anatomical location. Twelve 5-week-old male Wistar rats were randomly allocated to one of the following groups (n = 6 per group): sedentary control (SED) or high-intensity interval running (EX, 8 consecutive weeks). FHL2 identification in bone cells was performed by immuno-histochemistry on serial sections of radii. We hypothesized that impacts generated by running could activate, in vivo, a specific signaling pathway, through an integrin-mediated mechano-transductive process, leading to the synthesis of FHL2 in bone marrow cells. Our data demonstrated the systematic appearance of FHL2 (% labeled cells: 7.5%, p < 0.001) in bone marrow obtained from EX rats, whereas no FHL2 was revealed in SED rats. These results suggest that the mechanical impacts generated during high-intensity interval running activate a signaling pathway involving nuclear FHL2, such as that also observed with dexamethasone administration. Consequently, interval running could be proposed as a non-pharmacological strategy to contribute to bone marrow cell osteogenic differentiation.
Collapse
|
29
|
Snider CE, Bhattacharjee R, Igarashi MG, Gould KL. Fission yeast paxillin contains two Cdc15 binding motifs for robust recruitment to the cytokinetic ring. Mol Biol Cell 2022; 33:br4. [PMID: 35108037 PMCID: PMC9250355 DOI: 10.1091/mbc.e21-11-0560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The F-BAR protein Cdc15 mediates attachment of the cytokinetic ring (CR) to the plasma membrane and is essential for cytokinesis in Schizosaccharomyces pombe. While its N-terminal F-BAR domain is responsible for oligomerization and membrane binding, its C-terminal SH3 domain binds other partners at a distance from the membrane. We previously demonstrated that the essential cytokinetic formin Cdc12, through an N-terminal motif, directly binds the cytosolic face of the F-BAR domain. Here, we show that paxillin-like Pxl1, which is important for CR stability, contains a motif highly related to that in formin Cdc12, and also binds the Cdc15 F-BAR domain directly. Interestingly, Pxl1 has a second site for binding the Cdc15 SH3 domain. To understand the importance of these two Pxl1-Cdc15 interactions, we mapped and disrupted both. Disrupting the Pxl1-Cdc15 F-BAR domain interaction reduced Pxl1 levels in the CR, whereas disrupting Pxl1’s interaction with the Cdc15 SH3 domain, did not. Unexpectedly, abolishing Pxl1-Cdc15 interaction greatly reduced but did not eliminate CR Pxl1 and did not significantly affect cytokinesis. These data point to another mechanism of Pxl1 CR recruitment and show that very little CR Pxl1 is sufficient for its cytokinetic function.
Collapse
Affiliation(s)
- Chloe E Snider
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rahul Bhattacharjee
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Maya G Igarashi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kathleen L Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
30
|
Zhao Y, Yue S, Zhou X, Guo J, Ma S, Chen Q. O-GlcNAc transferase promotes the nuclear localization of the focal adhesion-associated protein Zyxin to regulate UV-induced cell death. J Biol Chem 2022; 298:101776. [PMID: 35227760 PMCID: PMC8988012 DOI: 10.1016/j.jbc.2022.101776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022] Open
Abstract
Zyxin is a zinc-binding phosphoprotein known to regulate cell migration, adhesion, and cell survival. Zyxin also plays a role in signal transduction between focal adhesions and the nuclear compartment. However, the mechanism of Zyxin shuttling to nucleus is still unclear. Here, we identify that the GlcNAc transferase (O-linked GlcNAc [O-GlcNAc] transferase) can O-GlcNAcylate Zyxin and regulate its nuclear localization. We show that O-GlcNAc transferase O-GlcNAcylates Zyxin at two residues, serine 169 (Ser-169) and Ser-246. In addition, O-GlcNAcylation of Ser-169, but not Ser-246, enhances its interaction with 14-3-3γ, which is a phosphoserine/threonine-binding protein and is reported to bind with phosphorylated Zyxin. Furthermore, we found that 14-3-3γ could promote the nuclear localization of Zyxin after Ser-169 O-GlcNAcylation by affecting the function of the N-terminal nuclear export signal sequence; functionally, UV treatment increases the O-GlcNAcylation of Zyxin, which may enhance the nuclear location of Zyxin. Finally, Zyxin in the nucleus maintains homeodomain-interacting protein kinase 2 stability and promotes UV-induced cell death. In conclusion, we uncover that the nuclear localization of Zyxin can be regulated by its O-GlcNAcylation, and that this protein may regulate UV-induced cell death.
Collapse
|
31
|
Balestra T, Manara MC, Laginestra MA, Pasello M, De Feo A, Bassi C, Guerzoni C, Landuzzi L, Lollini PL, Donati DM, Negrini M, Magnani M, Scotlandi K. Targeting CD99 Compromises the Oncogenic Effects of the Chimera EWS-FLI1 by Inducing Reexpression of Zyxin and Inhibition of GLI1 Activity. Mol Cancer Ther 2022; 21:58-69. [PMID: 34667115 DOI: 10.1158/1535-7163.mct-21-0189] [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: 03/02/2021] [Revised: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022]
Abstract
Ewing sarcoma, a highly aggressive pediatric tumor, is driven by EWS-FLI1, an oncogenic transcription factor that remodels the tumor genetic landscape. Epigenetic mechanisms play a pivotal role in Ewing sarcoma pathogenesis, and the therapeutic value of compounds targeting epigenetic pathways is being identified in preclinical models. Here, we showed that modulation of CD99, a cell surface molecule highly expressed in Ewing sarcoma cells, may alter transcriptional dysregulation in Ewing sarcoma through control of the zyxin-GLI1 axis. Zyxin is transcriptionally repressed, but GLI1 expression is maintained by EWS-FLI1. We demonstrated that targeting CD99 with antibodies, including the human diabody C7, or genetically inhibiting CD99 is sufficient to increase zyxin expression and induce its dynamic nuclear accumulation. Nuclear zyxin functionally affects GLI1, inhibiting targets such as NKX2-2, cyclin D1, and PTCH1 and upregulating GAS1, a tumor suppressor protein negatively regulated by SHH/GLI1 signaling. We used a battery of functional assays to demonstrate (i) the relationship between CD99/zyxin and tumor cell growth/migration and (ii) how CD99 deprivation from the Ewing sarcoma cell surface is sufficient to specifically affect the expression of some crucial EWS-FLI1 targets, both in vitro and in vivo, even in the presence of EWS-FLI1. This article reveals that the CD99/zyxin/GLI1 axis is promising therapeutic target for reducing Ewing sarcoma malignancy.
Collapse
Affiliation(s)
- Tommaso Balestra
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Maria Cristina Manara
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Michela Pasello
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandra De Feo
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Cristian Bassi
- Department of Translational Medicine and for Romagna, and "Laboratorio per le Tecnologie delle Terapie Avanzate" (LTTA), University of Ferrara, Ferrara, Italy
| | - Clara Guerzoni
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Lorena Landuzzi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Pier-Luigi Lollini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Davide Maria Donati
- Clinica Ortopedica III, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Massimo Negrini
- Department of Translational Medicine and for Romagna, and "Laboratorio per le Tecnologie delle Terapie Avanzate" (LTTA), University of Ferrara, Ferrara, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino, Fano, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|
32
|
Valencia FR, Sandoval E, Du J, Iu E, Liu J, Plotnikov SV. Force-dependent activation of actin elongation factor mDia1 protects the cytoskeleton from mechanical damage and promotes stress fiber repair. Dev Cell 2021; 56:3288-3302.e5. [PMID: 34822787 DOI: 10.1016/j.devcel.2021.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/02/2021] [Accepted: 11/02/2021] [Indexed: 01/16/2023]
Abstract
Plasticity of cell mechanics underlies a wide range of cell and tissue behaviors allowing cells to migrate through narrow spaces, resist shear forces, and safeguard against mechanical damage. Such plasticity depends on spatiotemporal regulation of the actomyosin cytoskeleton, but mechanisms of adaptive change in cell mechanics remain elusive. Here, we report a mechanism of mechanically activated actin polymerization at focal adhesions (FAs), specifically requiring the actin elongation factor mDia1. By combining live-cell imaging with mathematical modeling, we show that actin polymerization at FAs exhibits pulsatile dynamics where spikes of mDia1 activity are triggered by contractile forces. The suppression of mDia1-mediated actin polymerization increases tension on stress fibers (SFs) leading to an increased frequency of spontaneous SF damage and decreased efficiency of zyxin-mediated SF repair. We conclude that tension-controlled actin polymerization acts as a safety valve dampening excessive tension on the actin cytoskeleton and safeguarding SFs against mechanical damage.
Collapse
Affiliation(s)
- Fernando R Valencia
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Eduardo Sandoval
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joy Du
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Ernest Iu
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jian Liu
- Center for Cell Dynamics, Department of Cell Biology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
33
|
Essential role of zyxin in platelet biogenesis and glycoprotein Ib-IX surface expression. Cell Death Dis 2021; 12:955. [PMID: 34657146 PMCID: PMC8520529 DOI: 10.1038/s41419-021-04246-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
Platelets are generated from the cytoplasm of megakaryocytes (MKs) via actin cytoskeleton reorganization. Zyxin is a focal adhesion protein and wildly expressed in eukaryotes to regulate actin remodeling. Zyxin is upregulated during megakaryocytic differentiation; however, the role of zyxin in thrombopoiesis is unknown. Here we show that zyxin ablation results in profound macrothrombocytopenia. Platelet lifespan and thrombopoietin level were comparable between wild-type and zyxin-deficient mice, but MK maturation, demarcation membrane system formation, and proplatelet generation were obviously impaired in the absence of zyxin. Differential proteomic analysis of proteins associated with macrothrombocytopenia revealed that glycoprotein (GP) Ib-IX was significantly reduced in zyxin-deficient platelets. Moreover, GPIb-IX surface level was decreased in zyxin-deficient MKs. Knockdown of zyxin in a human megakaryocytic cell line resulted in GPIbα degradation by lysosomes leading to the reduction of GPIb-IX surface level. We further found that zyxin was colocalized with vasodilator-stimulated phosphoprotein (VASP), and loss of zyxin caused diffuse distribution of VASP and actin cytoskeleton disorganization in both platelets and MKs. Reconstitution of zyxin with VASP binding site in zyxin-deficient hematopoietic progenitor cell-derived MKs restored GPIb-IX surface expression and proplatelet generation. Taken together, our findings identify zyxin as a regulator of platelet biogenesis and GPIb-IX surface expression through VASP-mediated cytoskeleton reorganization, suggesting possible pathogenesis of macrothrombocytopenia.
Collapse
|
34
|
Abstract
TRIP6, a member of the ZYXIN-family of LIM domain proteins, is a focal adhesion component. Trip6 deletion in the mouse, reported here, reveals a function in the brain: ependymal and choroid plexus epithelial cells are carrying, unexpectedly, fewer and shorter cilia, are poorly differentiated, and the mice develop hydrocephalus. TRIP6 carries numerous protein interaction domains and its functions require homodimerization. Indeed, TRIP6 disruption in vitro (in a choroid plexus epithelial cell line), via RNAi or inhibition of its homodimerization, confirms its function in ciliogenesis. Using super-resolution microscopy, we demonstrate TRIP6 localization at the pericentriolar material and along the ciliary axoneme. The requirement for homodimerization which doubles its interaction sites, its punctate localization along the axoneme, and its co-localization with other cilia components suggest a scaffold/co-transporter function for TRIP6 in cilia. Thus, this work uncovers an essential role of a LIM-domain protein assembly factor in mammalian ciliogenesis.
Collapse
|
35
|
Kamperman T, Henke S, Crispim JF, Willemen NGA, Dijkstra PJ, Lee W, Offerhaus HL, Neubauer M, Smink AM, de Vos P, de Haan BJ, Karperien M, Shin SR, Leijten J. Tethering Cells via Enzymatic Oxidative Crosslinking Enables Mechanotransduction in Non-Cell-Adhesive Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102660. [PMID: 34476848 PMCID: PMC8530967 DOI: 10.1002/adma.202102660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/10/2021] [Indexed: 05/14/2023]
Abstract
Cell-matrix interactions govern cell behavior and tissue function by facilitating transduction of biomechanical cues. Engineered tissues often incorporate these interactions by employing cell-adhesive materials. However, using constitutively active cell-adhesive materials impedes control over cell fate and elicits inflammatory responses upon implantation. Here, an alternative cell-material interaction strategy that provides mechanotransducive properties via discrete inducible on-cell crosslinking (DOCKING) of materials, including those that are inherently non-cell-adhesive, is introduced. Specifically, tyramine-functionalized materials are tethered to tyrosines that are naturally present in extracellular protein domains via enzyme-mediated oxidative crosslinking. Temporal control over the stiffness of on-cell tethered 3D microniches reveals that DOCKING uniquely enables lineage programming of stem cells by targeting adhesome-related mechanotransduction pathways acting independently of cell volume changes and spreading. In short, DOCKING represents a bioinspired and cytocompatible cell-tethering strategy that offers new routes to study and engineer cell-material interactions, thereby advancing applications ranging from drug delivery, to cell-based therapy, and cultured meat.
Collapse
Affiliation(s)
- Tom Kamperman
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
- Division of Engineering in MedicineBrigham and Women's HospitalHarvard Medical School65 Landsdowne StreetCambridgeMA02139USA
| | - Sieger Henke
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - João F. Crispim
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Niels G. A. Willemen
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Pieter J. Dijkstra
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Wooje Lee
- Optical SciencesMESA+ Institute for NanotechnologyUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Herman L. Offerhaus
- Optical SciencesMESA+ Institute for NanotechnologyUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Martin Neubauer
- Physical Chemistry IIUniversity of BayreuthUniversitätsstrasse 30D‐95447BayreuthGermany
| | - Alexandra M. Smink
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Paul de Vos
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Bart J. de Haan
- Department of Pathology and Medical BiologySection of ImmunoendocrinologyUniversity of GroningenUniversity Medical Center GroningenHanzeplein 1 (EA11)Groningen9713 GZThe Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Su Ryon Shin
- Division of Engineering in MedicineBrigham and Women's HospitalHarvard Medical School65 Landsdowne StreetCambridgeMA02139USA
| | - Jeroen Leijten
- Department of Developmental BioEngineeringFaculty of Science and TechnologyTechnical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| |
Collapse
|
36
|
Sun H, Zhou R, Zheng Y, Wen Z, Zhang D, Zeng D, Wu J, Huang Z, Rong X, Huang N, Sun L, Bin J, Liao Y, Shi M, Liao W. CRIP1 cooperates with BRCA2 to drive the nuclear enrichment of RAD51 and to facilitate homologous repair upon DNA damage induced by chemotherapy. Oncogene 2021; 40:5342-5355. [PMID: 34262130 PMCID: PMC8390368 DOI: 10.1038/s41388-021-01932-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 06/09/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023]
Abstract
Homologous recombination (HR) repair is an important determinant of chemosensitivity. However, the mechanisms underlying HR regulation remain largely unknown. Cysteine-rich intestinal protein 1 (CRIP1) is a member of the LIM/double-zinc finger protein family and is overexpressed and associated with prognosis in several tumor types. However, to date, the functional role of CRIP1 in cancer biology is poorly understood. Here we found that CRIP1 downregulation causes HR repair deficiency with concomitant increase in cell sensitivity to cisplatin, epirubicin, and the poly ADP-ribose polymerase (PARP) inhibitor olaparib in gastric cancer cells. Mechanistically, upon DNA damage, CRIP1 is deubiquitinated and upregulated by activated AKT signaling. CRIP1, in turn, promotes nuclear enrichment of RAD51, which is a prerequisite step for HR commencement, by stabilizing BRCA2 to counteract FBXO5-targeted RAD51 degradation and by binding to the core domain of RAD51 (RAD51184-257) in coordination with BRCA2, to facilitate nuclear export signal masking interactions between BRCA2 and RAD51. Moreover, through mass spectrometry screening, we found that KPNA4 is at least one of the carriers controlling the nucleo-cytoplasmic distribution of the CRIP1-BRCA2-RAD51 complex in response to chemotherapy. Consistent with these findings, RAD51 inhibitors block the CRIP1-mediated HR process, thereby restoring chemotherapy sensitivity of gastric cancer cells with high CRIP1 expression. Analysis of patient specimens revealed an abnormally high level of CRIP1 expression in GC tissues compared to that in the adjacent normal mucosa and a significant negative association between CRIP1 expression and survival time in patient cohorts with different types of solid tumors undergoing genotoxic treatments. In conclusion, our study suggests an essential function of CRIP1 in promoting HR repair and facilitating gastric cancer cell adaptation to genotoxic therapy.
Collapse
Affiliation(s)
- Huiying Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Yannan Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zhaowei Wen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Dingling Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Zhenhua Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jianping Bin
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
37
|
Zeng J, Li M, Shi H, Guo J. Upregulation of FGD6 Predicts Poor Prognosis in Gastric Cancer. Front Med (Lausanne) 2021; 8:672595. [PMID: 34291059 PMCID: PMC8288026 DOI: 10.3389/fmed.2021.672595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/01/2021] [Indexed: 12/21/2022] Open
Abstract
Background: The aim of this study was to investigate the prognostic significance of faciogenital dysplasia 6 (FGD6) in gastric cancer (GC). Methods: The data of GC patients from The Cancer Genome Atlas (TCGA) database were used for the primary study. Then, our data were validated by the GEO database and RuiJin cohort. The relationship between the FGD6 level and various clinicopathological features was analyzed by logistic regression and univariate Cox regression. Multivariate Cox regression analysis was used to evaluate whether FGD6 was an independent prognostic factor for survival of patients with GC. The relationship between FGD6 and overall survival time was explored by the Kaplan–Meier method. In addition, gene set enrichment analysis (GSEA) was performed to investigate the possible biological processes of FGD6. Results: The FGD6 level was significantly overexpressed in GC tissues, compared with adjacent normal tissues. The high expression of FGD6 was related to a high histological grade, stage, and T classification and poor prognosis of GC. Multivariate Cox regression analysis showed that FGD6 was an independent prognostic factor for survival of patients with GC. GSEA identified that the high expression of FGD6 was mainly enriched in regulation of actin cytoskeleton. Conclusion: FGD6 may be a prognostic biomarker for predicting the outcome of patients with GC.
Collapse
Affiliation(s)
- Jianmin Zeng
- The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Man Li
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huasheng Shi
- Medical College, Qingdao University, Qingdao, China
| | - Jianhui Guo
- Second Department of General Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
38
|
Norizadeh Abbariki T, Gonda Z, Kemler D, Urbanek P, Wagner T, Litfin M, Wang ZQ, Herrlich P, Kassel O. The LIM domain protein nTRIP6 modulates the dynamics of myogenic differentiation. Sci Rep 2021; 11:12904. [PMID: 34145356 PMCID: PMC8213751 DOI: 10.1038/s41598-021-92331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 06/02/2021] [Indexed: 11/11/2022] Open
Abstract
The process of myogenesis which operates during skeletal muscle regeneration involves the activation of muscle stem cells, the so-called satellite cells. These then give rise to proliferating progenitors, the myoblasts which subsequently exit the cell cycle and differentiate into committed precursors, the myocytes. Ultimately, the fusion of myocytes leads to myofiber formation. Here we reveal a role for the transcriptional co-regulator nTRIP6, the nuclear isoform of the LIM-domain protein TRIP6, in the temporal control of myogenesis. In an in vitro model of myogenesis, the expression of nTRIP6 is transiently up-regulated at the transition between proliferation and differentiation, whereas that of the cytosolic isoform TRIP6 is not altered. Selectively blocking nTRIP6 function results in accelerated early differentiation followed by deregulated late differentiation and fusion. Thus, the transient increase in nTRIP6 expression appears to prevent premature differentiation. Accordingly, knocking out the Trip6 gene in satellite cells leads to deregulated skeletal muscle regeneration dynamics in the mouse. Thus, dynamic changes in nTRIP6 expression contributes to the temporal control of myogenesis.
Collapse
Affiliation(s)
- Tannaz Norizadeh Abbariki
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Zita Gonda
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Denise Kemler
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Pavel Urbanek
- Leibniz Institute for Age Research (Fritz Lipmann Institute, FLI), Jena, Germany
| | - Tabea Wagner
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Margarethe Litfin
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Zhao-Qi Wang
- Leibniz Institute for Age Research (Fritz Lipmann Institute, FLI), Jena, Germany
| | - Peter Herrlich
- Leibniz Institute for Age Research (Fritz Lipmann Institute, FLI), Jena, Germany
| | - Olivier Kassel
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
39
|
Sala S, Oakes PW. Stress fiber strain recognition by the LIM protein testin is cryptic and mediated by RhoA. Mol Biol Cell 2021; 32:1758-1771. [PMID: 34038160 PMCID: PMC8684727 DOI: 10.1091/mbc.e21-03-0156] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The actin cytoskeleton is a key regulator of mechanical processes in cells. The family of LIM domain proteins have recently emerged as important mechanoresponsive cytoskeletal elements capable of sensing strain in the actin cytoskeleton. The mechanisms regulating this mechanosensitive behavior, however, remain poorly understood. Here we show that the LIM domain protein testin is peculiar in that despite the full-length protein primarily appearing diffuse in the cytoplasm, the C-terminal LIM domains alone recognize focal adhesions and strained actin, while the N-terminal domains alone recognize stress fibers. Phosphorylation mutations in the dimerization regions of testin, however, reveal its mechanosensitivity and cause it to relocate to focal adhesions and sites of strain in the actin cytoskeleton. Finally, we demonstrate that activated RhoA causes testin to adorn stress fibers and become mechanosensitive. Together, our data show that testin’s mechanoresponse is regulated in cells and provide new insights into LIM domain protein recognition of the actin cytoskeleton’s mechanical state.
Collapse
Affiliation(s)
- Stefano Sala
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153
| | - Patrick W Oakes
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153
| |
Collapse
|
40
|
Anderson CA, Kovar DR, Gardel ML, Winkelman JD. LIM domain proteins in cell mechanobiology. Cytoskeleton (Hoboken) 2021; 78:303-311. [PMID: 34028199 DOI: 10.1002/cm.21677] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
The actin cytoskeleton is important for maintaining mechanical homeostasis in adherent cells, largely through its regulation of adhesion and cortical tension. The LIM (Lin-11, Isl1, MEC-3) domain-containing proteins are involved in a myriad of cellular mechanosensitive pathways. Recent work has discovered that LIM domains bind to mechanically stressed actin filaments, suggesting a novel and widely conserved mechanism of mechanosensing. This review summarizes the current state of knowledge of LIM protein mechanosensitivity.
Collapse
Affiliation(s)
- Caitlin A Anderson
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA.,Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA.,James Franck Institute, University of Chicago, Chicago, Illinois, USA.,Department of Physics, University of Chicago, Chicago, Illinois, USA.,Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Jonathan D Winkelman
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
41
|
Vincenzi M, Mercurio FA, Leone M. Protein Interaction Domains: Structural Features and Drug Discovery Applications (Part 2). Curr Med Chem 2021; 28:854-892. [PMID: 31942846 DOI: 10.2174/0929867327666200114114142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Proteins present a modular organization made up of several domains. Apart from the domains playing catalytic functions, many others are crucial to recruit interactors. The latter domains can be defined as "PIDs" (Protein Interaction Domains) and are responsible for pivotal outcomes in signal transduction and a certain array of normal physiological and disease-related pathways. Targeting such PIDs with small molecules and peptides able to modulate their interaction networks, may represent a valuable route to discover novel therapeutics. OBJECTIVE This work represents a continuation of a very recent review describing PIDs able to recognize post-translationally modified peptide segments. On the contrary, the second part concerns with PIDs that interact with simple peptide sequences provided with standard amino acids. METHODS Crucial structural information on different domain subfamilies and their interactomes was gained by a wide search in different online available databases (including the PDB (Protein Data Bank), the Pfam (Protein family), and the SMART (Simple Modular Architecture Research Tool)). Pubmed was also searched to explore the most recent literature related to the topic. RESULTS AND CONCLUSION PIDs are multifaceted: they have all diverse structural features and can recognize several consensus sequences. PIDs can be linked to different diseases onset and progression, like cancer or viral infections and find applications in the personalized medicine field. Many efforts have been centered on peptide/peptidomimetic inhibitors of PIDs mediated interactions but much more work needs to be conducted to improve drug-likeness and interaction affinities of identified compounds.
Collapse
Affiliation(s)
- Marian Vincenzi
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| | - Flavia Anna Mercurio
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| | - Marilisa Leone
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via Mezzocannone 16, 80134 Naples, Italy
| |
Collapse
|
42
|
Wang L, Sparks-Wallace A, Casteel JL, Howell MEA, Ning S. Algorithm-Based Meta-Analysis Reveals the Mechanistic Interaction of the Tumor Suppressor LIMD1 With Non-Small-Cell Lung Carcinoma. Front Oncol 2021; 11:632638. [PMID: 33869018 PMCID: PMC8044451 DOI: 10.3389/fonc.2021.632638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Non-small-cell lung carcinoma (NSCLC) is the major type of lung cancer, which is among the leading causes of cancer-related deaths worldwide. LIMD1 was previously identified as a tumor suppressor in lung cancer, but their detailed interaction in this setting remains unclear. In this study, we have carried out multiple genome-wide bioinformatic analyses for a comprehensive understanding of LIMD1 in NSCLC, using various online algorithm platforms that have been built for mega databases derived from both clinical and cell line samples. Our results indicate that LIMD1 expression level is significantly downregulated at both mRNA and protein levels in both lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), with a considerable contribution from its promoter methylation rather than its gene mutations. The Limd1 gene undergoes mutation only at a low rate in NSCLC (0.712%). We have further identified LIMD1-associated molecular signatures in NSCLC, including its natural antisense long non-coding RNA LIMD1-AS1 and a pool of membrane trafficking regulators. We have also identified a subgroup of tumor-infiltrating lymphocytes, especially neutrophils, whose tumor infiltration levels significantly correlate with LIMD1 level in both LUAD and LUSC. However, a significant correlation of LIMD1 with a subset of immune regulatory molecules, such as IL6R and TAP1, was only found in LUAD. Regarding the clinical outcomes, LIMD1 expression level only significantly correlates with the survival of LUAD (p<0.01) but not with that of LUSC (p>0.1) patients. These findings indicate that LIMD1 plays a survival role in LUAD patients at least by acting as an immune regulatory protein. To further understand the mechanisms underlying the tumor-suppressing function of LIMD1 in NSCLC, we show that LIMD1 downregulation remarkably correlates with the deregulation of multiple pathways that play decisive roles in the oncogenesis of NSCLC, especially those mediated by EGFR, KRAS, PIK3CA, Keap1, and p63, in both LUAD and LUSC, and those mediated by p53 and CDKN2A only in LUAD. This study has disclosed that LIMD1 can serve as a survival prognostic marker for LUAD patients and provides mechanistic insights into the interaction of LIMD1 with NSCLC, which provide valuable information for clinical applications.
Collapse
Affiliation(s)
- Ling Wang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Ayrianna Sparks-Wallace
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Jared L Casteel
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Mary E A Howell
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Shunbin Ning
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States.,Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
43
|
Venkatramanan S, Ibar C, Irvine KD. TRIP6 is required for tension at adherens junctions. J Cell Sci 2021; 134:jcs247866. [PMID: 33558314 PMCID: PMC7970510 DOI: 10.1242/jcs.247866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/29/2021] [Indexed: 01/08/2023] Open
Abstract
Hippo signaling mediates influences of cytoskeletal tension on organ growth. TRIP6 and LIMD1 have each been identified as being required for tension-dependent inhibition of the Hippo pathway LATS kinases and their recruitment to adherens junctions, but the relationship between TRIP6 and LIMD1 was unknown. Using siRNA-mediated gene knockdown, we show that TRIP6 is required for LIMD1 localization to adherens junctions, whereas LIMD1 is not required for TRIP6 localization. TRIP6, but not LIMD1, is also required for the recruitment of vinculin and VASP to adherens junctions. Knockdown of TRIP6 or vinculin, but not of LIMD1, also influences the localization of myosin and F-actin. In TRIP6 knockdown cells, actin stress fibers are lost apically but increased basally, and there is a corresponding increase in the recruitment of vinculin and VASP to basal focal adhesions. Our observations identify a role for TRIP6 in organizing F-actin and maintaining tension at adherens junctions that could account for its influence on LIMD1 and LATS. They also suggest that focal adhesions and adherens junctions compete for key proteins needed to maintain attachments to contractile F-actin.
Collapse
Affiliation(s)
- Srividya Venkatramanan
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Consuelo Ibar
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| |
Collapse
|
44
|
Boycott HE, Nguyen MN, Vrellaku B, Gehmlich K, Robinson P. Nitric Oxide and Mechano-Electrical Transduction in Cardiomyocytes. Front Physiol 2020; 11:606740. [PMID: 33384614 PMCID: PMC7770138 DOI: 10.3389/fphys.2020.606740] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
The ability§ of the heart to adapt to changes in the mechanical environment is critical for normal cardiac physiology. The role of nitric oxide is increasingly recognized as a mediator of mechanical signaling. Produced in the heart by nitric oxide synthases, nitric oxide affects almost all mechano-transduction pathways within the cardiomyocyte, with roles mediating mechano-sensing, mechano-electric feedback (via modulation of ion channel activity), and calcium handling. As more precise experimental techniques for applying mechanical stresses to cells are developed, the role of these forces in cardiomyocyte function can be further understood. Furthermore, specific inhibitors of different nitric oxide synthase isoforms are now available to elucidate the role of these enzymes in mediating mechano-electrical signaling. Understanding of the links between nitric oxide production and mechano-electrical signaling is incomplete, particularly whether mechanically sensitive ion channels are regulated by nitric oxide, and how this affects the cardiac action potential. This is of particular relevance to conditions such as atrial fibrillation and heart failure, in which nitric oxide production is reduced. Dysfunction of the nitric oxide/mechano-electrical signaling pathways are likely to be a feature of cardiac pathology (e.g., atrial fibrillation, cardiomyopathy, and heart failure) and a better understanding of the importance of nitric oxide signaling and its links to mechanical regulation of heart function may advance our understanding of these conditions.
Collapse
Affiliation(s)
- Hannah E. Boycott
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - My-Nhan Nguyen
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Besarte Vrellaku
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| | - Katja Gehmlich
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Li Y, Gao S, Han Y, Song L, Kong Y, Jiao Y, Huang S, Du J, Li Y. Variants of Focal Adhesion Scaffold Genes Cause Thoracic Aortic Aneurysm. Circ Res 2020; 128:8-23. [PMID: 33092471 DOI: 10.1161/circresaha.120.317361] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Thoracic aortic aneurysm (TAA) leads to substantial mortality worldwide. Familial and syndromic TAAs are highly correlated with genetics. However, the incidence of sporadic isolated TAA (iTAA) is much higher, and the genetic contribution is not yet clear. OBJECTIVE Here, we examined the genetic characteristics of sporadic iTAA. METHODS AND RESULTS We performed a genetic screen of 551 sporadic iTAA cases and 1071 controls via whole-exome sequencing. The prevalence of pathogenic mutations in known causal genes was 5.08% in the iTAA cohort. We selected 100 novel candidate genes using a strict strategy, and the suspected functional variants of these genes were significantly enriched in cases compared with controls and carried by 60.43% of patients. We found more severe phenotypes and a lower proportion of hypertension in cases with pathogenic mutations or suspected functional variants. Among the candidate genes, Testin (TES), which encodes a focal adhesion scaffold protein, was identified as a potential TAA causal gene, accounting for 4 patients with 2 missense variants in the LIM1 domain (c.751T>C encoding p.Y251H; c.838T>C encoding p.Y280H) and highly expressed in the aorta. The 2 variants led to a decrease in TES expression. The thoracic aorta was spontaneously dilated in the TesY249H knock-in and Tes-/- mice. Mechanistically, the p.Y249H variant or knockdown of TES led to the repression of vascular smooth muscle cell contraction genes and disturbed the vascular smooth muscle cell contractile phenotype. Interestingly, suspected functional variants of other focal adhesion scaffold genes, including TLN1 (Talin-1) and ZYX (zyxin), were also significantly enriched in patients with iTAA; moreover, their knockdown resulted in decreased contractility of vascular smooth muscle cells. CONCLUSIONS For the first time, this study revealed the genetic landscape across iTAA and showed that the focal adhesion scaffold genes are critical in the pathogenesis of iTAA.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yingchun Han
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Li Song
- BGI Genomics, BGI-Shenzhen, China (Li Song)
| | - Yu Kong
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yao Jiao
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| |
Collapse
|
46
|
Sun X, Phua DYZ, Axiotakis L, Smith MA, Blankman E, Gong R, Cail RC, Espinosa de Los Reyes S, Beckerle MC, Waterman CM, Alushin GM. Mechanosensing through Direct Binding of Tensed F-Actin by LIM Domains. Dev Cell 2020; 55:468-482.e7. [PMID: 33058779 DOI: 10.1016/j.devcel.2020.09.022] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/04/2020] [Accepted: 09/21/2020] [Indexed: 01/21/2023]
Abstract
Mechanical signals transmitted through the cytoplasmic actin cytoskeleton must be relayed to the nucleus to control gene expression. LIM domains are protein-protein interaction modules found in cytoskeletal proteins and transcriptional regulators. Here, we identify three LIM protein families (zyxin, paxillin, and FHL) whose members preferentially localize to the actin cytoskeleton in mechanically stimulated cells through their tandem LIM domains. A minimal actin-myosin reconstitution system reveals that representatives of all three families directly bind F-actin only in the presence of mechanical force. Point mutations at a site conserved in each LIM domain of these proteins disrupt tensed F-actin binding in vitro and cytoskeletal localization in cells, demonstrating a common, avidity-based mechanism. Finally, we find that binding to tensed F-actin in the cytoplasm excludes the cancer-associated transcriptional co-activator FHL2 from the nucleus in stiff microenvironments. This establishes direct force-activated F-actin binding as a mechanosensing mechanism by which cytoskeletal tension can govern nuclear localization.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Donovan Y Z Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Lucas Axiotakis
- Laboratory of Macromolecular Interactions, Cell Biology and Physiology Center, Division of Intramural Research, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Mark A Smith
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Elizabeth Blankman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Rui Gong
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Robert C Cail
- Laboratory of Macromolecular Interactions, Cell Biology and Physiology Center, Division of Intramural Research, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | | | - Mary C Beckerle
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Clare M Waterman
- Cell and Developmental Biology Center, Division of Intramural Research, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Macromolecular Interactions, Cell Biology and Physiology Center, Division of Intramural Research, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Winkelman JD, Anderson CA, Suarez C, Kovar DR, Gardel ML. Evolutionarily diverse LIM domain-containing proteins bind stressed actin filaments through a conserved mechanism. Proc Natl Acad Sci U S A 2020; 117:25532-25542. [PMID: 32989126 PMCID: PMC7568268 DOI: 10.1073/pnas.2004656117] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The actin cytoskeleton assembles into diverse load-bearing networks, including stress fibers (SFs), muscle sarcomeres, and the cytokinetic ring to both generate and sense mechanical forces. The LIM (Lin11, Isl- 1, and Mec-3) domain family is functionally diverse, but most members can associate with the actin cytoskeleton with apparent force sensitivity. Zyxin rapidly localizes via its LIM domains to failing SFs in cells, known as strain sites, to initiate SF repair and maintain mechanical homeostasis. The mechanism by which these LIM domains associate with stress fiber strain sites (SFSS) is not known. Additionally, it is unknown how widespread strain sensing is within the LIM protein family. We identify that the LIM domain-containing region of 18 proteins from the Zyxin, Paxillin, Tes, and Enigma proteins accumulate to SFSS. Moreover, the LIM domain region from the fission yeast protein paxillin like 1 (Pxl1) also localizes to SFSS in mammalian cells, suggesting that the strain sensing mechanism is ancient and highly conserved. We then used sequence and domain analysis to demonstrate that tandem LIM domains contribute additively, for SFSS localization. Employing in vitro reconstitution, we show that the LIM domain-containing region from mammalian zyxin and fission yeast Pxl1 binds to mechanically stressed F-actin networks but does not associate with relaxed actin filaments. We propose that tandem LIM domains recognize an F-actin conformation that is rare in the relaxed state but is enriched in the presence of mechanical stress.
Collapse
Affiliation(s)
| | - Caitlin A Anderson
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Cristian Suarez
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637;
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637;
- James Franck Institute, University of Chicago, Chicago, IL 60637
- Physics Department, University of Chicago, Chicago, IL 60637
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637
| |
Collapse
|
48
|
Sabino F, Madzharova E, Auf dem Keller U. Cell density-dependent proteolysis by HtrA1 induces translocation of zyxin to the nucleus and increased cell survival. Cell Death Dis 2020; 11:674. [PMID: 32826880 PMCID: PMC7442833 DOI: 10.1038/s41419-020-02883-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 01/01/2023]
Abstract
Proteases modulate critical processes in cutaneous tissue repair to orchestrate inflammation, cell proliferation and tissue remodeling. However, the functional consequences and implications in healing impairments of most cleavage events are not understood. Using iTRAQ-based Terminal Amine Isotopic Labeling of Substrates (TAILS) we had characterized proteolytic signatures in a porcine wound healing model and identified two neo-N termini derived from proteolytic cleavage of the focal adhesion protein and mechanotransducer zyxin. Here, we assign these proteolytic events to the activity of either caspase-1 or serine protease HtrA1 and analyze the biological relevance of the resultant zyxin truncations. By cellular expression of full-length and truncated zyxin proteins, we demonstrate nuclear translocation of a C-terminal zyxin fragment that could also be generated in vitro by HtrA1 cleavage and provide evidence for its anti-apoptotic activities, potentially by regulating the expression of modulators of cell proliferation, protein synthesis and genome stability. Targeted degradomics correlated endogenous generation of the same zyxin fragment with increased cell density in human primary dermal fibroblasts. Hence, this newly identified HtrA1-zyxin protease signaling axis might present a novel mechanism to transiently enhance cell survival in environments of increased cell density like in wound granulation tissue.
Collapse
Affiliation(s)
- Fabio Sabino
- Technical University of Denmark, Department of Biotechnology and Biomedicine, Søltofts Plads, 2800, Kongens Lyngby, Denmark
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093, Zurich, Switzerland
| | - Elizabeta Madzharova
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093, Zurich, Switzerland
| | - Ulrich Auf dem Keller
- Technical University of Denmark, Department of Biotechnology and Biomedicine, Søltofts Plads, 2800, Kongens Lyngby, Denmark.
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.
| |
Collapse
|
49
|
Huang X, Qu R, Ouyang J, Zhong S, Dai J. An Overview of the Cytoskeleton-Associated Role of PDLIM5. Front Physiol 2020; 11:975. [PMID: 32848888 PMCID: PMC7426503 DOI: 10.3389/fphys.2020.00975] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Regenerative medicine represented by stem cell technology has become one of the pillar medical technologies for human disease treatment. Cytoskeleton plays important roles in maintaining cell morphology, bearing external forces, and maintaining the effectiveness of cell internal structure, among which cytoskeleton related proteins are involved in and play an indispensable role in the changes of cytoskeleton. PDLIM5 is a cytoskeleton-related protein that, like other cytoskeletal proteins, acts as a binding protein. PDZ and LIM domain 5 (PDLIM5), also known as ENH (Enigma homolog), is a cytoplasmic protein with a molecular mass of about 63 KDa that consists of a PDZ domain at the N-terminus and three LIM domains at the C-terminus. PDLIM5 binds to the cytoskeleton and membrane proteins through its PDZ domain and interacts with various signaling molecules, including protein kinases and transcription factors, through its LIM domain. As a cytoskeleton-related protein, PDLIM5 plays an important role in regulating cell proliferation, differentiation and cell fate decision in multiple tissues and cell types. In this review, we briefly summarize the state of knowledge on the PDLIM5 gene, structural properties, and molecular functional mechanisms of the PDLIM5 protein, and its role in cells, tissues, and organ systems, and describe the possible underlying molecular signaling pathways. In the last part of this review, we will focus on discussing the limitations of existing research and the future prospects of PDLIM5 research in turn.
Collapse
Affiliation(s)
- Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Hsu HW, Liao CP, Chiang YC, Syu RT, Pan CL. Caenorhabditis elegans Flamingo FMI-1 controls dendrite self-avoidance through F-actin assembly. Development 2020; 147:dev179168. [PMID: 32631831 DOI: 10.1242/dev.179168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
Self-avoidance is a conserved mechanism that prevents crossover between sister dendrites from the same neuron, ensuring proper functioning of the neuronal circuits. Several adhesion molecules are known to be important for dendrite self-avoidance, but the underlying molecular mechanisms are incompletely defined. Here, we show that FMI-1/Flamingo, an atypical cadherin, is required autonomously for self-avoidance in the multidendritic PVD neuron of Caenorhabditis elegans The fmi-1 mutant shows increased crossover between sister PVD dendrites. Our genetic analysis suggests that FMI-1 promotes transient F-actin assembly at the tips of contacting sister dendrites to facilitate their efficient retraction during self-avoidance events, probably by interacting with WSP-1/N-WASP. Mutations of vang-1, which encodes the planar cell polarity protein Vangl2 previously shown to inhibit F-actin assembly, suppress self-avoidance defects of the fmi-1 mutant. FMI-1 downregulates VANG-1 levels probably through forming protein complexes. Our study identifies molecular links between Flamingo and the F-actin cytoskeleton that facilitate efficient dendrite self-avoidance.
Collapse
Affiliation(s)
- Hao-Wei Hsu
- Institute of Molecular Medicine and Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chien-Po Liao
- Institute of Molecular Medicine and Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Yueh-Chen Chiang
- Institute of Molecular Medicine and Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Ru-Ting Syu
- Institute of Molecular Medicine and Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chun-Liang Pan
- Institute of Molecular Medicine and Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| |
Collapse
|