1
|
Li Y, Guo Q, Wang H, Wang R, Kang W, Zhang CL, Zhang D, Xiao K, Sun Z. Associations between vascular endothelial growth factor polymorphisms and response to 5-FU-based pharmaceutical therapy in esophageal squamous cell carcinoma: A meta-analysis. TUMORI JOURNAL 2025; 111:112-120. [PMID: 40012106 DOI: 10.1177/03008916251322057] [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] [Indexed: 02/28/2025]
Abstract
BACKGROUND AND AIMS Vascular endothelial growth factor (VEGF) gene polymorphisms are associated with the response to pharmaceutical therapy in many cancers. This study aimed to investigate the effects of VEGF gene polymorphisms in esophageal squamous cell carcinoma patients receiving pharmaceutical therapy. METHODS This literature-based meta-analysis was performed with keywords related to VEGF gene polymorphisms and clinical response in esophageal squamous carcinoma patients receiving pharmaceutical therapy (including 5-FU, cisplatin, oxaliplatin, and calcium folinate). After a series of bias grading analyses and DerSimonian-Laird method analysis, odds ratios and 95% confidence intervals were calculated to examine the potential relationships. Sensitivity and subgroup analyses were subsequently performed to determine the major causes of heterogeneity. RESULTS Heterogeneity was dramatically reduced after the removal of one study from the analysis (I2 = 37%, P = 0.19). The remaining studies involved 5-FU-based treatment. The presence of VEGF G-1154A and VEGF-634C/G was found to be correlated with patient response to 5-FU/CDDP-based treatment, whereas VEGF-2549I/D was correlated with response to 5-FU/oxaliplatin-based treatment, and VEGF-936C/T was associated with both 5-FU/CDDP- and 5-FU/oxaliplatin-based treatment response. CONCLUSION VEGF gene polymorphisms affect the response of esophageal squamous carcinoma patients receiving pharmaceutical therapy, especially 5-FU-based treatments.
Collapse
Affiliation(s)
- Yonghui Li
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Qiang Guo
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Haibo Wang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Ruiyao Wang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - WenLi Kang
- Department of Obstetrical, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Cheng Long Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Duo Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Kuo Xiao
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - ZhenQing Sun
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| |
Collapse
|
2
|
Runyan LA, Kudryashova E, Agrawal R, Mohamed M, Kudryashov DS. Human plastins are novel cytoskeletal pH sensors with a reduced F-actin bundling capacity at basic pH. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645573. [PMID: 40196613 PMCID: PMC11974883 DOI: 10.1101/2025.03.26.645573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intracellular pH (pHi) is a fundamental component of cell homeostasis. Controlled elevations in pHi precede and accompany cell polarization, cytokinesis, and directional migration. pH dysregulation contributes to cancer, neurodegenerative diseases, diabetes, and other metabolic disorders. While cytoskeletal rearrangements are crucial for these processes, only a few cytoskeletal proteins, namely Cdc42, cofilin, talin, cortactin, α-actinin, and AIP1 have been documented as pH sensors. Here, we report that actin-bundling proteins plastin 2 (PLS2, aka LCP1) and plastin 3 (PLS3) respond to physiological scale pH fluctuations by a reduced F-actin bundling at alkaline pH. The inhibition of PLS2 actin-bundling activity at elevated pH stems from the reduced affinity of the N-terminal actin-binding domain (ABD1) to actin. In fibroblast cells, elevated cytosolic pH caused the dissociation of ectopically expressed PLS2 from actin structures, whereas acidic conditions promoted its tighter association with focal adhesions and stress fibers. We identified His207 as one of the pH-sensing residues whose mutation to Lys and Tyr reduces pH sensitivity by enhancing and inhibiting the bundling ability, respectively. Our results suggest that weaker actin bundling by plastin isoforms at alkaline pH favors higher dynamics of the actin cytoskeleton. Therefore, like other cytoskeleton pH sensors, plastins promote disassembly and faster dynamics of cytoskeletal components during cytokinesis and cell migration. Since both plastins are implemented in cancer, their pH sensitivity may contribute to the accelerated proliferation and enhanced invasive and metastatic potentials of cancer cells at alkaline pHi.
Collapse
Affiliation(s)
- Lucas A. Runyan
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA, 43210
| | - Elena Kudryashova
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA, 43210
| | - Richa Agrawal
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA, 43210
| | - Mubarik Mohamed
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA, 43210
| | - Dmitri S. Kudryashov
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, USA, 43210
| |
Collapse
|
3
|
Zhou S, Liu B, Liu J, Yi B, Wang X. Spatiotemporal dissection of collective cell migration and tissue morphogenesis during development by optogenetics. Semin Cell Dev Biol 2025; 166:36-51. [PMID: 39729778 DOI: 10.1016/j.semcdb.2024.12.004] [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: 09/16/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/29/2024]
Abstract
Collective cell migration and tissue morphogenesis play a variety of important roles in the development of many species. Tissue morphogenesis often generates mechanical forces that alter cell shapes and arrangements, resembling collective cell migration-like behaviors. Genetic methods have been widely used to study collective cell migration and its like behavior, advancing our understanding of these processes during development. However, a growing body of research shows that collective cell migration during development is not a simple behavior but is often combined with other cellular and tissue processes. In addition, different surrounding environments can also influence migrating cells, further complicating collective cell migration during development. Due to the complexity of developmental processes and tissues, traditional genetic approaches often encounter challenges and limitations. Thus, some methods with spatiotemporal control become urgent in dissecting collective cell migration and tissue morphogenesis during development. Optogenetics is a method that combines optics and genetics, providing a perfect strategy for spatiotemporally controlling corresponding protein activity in subcellular, cellular or tissue levels. In this review, we introduce the basic mechanisms underlying different optogenetic tools. Then, we demonstrate how optogenetic methods have been applied in vivo to dissect collective cell migration and tissue morphogenesis during development. Additionally, we describe some promising optogenetic approaches for advancing this field. Together, this review will guide and facilitate future studies of collective cell migration in vivo and tissue morphogenesis by optogenetics.
Collapse
Affiliation(s)
- Sijia Zhou
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, China; Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, Toulouse, France.
| | - Bing Liu
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, Toulouse, France.
| | - Jiaying Liu
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, Toulouse, France
| | - Bin Yi
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | - Xiaobo Wang
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Integrative (CBI), University of Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
4
|
Li Z, Shao R, Xin H, Zhu Y, Jiang S, Wu J, Yan H, Jia T, Ge M, Shi X. Paxillin and Kindlin: Research Progress and Biological Functions. Biomolecules 2025; 15:173. [PMID: 40001476 PMCID: PMC11853175 DOI: 10.3390/biom15020173] [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: 11/17/2024] [Revised: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Paxillin and kindlin are essential regulatory proteins involved in cell adhesion, migration, and signal transduction. Paxillin influences cytoskeletal dynamics by interacting with multiple signaling proteins, while kindlin regulates integrin activation, affecting adhesion and motility. This review examines the structures and functions of these proteins, focusing on their roles in cancer progression, immune response, and therapeutic potential. The cooperation between paxillin and kindlin in integrin activation and focal adhesion dynamics offers valuable insights into tumor metastasis, immune function, and tissue repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xiaofeng Shi
- The Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing 210003, China; (Z.L.)
| |
Collapse
|
5
|
Courte J, Chung C, Jain N, Salazar C, Phuchane N, Grosser S, Lam C, Morsut L. Programming the elongation of mammalian cell aggregates with synthetic gene circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627621. [PMID: 39713354 PMCID: PMC11661162 DOI: 10.1101/2024.12.11.627621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
A key goal of synthetic morphogenesis is the identification and implementation of methods to control morphogenesis. One line of research is the use of synthetic genetic circuits guiding the self-organization of cell ensembles. This approach has led to several recent successes, including control of cellular rearrangements in 3D via control of cell-cell adhesion by user-designed artificial genetic circuits. However, the methods employed to reach such achievements can still be optimized along three lines: identification of circuits happens by hand, 3D structures are spherical, and effectors are limited to cell-cell adhesion. Here we show the identification, in a computational framework, of genetic circuits for volumetric axial elongation via control of proliferation, tissue fluidity, and cell-cell signaling. We then seek to implement this design in mammalian cell aggregates in vitro. We start by identifying effectors to control tissue growth and fluidity in vitro. We then combine these new modules to construct complete circuits that control cell behaviors of interest in space and time, resulting in measurable tissue deformation along an axis that depends on the engineered signaling modules. Finally, we contextualize in vitro and in silico implementations within a unified morphospace to suggest further elaboration of this initial family of circuits towards more robust programmed axial elongation. These results and integrated in vitro/in silico pipeline demonstrate a promising method for designing, screening, and implementing synthetic genetic circuits of morphogenesis, opening the way to the programming of various user-defined tissue shapes.
Collapse
Affiliation(s)
- Josquin Courte
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christian Chung
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Naisargee Jain
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Catcher Salazar
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Neo Phuchane
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Steffen Grosser
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Calvin Lam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Leonardo Morsut
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Li XM, Xu K, Wang JY, Guo JY, Wang XH, Zeng L, Wan B, Wang J, Chu BB, Yang GY, Pan JJ, Hao WB. The actin cytoskeleton is important for pseudorabies virus infection. Virology 2024; 600:110233. [PMID: 39255726 DOI: 10.1016/j.virol.2024.110233] [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: 04/19/2024] [Revised: 08/06/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
Viruses are dependent on the host factors for their replication and survival. Therefore, identification of host factors that druggable for antiviral development is crucial. The actin cytoskeleton plays an important role in the virus infection. The dynamics change of actin and its function are regulated by multiple actin-associated proteins (AAPs). However, the role and mechanism of various AAPs in the life cycle of virus are still enigmatic. In this study, we analyzed the roles of actin and AAPs in the replication of pseudorabies virus (PRV). Using a library of compounds targeting AAPs, our data found that multiple AAPs, such as Rho-GTPases, Rock, Myosin and Formin were involved in PRV infection. Besides, our result demonstrated that the actin-binding protein Drebrin was also participated in PRV infection. Further studies are necessary to elucidate the molecular mechanism of AAPs in the virus life cycle, in the hope of mining host factors for antiviral developments.
Collapse
Affiliation(s)
- Xin-Man Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Kun Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Jin-Yuan Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Jie-Yuan Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Xiao-Han Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China
| | - Bo Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Henan University of Animal Husbandry and Economy, Zhengzhou, 450047, China
| | - Jia-Jia Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China.
| | - Wen-Bo Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
7
|
Li T, Song Y, Wei L, Song X, Duan R. Disulfidptosis: a novel cell death modality induced by actin cytoskeleton collapse and a promising target for cancer therapeutics. Cell Commun Signal 2024; 22:491. [PMID: 39394612 PMCID: PMC11470700 DOI: 10.1186/s12964-024-01871-9] [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: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Disulfidptosis is a novel discovered form of programmed cell death (PCD) that diverges from apoptosis, necroptosis, ferroptosis, and cuproptosis, stemming from disulfide stress-induced cytoskeletal collapse. In cancer cells exhibiting heightened expression of the solute carrier family 7 member 11 (SLC7A11), excessive cystine importation and reduction will deplete nicotinamide adenine dinucleotide phosphate (NADPH) under glucose deprivation, followed by an increase in intracellular disulfide stress and aberrant disulfide bond formation within actin networks, ultimately culminating in cytoskeletal collapse and disulfidptosis. Disulfidptosis involves crucial physiological processes in eukaryotic cells, such as cystine and glucose uptake, NADPH metabolism, and actin dynamics. The Rac1-WRC pathway-mediated actin polymerization is also implicated in this cell death due to its contribution to disulfide bond formation. However, the precise mechanisms underlying disulfidptosis and its role in tumors are not well understood. This is probably due to the multifaceted functionalities of SLC7A11 within cells and the complexities of the downstream pathways driving disulfidptosis. This review describes the critical roles of SLC7A11 in cells and summarizes recent research advancements in the potential pathways of disulfidptosis. Moreover, the less-studied aspects of this newly discovered cell death process are highlighted to stimulate further investigations in this field.
Collapse
Affiliation(s)
- Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Xiangyi Song
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China.
| |
Collapse
|
8
|
Popov IK, Tao J, Chang C. The RhoGEF protein Plekhg5 self-associates via its PH domain to regulate apical cell constriction. Mol Biol Cell 2024; 35:ar134. [PMID: 39196644 PMCID: PMC11481697 DOI: 10.1091/mbc.e24-04-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 08/30/2024] Open
Abstract
RhoGEFs are critical activators of Rho family small GTPases and regulate diverse biological processes, such as cell division and tissue morphogenesis. We reported previously that the RhoGEF gene plekhg5 controls apical constriction of bottle cells at the blastopore lip during Xenopus gastrulation, but the detailed mechanism of plekhg5 action is not understood in depth. In this study, we show that localization of Plekhg5 in the apical cortex depends on its N-terminal sequences and intact guanine nucleotide exchange activity, whereas the C-terminal sequences prevent ectopic localization of the protein to the basolateral compartment. We also reveal that Plekhg5 self-associates via its PH domain, and this interaction leads to functional rescue of two mutants that lack the N-terminal region and the guanine nucleotide exchange factor activity, respectively, in trans. A point mutation in the PH domain corresponding to a variant associated with human disease leads to loss of self-association and failure of the mutant to induce apical constriction. Taken together, our results suggest that PH-mediated self-association and N-terminal domain-mediated subcellular localization are both crucial for the function of Plekhg5 in inducing apical constriction.
Collapse
Affiliation(s)
- Ivan K. Popov
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jiahui Tao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
9
|
Ksovreli M, Kachlishvili T, Skhvitaridze M, Nadaraia L, Goliadze R, Kamashidze L, Zurabiani K, Batsatsashvili T, Kvachantiradze N, Gverdtsiteli M, Kantaria T, Piot O, Courageot MP, Terryn C, Tchelidze P, Katsarava R, Kulikova N. Wound Closure Promotion by Leucine-Based Pseudo-Proteins: An In Vitro Study. Int J Mol Sci 2024; 25:9641. [PMID: 39273588 PMCID: PMC11395615 DOI: 10.3390/ijms25179641] [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: 08/20/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Our research explores leucine-based pseudo-proteins (LPPs) for advanced wound dressings, focusing on their effects on wound healing in an in vitro model. We assessed three types of LPP films for their ability to enhance wound closure rates and modulate cytokine production. They all significantly improved wound closure compared to traditional methods, with the 8L6 and copolymer films showing the most pronounced effects. Notably, the latter exhibited an optimal cytokine profile: an initial burst of pro-inflammatory TNF-α, followed by a controlled release of IL-6 during the proliferative phase and a significant increase in anti-inflammatory IL-10 during remodeling. This balanced cytokine response suggests that the copolymer film not only accelerates wound closure but also supports a well-regulated healing process, potentially reducing fibrosis and abnormal scarring, underscoring the potential of copolymer LPPs as advanced wound dressing materials. Future research will aim to elucidate the specific signaling pathways activated by the copolymer LPP to better understand its mechanism of action. Overall, LPP films offer a promising approach to improving wound care and could lead to more effective treatments for complex wounds.
Collapse
Affiliation(s)
- Mariam Ksovreli
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Tinatin Kachlishvili
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Mariam Skhvitaridze
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Lili Nadaraia
- Carl Zeiss Scientific and Education Center, New Vision University, 0159 Tbilisi, Georgia
| | - Rusudan Goliadze
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Luka Kamashidze
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Knarita Zurabiani
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Tatuli Batsatsashvili
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Nino Kvachantiradze
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Marekhi Gverdtsiteli
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Temur Kantaria
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Olivier Piot
- BioSpectroscopie Translationnelle (BioSpecT) Unit, University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Marie-Pierre Courageot
- BioSpectroscopie Translationnelle (BioSpecT) Unit, University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Christine Terryn
- La plateforme en Imagerie Cellulaire et Tissulaire (PICT), University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Pavel Tchelidze
- Faculty of Healthcare, East European University, 0159 Tbilisi, Georgia
| | - Ramaz Katsarava
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| | - Nina Kulikova
- Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 0159 Tbilisi, Georgia
| |
Collapse
|
10
|
Zhang Q, Lu B. The mRNA and microRNA Landscape of the Blastema Niche in Regenerating Newt Limbs. Int J Mol Sci 2024; 25:9225. [PMID: 39273174 PMCID: PMC11395517 DOI: 10.3390/ijms25179225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Newts are excellent vertebrate models for investigating tissue regeneration due to their remarkable regenerative capabilities. To investigate the mRNA and microRNAs (miRNAs) profiles within the blastema niche of regenerating newt limbs, we amputated the limbs of Chinese fire belly newts (Cynops orientalis) and conducted comprehensive analyses of the transcriptome and microRNA profiles at five distinct time points post-amputation (0 hours, 1 day, 5 days 10 days and 20 days). We identified 24 significantly differentially expressed (DE) genes and 20 significantly DE miRNAs. Utilizing weighted gene co-expression network analysis (WGCNA) and gene ontology (GO) enrichment analysis, we identified four genes likely to playing crucial roles in the early stages of limb regeneration: Cemip, Rhou, Gpd2 and Pcna. Moreover, mRNA-miRNA integration analysis uncovered seven human miRNAs (miR-19b-1, miR-19b-2, miR-21-5p, miR-127-5p, miR-150-5p, miR-194-5p, and miR-210-5p) may regulate the expression of these four key genes. The temporal expression patterns of these key genes and miRNAs further validated the robustness of the identified mRNA-miRNA landscape. Our study successfully identified candidate key genes and elucidated a portion of the genetic regulatory mechanisms involved in newt limb regeneration. These findings offer valuable insights for further exploration of the intricate processes of tissue regeneration.
Collapse
Affiliation(s)
- Qi Zhang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Lu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| |
Collapse
|
11
|
Nheu D, Petratos S. How does Nogo-A signalling influence mitochondrial function during multiple sclerosis pathogenesis? Neurosci Biobehav Rev 2024; 163:105767. [PMID: 38885889 DOI: 10.1016/j.neubiorev.2024.105767] [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: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.
Collapse
Affiliation(s)
- Danica Nheu
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia.
| |
Collapse
|
12
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [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] [Indexed: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
13
|
Iyer KS, Maruri DP, Schmidtke DW, Petroll WM, Varner VD. Treatment with both TGF-β1 and PDGF-BB disrupts the stiffness-dependent myofibroblast differentiation of corneal keratocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582803. [PMID: 38496568 PMCID: PMC10942298 DOI: 10.1101/2024.02.29.582803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
During corneal wound healing, stromal keratocytes transform into a repair phenotype that is driven by the release of cytokines, like transforming growth factor-beta 1 (TGF-β1) and platelet-derived growth factor-BB (PDGF-BB). Previous work has shown that TGF-β1 promotes the myofibroblast differentiation of corneal keratocytes in a manner that depends on PDGF signaling. In addition, changes in mechanical properties are known to regulate the TGF-β1-mediated differentiation of cultured keratocytes. While PDGF signaling acts synergistically with TGF-β1 during myofibroblast differentiation, how treatment with multiple growth factors affects stiffness-dependent differences in keratocyte behavior is unknown. Here, we treated primary corneal keratocytes with PDGF-BB and TGF-β1 and cultured them on polyacrylamide (PA) substrata of different stiffnesses. In the presence of TGF-β1 alone, the cells underwent stiffness-dependent myofibroblast differentiation. On stiff substrata, the cells developed robust stress fibers, exhibited high levels of ⍺-SMA staining, formed large focal adhesions (FAs), and exerted elevated contractile forces, whereas cells in a compliant microenvironment showed low levels of ⍺-SMA immunofluorescence, formed smaller focal adhesions, and exerted decreased contractile forces. When the cultured keratocytes were treated simultaneously with PDGF-BB however, increased levels of ⍺-SMA staining and stress fiber formation were observed on compliant substrata, even though the cells did not exhibit elevated contractility or focal adhesion size. Pharmacological inhibition of PDGF signaling disrupted the myofibroblast differentiation of cells cultured on substrata of all stiffnesses. These results indicate that treatment with PDGF-BB can decouple molecular markers of myofibroblast differentiation from the elevated contractile phenotype otherwise associated with these cells, suggesting that crosstalk in the mechanotransductive signaling pathways downstream of TGF-β1 and PDGF-BB can regulate the stiffness-dependent differentiation of cultured keratocytes. Statement of Significance In vitro experiments have shown that changes in ECM stiffness can regulate the differentiation of myofibroblasts. Typically, these assays involve the use of individual growth factors, but it is unclear how stiffness-dependent differences in cell behavior are affected by multiple cytokines. Here, we used primary corneal keratocytes to show that treatment with both TGF-β1 and PDGF-BB disrupts the dependency of myofibroblast differentiation on substratum stiffness. In the presence of both growth factors, keratocytes on soft substrates exhibited elevated ⍺-SMA immunofluorescence without a corresponding increase in contractility or focal adhesion formation. This result suggests that molecular markers of myofibroblast differentiation can be dissociated from the elevated contractile behavior associated with the myofibroblast phenotype, suggesting potential crosstalk in mechanotransductive signaling pathways downstream of TGF-β1 and PDGF-BB.
Collapse
|
14
|
He J, Tang MY, Liu LX, Kong CX, Chen W, Wang L, Zhi SB, Sun HW, Huang YC, Chen GY, Xin HB, Deng KY. Myeloid Deletion of Cdc42 Protects Liver From Hepatic Ischemia-Reperfusion Injury via Inhibiting Macrophage-Mediated Inflammation in Mice. Cell Mol Gastroenterol Hepatol 2024; 17:965-981. [PMID: 38342302 PMCID: PMC11047801 DOI: 10.1016/j.jcmgh.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND & AIMS Hepatic ischemia-reperfusion injury (HIRI) often occurs in liver surgery, such as partial hepatectomy and liver transplantation, in which myeloid macrophage-mediated inflammation plays a critical role. Cell division cycle 42 (Cdc42) regulates cell migration, cytoskeleton rearrangement, and cell polarity. In this study, we explore the role of myeloid Cdc42 in HIRI. METHODS Mouse HIRI models were established with 1-hour ischemia followed by 12-hour reperfusion in myeloid Cdc42 knockout (Cdc42mye) and Cdc42flox mice. Myeloid-derived macrophages were traced with RosamTmG fluorescent reporter under LyzCre-mediated excision. The experiments for serum or hepatic enzymic activities, histologic and immunologic analysis, gene expressions, flow cytometry analysis, and cytokine antibody array were performed. RESULTS Myeloid deletion of Cdc42 significantly alleviated hepatic damages with the reduction of hepatic necrosis and inflammation, and reserved hepatic functions following HIRI in mice. Myeloid Cdc42 deficiency suppressed the infiltration of myeloid macrophages, reduced the secretion of proinflammatory cytokines, restrained M1 polarization, and promoted M2 polarization of myeloid macrophages in livers. In addition, inactivation of Cdc42 promoted M2 polarization via suppressing the phosphorylation of STAT1 and promoting phosphorylation of STAT3 and STAT6 in myeloid macrophages. Furthermore, pretreatment with Cdc42 inhibitor, ML141, also protected mice from hepatic ischemia-reperfusion injury. CONCLUSIONS Inhibition or deletion of myeloid Cdc42 protects liver from HIRI via restraining the infiltration of myeloid macrophages, suppressing proinflammatory response, and promoting M2 polarization in macrophages.
Collapse
Affiliation(s)
- Jing He
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Meng-Yu Tang
- College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Li-Xin Liu
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China; College of Pharmacy, Nanchang University, Nanchang, Jiangxi, PR China
| | - Chen-Xian Kong
- College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Wen Chen
- College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China
| | - Lu Wang
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Shao-Bin Zhi
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Hong-Wei Sun
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Yu-Chun Huang
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China
| | - Guo-Yu Chen
- The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi, PR China
| | - Hong-Bo Xin
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China; College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China; College of Pharmacy, Nanchang University, Nanchang, Jiangxi, PR China.
| | - Ke-Yu Deng
- The National Engineering Research Center for Bioengineering Drugs and Technology, Institution of Translation Medicine, Nanchang University, Nanchang, Jiangxi, PR China; College of Life Science, Nanchang University, Nanchang, Jiangxi, PR China; College of Pharmacy, Nanchang University, Nanchang, Jiangxi, PR China.
| |
Collapse
|
15
|
Fukatsu S, Miyamoto Y, Oka Y, Ishibashi M, Shirai R, Ishida Y, Endo S, Katoh H, Yamauchi J. Investigating the Protective Effects of a Citrus Flavonoid on the Retardation Morphogenesis of the Oligodendroglia-like Cell Line by Rnd2 Knockdown. Neurol Int 2023; 16:33-61. [PMID: 38251051 PMCID: PMC10801557 DOI: 10.3390/neurolint16010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Recent discoveries suggest links between abnormalities in cell morphogenesis in the brain and the functional deficiency of molecules controlling signal transduction in glial cells such as oligodendroglia. Rnd2 is one such molecule and one of the Rho family monomeric GTP-binding proteins. Despite the currently known functions of Rnd2, its precise roles as it relates to cell morphogenesis and disease state remain to be elucidated. First, we showed that signaling through the loss of function of the rnd2 gene affected the regulation of oligodendroglial cell-like morphological differentiation using the FBD-102b cell line, which is often utilized as a differentiation model. The knockdown of Rnd2 using the clustered regularly interspaced palindromic repeats (CRISPR)/CasRx system or RNA interference was shown to slow morphological differentiation. Second, the knockdown of Prag1 or Fyn kinase, a signaling molecule acting downstream of Rnd2, slowed differentiation. Rnd2 or Prag1 knockdown also decreased Fyn phosphorylation, which is critical for its activation and for oligodendroglial cell differentiation and myelination. Of note, hesperetin, a citrus flavonoid with protective effects on oligodendroglial cells and neurons, can recover differentiation states induced by the knockdown of Rnd2/Prag1/Fyn. Here, we showed that signaling through Rnd2/Prag1/Fyn is involved in the regulation of oligodendroglial cell-like morphological differentiation. The effects of knocking down the signaling cascade molecule can be recovered by hesperetin, highlighting an important molecular structure involved in morphological differentiation.
Collapse
Affiliation(s)
- Shoya Fukatsu
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
| | - Yu Oka
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Maki Ishibashi
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Remina Shirai
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
| | - Yuki Ishida
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Shin Endo
- Personal Health Care Division, Hayashibara Co., Ltd., Okayama 702-8006, Japan
| | - Hironori Katoh
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka 599-8531, Japan;
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (S.F.); (Y.M.); (R.S.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
16
|
Nanda S, Calderon A, Sachan A, Duong TT, Koch J, Xin X, Solouk-Stahlberg D, Wu YW, Nalbant P, Dehmelt L. Rho GTPase activity crosstalk mediated by Arhgef11 and Arhgef12 coordinates cell protrusion-retraction cycles. Nat Commun 2023; 14:8356. [PMID: 38102112 PMCID: PMC10724141 DOI: 10.1038/s41467-023-43875-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Rho GTPases play a key role in the spatio-temporal coordination of cytoskeletal dynamics during cell migration. Here, we directly investigate crosstalk between the major Rho GTPases Rho, Rac and Cdc42 by combining rapid activity perturbation with activity measurements in mammalian cells. These studies reveal that Rac stimulates Rho activity. Direct measurement of spatio-temporal activity patterns show that Rac activity is tightly and precisely coupled to local cell protrusions, followed by Rho activation during retraction. Furthermore, we find that the Rho-activating Lbc-type GEFs Arhgef11 and Arhgef12 are enriched at transient cell protrusions and retractions and recruited to the plasma membrane by active Rac. In addition, their depletion reduces activity crosstalk, cell protrusion-retraction dynamics and migration distance and increases migration directionality. Thus, our study shows that Arhgef11 and Arhgef12 facilitate exploratory cell migration by coordinating cell protrusion and retraction by coupling the activity of the associated regulators Rac and Rho.
Collapse
Affiliation(s)
- Suchet Nanda
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Abram Calderon
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Arya Sachan
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
| | - Thanh-Thuy Duong
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Johannes Koch
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Xiaoyi Xin
- SciLifeLab and Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Djamschid Solouk-Stahlberg
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Yao-Wen Wu
- SciLifeLab and Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany.
| | - Leif Dehmelt
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany.
| |
Collapse
|
17
|
Lv C, Huang Y, Yan R, Gao Y. Vascular endothelial growth factor induces the migration of human airway smooth muscle cells by activating the RhoA/ROCK pathway. BMC Pulm Med 2023; 23:505. [PMID: 38093231 PMCID: PMC10720058 DOI: 10.1186/s12890-023-02803-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Airway remodeling due to increased airway smooth muscle cell (ASMC) mass, likely due to enhanced proliferation, hypertrophy, and migration, has been proven to be highly correlated with decreased lung function in asthma patients. Vascular endothelial growth factor (VEGF) mediates vascular and extravascular remodeling and inflammation and has been proven to be involved in the progression of asthma. Previous studies have focused on the effects of VEGF on ASMC proliferation, but few researchers have focused on the effects of VEGF on human ASMC migration. The purpose of this study was to explore the effect of VEGF on the migration of ASMCs and its related signaling pathway mechanism to provide evidence for the treatment of airway remodeling. METHODS We examined the effects of VEGF induction on ASMC migration and explored the mechanisms involved in ASMC migration. RESULTS We found by wound healing and Transwell assays that VEGF promoted ASMC migration. Through the Cell Counting Kit-8 (CCK-8) experiment, we found that VEGF had no significant effect on the proliferation of ASMCs, which excluded the involvement of cell proliferation in the process of wound healing. Moreover, a cellular immunofluorescence assay showed that VEGF promoted F-actin reorganization, and Western blotting showed that VEGF improved RhoA activation and myosin phosphatase targeting subunit-1 (MYPT1) and myosin light chain (MLC) phosphorylation in ASMCs. Treatment with the ROCK inhibitor Y27632 significantly attenuated the effects of VEGF on MYPT1/MLC activation and cell migration. CONCLUSION In conclusion, the results suggest that the promigratory function of VEGF activates the RhoA/ROCK pathway, induces F-actin reorganization, improves the migration of ASMCs, and provides a better rationale for targeting the RhoA/ROCK pathway for therapeutic approaches in airway remodeling.
Collapse
Affiliation(s)
- Chengtian Lv
- Department of Pulmonary and Critical Care Medicine; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuwen Huang
- Department of Pulmonary and Critical Care Medicine; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruirong Yan
- Department of Pulmonary and Critical Care Medicine; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuanmei Gao
- Department of Pulmonary and Critical Care Medicine; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Assoian RK, Xu T, Roberts E. Arterial mechanics, extracellular matrix, and smooth muscle differentiation in carotid arteries deficient for Rac1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.15.567271. [PMID: 38014108 PMCID: PMC10680774 DOI: 10.1101/2023.11.15.567271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Stiffening of the extracellular matrix (ECM) occurs after vascular injury and contributes to the injury-associated proliferation of vascular smooth muscle cells (SMCs). ECM stiffness also activates Rac-GTP, and SMC Rac1 deletion strongly reduces the proliferative response to injury in vivo . However, ECM stiffening and Rac can affect SMC differentiation, which, in itself, can influence ECM stiffness and proliferation. Here, we used pressure myography and immunofluorescence analysis of mouse carotid arteries to ask if the reported effect of Rac1 deletion on in vivo SMC proliferation might be secondary to a Rac effect on basal arterial stiffness or SMC differentiation. The results show that Rac1 deletion does not affect the abundance of arterial collagen-I, -III, or -V, the integrity of arterial elastin, or the arterial responses to pressure, including the axial and circumferential stretch-strain relationships that are assessments of arterial stiffness. Medial abundance of alpha-smooth muscle actin and smooth muscle-myosin heavy chain, markers of the SMC differentiated phenotype, were not statistically different in carotid arteries containing or deficient in Rac1. Nor did Rac1 deficiency have a statistically significant effect on carotid artery contraction to KCl. Overall, these data argue that the inhibitory effect of Rac1 deletion on in vivo SMC proliferation reflects a primary effect of Rac1 signaling to the cell cycle rather than a secondary effect associated with altered SMC differentiation or arterial stiffness.
Collapse
|
19
|
Li XM, Wang SP, Wang JY, Tang T, Wan B, Zeng L, Wang J, Chu BB, Yang GY, Pan JJ. RhoA suppresses pseudorabies virus replication in vitro. Virol J 2023; 20:264. [PMID: 37968757 PMCID: PMC10652432 DOI: 10.1186/s12985-023-02229-2] [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: 08/29/2023] [Accepted: 11/05/2023] [Indexed: 11/17/2023] Open
Abstract
The porcine pseudorabies virus (PRV) is one of the most devastating pathogens and brings great economic losses to the swine industry worldwide. Viruses are intracellular parasites that have evolved numerous strategies to subvert and utilize different host processes for their life cycle. Among the different systems of the host cell, the cytoskeleton is one of the most important which not only facilitate viral invasion and spread into neighboring cells, but also help viruses to evade the host immune system. RhoA is a key regulator of cytoskeleton system that may participate in virus infection. In this study, we characterized the function of RhoA in the PRV replication by chemical drugs treatment, gene knockdown and gene over-expression strategy. Inhibition of RhoA by specific inhibitor and gene knockdown promoted PRV proliferation. On the contrary, overexpression of RhoA or activation of RhoA by chemical drug inhibited PRV infection. Besides, our data demonstrated that PRV infection induced the disruption of actin stress fiber, which was consistent with previous report. In turn, the actin specific inhibitor cytochalasin D markedly disrupted the normal fibrous structure of intracellular actin cytoskeleton and decreased the PRV replication, suggesting that actin cytoskeleton polymerization contributed to PRV replication in vitro. In summary, our data displayed that RhoA was a host restriction factor that inhibited PRV replication, which may deepen our understanding the pathogenesis of PRV and provide further insight into the prevention of PRV infection and the development of anti-viral drugs.
Collapse
Affiliation(s)
- Xin-Man Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
| | - Shi-Ping Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
| | - Jin-Yuan Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
| | - Ting Tang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
| | - Bo Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, 450046, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, 450046, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, 450046, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, 450047, China
| | - Jia-Jia Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, 450046, China.
- Key Laboratory of Animal Growth and Development of Henan Province, Zhengzhou, 450046, China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
20
|
Dang I, Brazzo JA, Bae Y, Assoian RK. Key role for Rac in the early transcriptional response to extracellular matrix stiffness and stiffness-dependent repression of ATF3. J Cell Sci 2023; 136:jcs260636. [PMID: 37737020 PMCID: PMC10617619 DOI: 10.1242/jcs.260636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
The Rho family GTPases Rac and Rho play critical roles in transmitting mechanical information contained within the extracellular matrix (ECM) to the cell. Rac and Rho have well-described roles in regulating stiffness-dependent actin remodeling, proliferation and motility. However, much less is known about the relative roles of these GTPases in stiffness-dependent transcription, particularly at the genome-wide level. Here, we selectively inhibited Rac and Rho in mouse embryonic fibroblasts cultured on deformable substrata and used RNA sequencing to elucidate and compare the contribution of these GTPases to the early transcriptional response to ECM stiffness. Surprisingly, we found that the stiffness-dependent activation of Rac was dominant over Rho in the initial transcriptional response to ECM stiffness. We also identified activating transcription factor 3 (ATF3) as a major target of stiffness- and Rac-mediated signaling and show that ATF3 repression by ECM stiffness helps to explain how the stiffness-dependent activation of Rac results in the induction of cyclin D1.
Collapse
Affiliation(s)
- Irène Dang
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Richard K. Assoian
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Punessen NC, Pena C, Sandberg A, Koza LA, Linseman DA. A novel anti-apoptotic role for Cdc42/ACK-1 signaling in neurons. Mol Cell Neurosci 2023; 126:103865. [PMID: 37263460 DOI: 10.1016/j.mcn.2023.103865] [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: 04/01/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023] Open
Abstract
Neurodegenerative diseases such as amyotrophic lateral sclerosis, Alzheimer's and Parkinson's disease are caused by a progressive and aberrant destruction of neurons in the brain and spinal cord. These disorders lack effective long-term treatments that impact the underlying mechanisms of pathogenesis and as a result, existing options focus primarily on alleviating symptomology. Dysregulated programmed cell death (i.e., apoptosis) is a significant contributor to neurodegeneration, and is controlled by a number of different factors. Rho family GTPases are molecular switches with recognized importance in proper neuronal development and migration that have more recently emerged as central regulators of apoptosis and neuronal survival. Here, we investigated a role for the Rho GTPase family member, Cdc42, and its downstream effectors, in neuronal survival and apoptosis. We initially induced apoptosis in primary cultures of rat cerebellar granule neurons (CGNs) by removing both growth factor-containing serum and depolarizing potassium from the cell medium. We then utilized both chemical inhibitors and adenoviral shRNA targeted to Cdc42 to block the function of Cdc42 or its downstream effectors under either control or apoptotic conditions. Our in vitro studies demonstrate that functional inhibition of Cdc42 or its downstream effector, activated Cdc42-associated tyrosine kinase-1 (ACK-1), had no adverse effects on CGN survival under control conditions, but significantly sensitized neurons to cell death under apoptotic conditions. In conclusion, our results suggest a key pro-survival role for Cdc42/ACK-1 signaling in neurons, particularly in regulating neuronal susceptibility to pro-apoptotic stress such as that observed in neurodegenerative disorders.
Collapse
Affiliation(s)
- Noelle C Punessen
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Claudia Pena
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Alexandra Sandberg
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Lilia A Koza
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Daniel A Linseman
- Department of Biological Sciences, University of Denver, Denver, CO, USA; Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA.
| |
Collapse
|
22
|
Bang J, Son KH, Heo HR, Park E, Kwak HJ, Uhm KO, Chung MH, Kim YY, Lim HJ. Exogenous 8-Hydroxydeoxyguanosine Attenuates PM 2.5-Induced Inflammation in Human Bronchial Epithelial Cells by Decreasing NLRP3 Inflammasome Activation. Antioxidants (Basel) 2023; 12:1189. [PMID: 37371919 DOI: 10.3390/antiox12061189] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/20/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Particulate matter 2.5 (PM2.5) induces lung injury by increasing the generation of reactive oxygen species (ROS) and inflammation. ROS aggravates NLRP3 inflammasome activation, which activates caspase-1, IL-1β, and IL-18 and induces pyroptosis; these factors propagate inflammation. In contrast, treatment with exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases RAC1 activity and eventually decreases dinucleotide phosphate oxidase (NOX) and ROS generation. To establish modalities that would mitigate PM2.5-induced lung injury, we evaluated whether 8-OHdG decreased PM2.5-induced ROS generation and NLRP3 inflammasome activation in BEAS-2B cells. CCK-8 and lactate dehydrogenase assays were used to determine the treatment concentration. Fluorescence intensity, Western blotting, enzyme-linked immunosorbent assay, and immunoblotting assays were also performed. Treatment with 80 μg/mL PM2.5 increased ROS generation, RAC1 activity, NOX1 expression, NLRP3 inflammasome (NLRP3, ASC, and caspase-1) activity, and IL-1β and IL-18 levels in cells; treatment with 10 μg/mL 8-OHdG significantly attenuated these effects. Furthermore, similar results, such as reduced expression of NOX1, NLRP3, ASC, and caspase-1, were observed in PM2.5-treated BEAS-2B cells when treated with an RAC1 inhibitor. These results show that 8-OHdG mitigates ROS generation and NLRP3 inflammation by inhibiting RAC1 activity and NOX1 expression in respiratory cells exposed to PM2.5.
Collapse
Affiliation(s)
- Jihye Bang
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Kuk Hui Son
- Gachon University Gil Medical Center, Department of Thoracic and Cardiovascular Surgery, College of Medicine, Gachon University, 21, Namdong-daero 774 beon-gil, Namdong-gu, Incheon 21565, Republic of Korea
| | - Hye-Ryeon Heo
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Eunsook Park
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Hyun-Jeong Kwak
- Major of Life Science, Division of Bioconvergence, College of Convergence and Integrated Science, Kyonggi University, 154-42 Gwanggosan-ro, Yeongtong-gu, Suwon-si 16227, Republic of Korea
| | - Kyung-Ok Uhm
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Myung-Hee Chung
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| | - Young-Youl Kim
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Hyun Joung Lim
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| |
Collapse
|
23
|
Grande V, Schuld J, van der Ven PFM, Gruss OJ, Fürst DO. Filamin-A-interacting protein 1 (FILIP1) is a dual compartment protein linking myofibrils and microtubules during myogenic differentiation and upon mechanical stress. Cell Tissue Res 2023:10.1007/s00441-023-03776-4. [PMID: 37178194 DOI: 10.1007/s00441-023-03776-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023]
Abstract
Variations in the gene encoding filamin-A-interacting protein 1 (FILIP1) were identified to be associated with a combination of neurological and muscular symptoms. While FILIP1 was shown to regulate motility of brain ventricular zone cells, a process important for corticogenesis, the function of the protein in muscle cells has been less well characterized. The expression of FILIP1 in regenerating muscle fibres predicted a role in early muscle differentiation. Here we analysed expression and localization of FILIP1 and its binding partners filamin-C (FLNc) and microtubule plus-end-binding protein EB3 in differentiating cultured myotubes and adult skeletal muscle. Prior to the development of cross-striated myofibrils, FILIP1 is associated with microtubules and colocalizes with EB3. During further myofibril maturation its localization changes, and FILIP1 localizes to myofibrillar Z-discs together with the actin-binding protein FLNc. Forced contractions of myotubes by electrical pulse stimulation (EPS) induce focal disruptions in myofibrils and translocation of both proteins from Z-discs to these lesions, suggesting a role in induction and/or repair of these structures. The immediate proximity of tyrosylated, dynamic microtubules and EB3 to lesions implies that also these play a role in these processes. This implication is supported by the fact that in nocodazole-treated myotubes that lack functional microtubules, the number of lesions induced by EPS is significantly reduced. In summary, we here show that FILIP1 is a cytolinker protein that is associated with both microtubules and actin filaments, and might play a role in the assembly of myofibrils and their stabilization upon mechanical stress to protect them from damage.
Collapse
Affiliation(s)
- Valentina Grande
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121, Bonn, Germany
| | - Julia Schuld
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121, Bonn, Germany
| | - Peter F M van der Ven
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121, Bonn, Germany
| | - Oliver J Gruss
- Institute of Genetics, University of Bonn, Karlrobert-Kreiten-Str. 13, 53115, Bonn, Germany
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121, Bonn, Germany.
| |
Collapse
|
24
|
Zhao RY, Wei PJ, Sun X, Zhang DH, He QY, Liu J, Chang JL, Yang Y, Guo ZN. Role of lipocalin 2 in stroke. Neurobiol Dis 2023; 179:106044. [PMID: 36804285 DOI: 10.1016/j.nbd.2023.106044] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/22/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Stroke is the second leading cause of death worldwide; however, the treatment choices available to neurologists are limited in clinical practice. Lipocalin 2 (LCN2) is a secreted protein, belonging to the lipocalin superfamily, with multiple biological functions in mediating innate immune response, inflammatory response, iron-homeostasis, cell migration and differentiation, energy metabolism, and other processes in the body. LCN2 is expressed at low levels in the brain under normal physiological conditions, but its expression is significantly up-regulated in multiple acute stimulations and chronic pathologies. An up-regulation of LCN2 has been found in the blood/cerebrospinal fluid of patients with ischemic/hemorrhagic stroke, and could serve as a potential biomarker for the prediction of the severity of acute stroke. LCN2 activates reactive astrocytes and microglia, promotes neutrophil infiltration, amplifies post-stroke inflammation, promotes blood-brain barrier disruption, white matter injury, and neuronal death. Moreover, LCN2 is involved in brain injury induced by thrombin and erythrocyte lysates, as well as microvascular thrombosis after hemorrhage. In this paper, we review the role of LCN2 in the pathological processes of ischemic stroke; intracerebral hemorrhage; subarachnoid hemorrhage; and stroke-related brain diseases, such as vascular dementia and post-stroke depression, and their underlying mechanisms. We hope that this review will help elucidate the value of LCN2 as a therapeutic target in stroke.
Collapse
Affiliation(s)
- Ruo-Yu Zhao
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Peng-Ju Wei
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Sun
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Jie Liu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Jun-Lei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, the First Hospital of Jilin University, Chang Chun, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, the First Hospital of Jilin University, Chang Chun, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China.
| |
Collapse
|
25
|
Light-driven biological actuators to probe the rheology of 3D microtissues. Nat Commun 2023; 14:717. [PMID: 36759504 PMCID: PMC9911700 DOI: 10.1038/s41467-023-36371-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
The mechanical properties of biological tissues are key to their physical integrity and function. Although external loading or biochemical treatments allow the estimation of these properties globally, it remains difficult to assess how such external stimuli compare with cell-generated contractions. Here we engineer microtissues composed of optogenetically-modified fibroblasts encapsulated within collagen. Using light to control the activity of RhoA, a major regulator of cellular contractility, we induce local contractions within microtissues, while monitoring microtissue stress and strain. We investigate the regulation of these local contractions and their spatio-temporal distribution. We demonstrate the potential of our technique for quantifying tissue elasticity and strain propagation, before examining the possibility of using light to create and map local anisotropies in mechanically heterogeneous microtissues. Altogether, our results open an avenue to guide the formation of tissues while non-destructively charting their rheology in real time, using their own constituting cells as internal actuators.
Collapse
|
26
|
Reeb T, Rhea L, Adelizzi E, Garnica B, Dunnwald E, Dunnwald M. ARHGAP29 is required for keratinocyte proliferation and migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.525978. [PMID: 36778214 PMCID: PMC9915469 DOI: 10.1101/2023.01.30.525978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND RhoA GTPase plays critical roles in actin cytoskeletal remodeling required for controlling a diverse range of cellular functions including cell proliferation, cell adhesions, migration and changes in cell shape. RhoA cycles between an active GTP-bound and an inactive GDP-bound form, a process that is regulated by guanine nucleotide exchange factors (GEFs), and GTPase-activating proteins (GAPs). ARHGAP29 is a GAP expressed in keratinocytes of the skin and is decreased in the absence of Interferon Regulator Factor 6, a critical regulator of cell proliferation and migration. However, the role for ARHGAP29 in keratinocyte biology is unknown. RESULTS Novel ARHGAP29 knockdown keratinocyte cell lines were generated using both CRISPR/Cas9 and shRNA technologies. Knockdown cells exhibited significant reduction of ARHGAP29 protein (50-80%) and displayed increased filamentous actin (stress fibers), phospho-myosin light chain (contractility), cell area and population doubling time. Furthermore, we found that ARHGAP29 knockdown keratinocytes displayed significant delays in scratch wound closure in both single cell and collective cell migration conditions. Particularly, our results show a reduction in path lengths, speed, directionality and persistence in keratinocytes with reduced ARHGAP29. The delay in scratch closure was rescued by both adding back ARHGAP29 or adding a ROCK inhibitor to ARHGAP29 knockdown cells. CONCLUSIONS These data demonstrate that ARHGAP29 is required for keratinocyte morphology, proliferation and migration mediated through the RhoA pathway.
Collapse
|
27
|
Liu X, Wang M, Zhang L, Huang L. LncRNA ZFAS1 contributes to osteosarcoma progression via miR-520b and miR-520e-mediated inhibition of RHOC signaling. Clinics (Sao Paulo) 2023; 78:100143. [PMID: 36473367 PMCID: PMC9727593 DOI: 10.1016/j.clinsp.2022.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES We examined the expression of Lnc-ZFAS1 in osteosarcoma and comprehensively evaluated its effects on osteosarcoma in vitro and vivo. Moreover, we revealed the regulatory mechanism between Lnc-ZFAS1 and miR-520b/miR-520e-mediated RHOC and provided a novel clue for ameliorating osteosarcoma. METHOD The expression of Long non-coding RNA Zinc Finger Antisense 1 (LncRNA ZFAS1) osteosarcoma tissues and normal tissues in the TCGA database was analyzed. Then, LncRNA ZFAS1 expression was further verified in clinical samples and osteosarcoma cell lines (U2OS and KHOS), as well as the human osteoblast cell line hFOB1.19 by qRT-PCR. Thereafter, LncRNA ZFAS1 was overexpressed or silenced to explore its effects on cell proliferation, apoptosis, migration, invasion, and Epithelial-Mesenchymal Transition (EMT). The fundamental mechanism through which Lnc-ZFAS1 affects osteosarcoma progression was further investigated and verified. RESULTS We found that LncRNA ZFAS1 was upregulated in osteosarcoma, and Lnc-ZFAS1 overexpression facilitated osteosarcoma cells proliferation, migration, invasion and EMT, while Lnc-ZFAS1 silence exerted reverse influence. Mechanistically, Lnc-ZFAS1 functionally acted as a sponger of microRNA-520b (miR-520b) and microRNA-520e (miR-520e) to up-regulate Ras Homologue C (RHOC). In addition, depleted Lnc-ZFAS1 restrained osteosarcoma cells proliferation, migration, and invasion, which could be rescued by RHOC overexpression. Lnc-ZFAS1 was upregulated in osteosarcoma and Lnc-ZFAS1 could exert promoted impact upon osteosarcoma cells proliferation, migration, invasion, and EMT in vitro. CONCLUSIONS Lnc-ZFAS1 acted sponger of miR-520b and miR-520e to promote RHOC, indicating that Lnc-ZFAS1/miR-520b/RHOC and Lnc-ZFAS1/miR-520e/RHOC axes might serve as potential therapeutic strategies against osteosarcoma.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China
| | - Mingyang Wang
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China
| | - Liwen Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China
| | - Lei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Dalian Medical University, Liaoning, People's Republic of China.
| |
Collapse
|
28
|
Dock10 Regulates Cardiac Function under Neurohormonal Stress. Int J Mol Sci 2022; 23:ijms23179616. [PMID: 36077014 PMCID: PMC9455810 DOI: 10.3390/ijms23179616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022] Open
Abstract
Dedicator of cytokinesis 10 (Dock10) is a guanine nucleotide exchange factor for Cdc42 and Rac1 that regulates the JNK (c-Jun N-terminal kinase) and p38 MAPK (mitogen-activated protein kinase) signaling cascades. In this study, we characterized the roles of Dock10 in the myocardium. In vitro: we ablated Dock10 in neonatal mouse floxed Dock10 cardiomyocytes (NMCMs) and cardiofibroblasts (NMCFs) by transduction with an adenovirus expressing Cre-recombinase. In vivo, we studied mice in which the Dock10 gene was constitutively and globally deleted (Dock10 KO) and mice with cardiac myocyte-specific Dock10 KO (Dock10 CKO) at baseline and in response to two weeks of Angiotensin II (Ang II) infusion. In vitro, Dock10 ablation differentially inhibited the α-adrenergic stimulation of p38 and JNK in NMCM and NMCF, respectively. In vivo, the stimulation of both signaling pathways was markedly attenuated in the heart. The Dock10 KO mice had normal body weight and cardiac size. However, echocardiography revealed mildly reduced systolic function, and IonOptix recordings demonstrated reduced contractility and elevated diastolic calcium levels in isolated cardiomyocytes. Remarkably, Dock10 KO, but not Dock10 CKO, exaggerated the pathological response to Ang II infusion. These data suggest that Dock10 regulates cardiac stress-related signaling. Although Dock10 can regulate MAPK signaling in both cardiomyocytes and cardiofibroblasts, the inhibition of pathological cardiac remodeling is not apparently due to the Dock10 signaling in the cardiomyocyte.
Collapse
|
29
|
Ahangar P, Strudwick XL, Cowin AJ. Wound Healing from an Actin Cytoskeletal Perspective. Cold Spring Harb Perspect Biol 2022; 14:a041235. [PMID: 35074864 PMCID: PMC9341468 DOI: 10.1101/cshperspect.a041235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing requires a complex cascade of highly controlled and conserved cellular and molecular processes. These involve numerous cell types and extracellular matrix molecules regulated by the actin cytoskeleton. This microscopic network of filaments is present within the cytoplasm of all cells and provides the shape and mechanical support required for cell movement and proliferation. Here, an overview of the processes of wound healing are described from the perspective of the cell in relation to the actin cytoskeleton. Key points of discussion include the role of actin, its binding proteins, signaling pathways, and events that play significant roles in the phases of wound healing. The identification of cytoskeletal targets that can be used to manipulate and improve wound healing is included as an emerging area of focus that may inform future therapeutic approaches to improve healing of complex wounds.
Collapse
Affiliation(s)
- Parinaz Ahangar
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Allison J Cowin
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| |
Collapse
|
30
|
Shutova MS, Boehncke WH. Mechanotransduction in Skin Inflammation. Cells 2022; 11:2026. [PMID: 35805110 PMCID: PMC9265324 DOI: 10.3390/cells11132026] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
In the process of mechanotransduction, the cells in the body perceive and interpret mechanical stimuli to maintain tissue homeostasis and respond to the environmental changes. Increasing evidence points towards dysregulated mechanotransduction as a pathologically relevant factor in human diseases, including inflammatory conditions. Skin is the organ that constantly undergoes considerable mechanical stresses, and the ability of mechanical factors to provoke inflammatory processes in the skin has long been known, with the Koebner phenomenon being an example. However, the molecular mechanisms and key factors linking mechanotransduction and cutaneous inflammation remain understudied. In this review, we outline the key players in the tissue's mechanical homeostasis, the available data, and the gaps in our current understanding of their aberrant regulation in chronic cutaneous inflammation. We mainly focus on psoriasis as one of the most studied skin inflammatory diseases; we also discuss mechanotransduction in the context of skin fibrosis as a result of chronic inflammation. Even though the role of mechanotransduction in inflammation of the simple epithelia of internal organs is being actively studied, we conclude that the mechanoregulation in the stratified epidermis of the skin requires more attention in future translational research.
Collapse
Affiliation(s)
- Maria S. Shutova
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| |
Collapse
|
31
|
Sarkar P, Kumar GA, Shrivastava S, Chattopadhyay A. Chronic cholesterol depletion increases F-actin levels and induces cytoskeletal reorganization via a dual mechanism. J Lipid Res 2022; 63:100206. [PMID: 35390404 PMCID: PMC9096963 DOI: 10.1016/j.jlr.2022.100206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Previous work from us and others has suggested that cholesterol is an important lipid in the context of the organization of the actin cytoskeleton. However, reorganization of the actin cytoskeleton upon modulation of membrane cholesterol is rarely addressed in the literature. In this work, we explored the signaling crosstalk between cholesterol and the actin cytoskeleton by using a high-resolution confocal microscopic approach to quantitatively measure changes in F-actin content upon cholesterol depletion. Our results show that F-actin content significantly increases upon chronic cholesterol depletion, but not during acute cholesterol depletion. In addition, utilizing inhibitors targeting the cholesterol biosynthetic pathway at different steps, we show that reorganization of the actin cytoskeleton could occur due to the synergistic effect of multiple pathways, including prenylated Rho GTPases and availability of membrane phosphatidylinositol 4,5-bisphosphate. These results constitute one of the first comprehensive dissections of the mechanistic basis underlying the interplay between cellular actin levels and cholesterol biosynthesis. We envision these results will be relevant for future understating of the remodeling of the actin cytoskeleton in pathological conditions with altered cholesterol.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
32
|
De Ieso ML, Kuhn M, Bernatchez P, Elliott MH, Stamer WD. A Role of Caveolae in Trabecular Meshwork Mechanosensing and Contractile Tone. Front Cell Dev Biol 2022; 10:855097. [PMID: 35372369 PMCID: PMC8969750 DOI: 10.3389/fcell.2022.855097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Polymorphisms in the CAV1/2 gene loci impart increased risk for primary open-angle glaucoma (POAG). CAV1 encodes caveolin-1 (Cav1), which is required for biosynthesis of plasma membrane invaginations called caveolae. Cav1 knockout mice exhibit elevated intraocular pressure (IOP) and decreased outflow facility, but the mechanistic role of Cav1 in IOP homeostasis is unknown. We hypothesized that caveolae sequester/inhibit RhoA, to regulate trabecular meshwork (TM) mechanosensing and contractile tone. Using phosphorylated myosin light chain (pMLC) as a surrogate indicator for Rho/ROCK activity and contractile tone, we found that pMLC was elevated in Cav1-deficient TM cells compared to control (131 ± 10%, n = 10, p = 0.016). Elevation of pMLC levels following Cav1 knockdown occurred in cells on a soft surface (137 ± 7%, n = 24, p < 0.0001), but not on a hard surface (122 ± 17%, n = 12, p = 0.22). In Cav1-deficient TM cells where pMLC was elevated, Rho activity was also increased (123 ± 7%, n = 6, p = 0.017), suggesting activation of the Rho/ROCK pathway. Cyclic stretch reduced pMLC/MLC levels in TM cells (69 ± 7% n = 9, p = 0.002) and in Cav1-deficient TM cells, although not significantly (77 ± 11% n = 10, p = 0.059). Treatment with the Cav1 scaffolding domain mimetic, cavtratin (1 μM) caused a reduction in pMLC (70 ± 5% n = 7, p = 0.001), as did treatment with the scaffolding domain mutant cavnoxin (1 μM) (82 ± 7% n = 7, p = 0.04). Data suggest that caveolae differentially regulate RhoA signaling, and that caveolae participate in TM mechanotransduction. Cav1 regulation of these key TM functions provide evidence for underlying mechanisms linking polymorphisms in the Cav1/2 gene loci with increased POAG risk.
Collapse
Affiliation(s)
- Michael L. De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, United States
| | - Megan Kuhn
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, United States
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Heart + Lung Innovation Centre, St. Paul’s Hospital, Vancouver, BC, Canada
| | - Michael H. Elliott
- Department of Ophthalmology, Dean McGee Eye Institute University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC, United States
| |
Collapse
|
33
|
CIP4 targeted to recruit GTP-Cdc42 involving in invadopodia formation via NF-κB signaling pathway promotes invasion and metastasis of CRC. Mol Ther Oncolytics 2022; 24:873-886. [PMID: 35317515 PMCID: PMC8924540 DOI: 10.1016/j.omto.2022.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/21/2022] [Indexed: 12/04/2022] Open
Abstract
Cdc42-interacting protein 4 (CIP4), a member of the F-BAR family, which plays an important role in regulating cell membrane and actin, has been reported to interact with Cdc42 and be closely associated with tumor invadopodia formation. In this study, we found that CIP4 expression was significantly higher in human CRC tissues and correlated with the CRC infiltrating depth and metastasis, as well as the lower survival rate in patients. In cultured CRC cells, knockdown of CIP4 inhibited cell migration and invasion ability in vitro and tumor metastasis in vivo, while the overexpression of CIP4 promoted invadopodia formation and matrix degradation ability. We then identified GTP-Cdc42 as a directly interactive protein of CIP4, which was upregulated and recruited by CIP4. Furthermore, activated NF-κB signaling pathway was found in CIP4 overexpression of CRC cells contributing to invadopodia formation, while the inhibition of either CIP4 or Cdc42 led to the suppression of the NF-κB pathway and resulted in a decreased quantity of invadopodia. Our findings suggested that CIP4 targets to recruit GTP-Cdc42 and directly combines with it to accelerate invadopodia formation and function by activating NF-κB signaling pathway, thus promoting CRC infiltration and metastasis.
Collapse
|
34
|
To Stick or Not to Stick: Adhesions in Orofacial Clefts. BIOLOGY 2022; 11:biology11020153. [PMID: 35205020 PMCID: PMC8869391 DOI: 10.3390/biology11020153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 11/17/2022]
Abstract
Morphogenesis requires a tight coordination between mechanical forces and biochemical signals to inform individual cellular behavior. For these developmental processes to happen correctly the organism requires precise spatial and temporal coordination of the adhesion, migration, growth, differentiation, and apoptosis of cells originating from the three key embryonic layers, namely the ectoderm, mesoderm, and endoderm. The cytoskeleton and its remodeling are essential to organize and amplify many of the signaling pathways required for proper morphogenesis. In particular, the interaction of the cell junctions with the cytoskeleton functions to amplify the behavior of individual cells into collective events that are critical for development. In this review we summarize the key morphogenic events that occur during the formation of the face and the palate, as well as the protein complexes required for cell-to-cell adhesions. We then integrate the current knowledge into a comprehensive review of how mutations in cell-to-cell adhesion genes lead to abnormal craniofacial development, with a particular focus on cleft lip with or without cleft palate.
Collapse
|
35
|
Wen Q, Weng H, Liu T, Yu L, Zhao T, Qin J, Li S, Wu Q, Fadel T, Qu Y, Zhou L. Inactivating Celsr2 promotes motor axon fasciculation and regeneration in mouse and human. Brain 2022; 145:670-683. [PMID: 34983065 PMCID: PMC9014747 DOI: 10.1093/brain/awab317] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 11/18/2022] Open
Abstract
Understanding new modulators of axon regeneration is central to neural repair. Our previous work demonstrated critical roles of atypical cadherin Celsr2 during neural development, including cilia organization, neuron migration and axon navigation. Here, we address its role in axon regeneration. We show that Celsr2 is highly expressed in both mouse and human spinal motor neurons. Celsr2 knockout promotes axon regeneration and fasciculation in mouse cultured spinal explants. Similarly, cultured Celsr2 mutant motor neurons extend longer neurites and larger growth cones, with increased expression of end-binding protein 3 and higher potassium-induced calcium influx. Mice with Celsr2 conditional knockout in spinal motor neurons do not exhibit any behavioural deficits; however, after branchial plexus injury, axon regeneration and functional forelimb locomotor recovery are significantly improved. Similarly, knockdown of CELSR2 using shRNA interference in cultured human spinal motor explants and motor neurons increases axonal fasciculation and growth. In mouse adult spinal cord after root avulsion, in mouse embryonic spinal cords, and in cultured human motor neurons, Celsr2 downregulation is accompanied by increased levels of GTP-bound Rac1 and Cdc42, and of JNK and c-Jun. In conclusion, Celsr2 negatively regulates motor axon regeneration and is a potential target to improve neural repair.
Collapse
Affiliation(s)
- Quan Wen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, P. R. China
| | - Huandi Weng
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, P. R. China
| | - Tao Liu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, P. R. China
| | - Lingtai Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, P. R. China
| | - Tainyun Zhao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R. China
| | - Jingwen Qin
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R. China
| | - Si Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Qingfeng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Tissir Fadel
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium.,College of Life and Health Sciences, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Yibo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, P. R. China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain- Inspired Intelligence, Guangzhou 510515, P.R. China.,Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, P. R. China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, P. R. China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, P.R. China.,Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, P. R. China.,The first affiliated hospital of Jian University, Guangzhou 510632, P. R. China
| |
Collapse
|
36
|
Mei Q, Li H, Liu Y, Wang X, Xiang W. Advances in the study of CDC42 in the female reproductive system. J Cell Mol Med 2021; 26:16-24. [PMID: 34859585 PMCID: PMC8742232 DOI: 10.1111/jcmm.17088] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
CDC42 is a member of the Rho‐GTPase family and is involved in a variety of cellular functions including regulation of cell cycle progression, constitution of the actin backbone and membrane transport. In particular, CDC42 plays a key role in the establishment of polarity in female vertebrate oocytes, and essential to this major regulatory role is its local occupation of specific regions of the cell to ensure that the contractile ring is assembled at the right time and place to ensure proper gametogenesis. The multifactor controlled ‘inactivation‐activation’ process of CDC42 also allows it to play an important role in the multilevel signalling network, and the synergistic regulation of multiple genes ensures maximum precision during gametogenesis. The purpose of this paper is to review the role of CDC42 in the control of gametogenesis and to explore its related mechanisms, with the aim of further understanding the great research potential of CDC42 in female vertebrate germ cells and its future clinical translation.
Collapse
Affiliation(s)
- Qiaojuan Mei
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiying Li
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Liu
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Wang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpei Xiang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Graziani V, Rodriguez-Hernandez I, Maiques O, Sanz-Moreno V. The amoeboid state as part of the epithelial-to-mesenchymal transition programme. Trends Cell Biol 2021; 32:228-242. [PMID: 34836782 DOI: 10.1016/j.tcb.2021.10.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023]
Abstract
Cell migration is essential for many biological processes, while abnormal cell migration is characteristic of cancer cells. Epithelial cells become motile by undergoing epithelial-to-mesenchymal transition (EMT), and mesenchymal cells increase migration speed by adopting amoeboid features. This review highlights how amoeboid behaviour is not merely a migration mode but rather a cellular state - within the EMT spectra - by which cancer cells survive, invade and colonise challenging microenvironments. Molecular biomarkers and physicochemical triggers associated with amoeboid behaviour are discussed, including an amoeboid associated tumour microenvironment. We reflect on how amoeboid characteristics support metastasis and how their liabilities could turn into therapeutic opportunities.
Collapse
Affiliation(s)
- Vittoria Graziani
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | | | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | | |
Collapse
|
38
|
McGarry DJ, Armstrong G, Castino G, Mason S, Clark W, Shaw R, McGarry L, Blyth K, Olson MF. MICAL1 regulates actin cytoskeleton organization, directional cell migration and the growth of human breast cancer cells as orthotopic xenograft tumours. Cancer Lett 2021; 519:226-236. [PMID: 34314753 DOI: 10.1016/j.canlet.2021.07.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 02/09/2023]
Abstract
The Molecule Interacting with CasL 1 (MICAL1) monooxygenase has emerged as an important regulator of cytoskeleton organization via actin oxidation. Although filamentous actin (F-actin) increases MICAL1 monooxygenase activity, hydrogen peroxide (H2O2) is also generated in the absence of F-actin, suggesting that diffusible H2O2 might have additional functions. MICAL1 gene disruption by CRISPR/Cas9 in MDA MB 231 human breast cancer cells knocked out (KO) protein expression, which affected F-actin organization, cell size and motility. Transcriptomic profiling revealed that MICAL1 deletion significantly affected the expression of over 700 genes, with the majority being reduced in their expression levels. In addition, the absolute magnitudes of reduced gene expression were significantly greater than the magnitudes of increased gene expression. Gene set enrichment analysis (GSEA) identified receptor regulator activity as the most significant negatively enriched molecular function gene set. The prominent influence exerted by MICAL1 on F-actin structures was also associated with changes in the expression of several serum-response factor (SRF) regulated genes in KO cells. Moreover, MICAL1 disruption attenuated breast cancer tumour growth in vivo. Elevated MICAL1 gene expression was observed in invasive breast cancer samples from human patients relative to normal tissue, while MICAL1 amplification or point mutations were associated with reduced progression free survival. Collectively, these results demonstrate that MICAL1 gene disruption altered cytoskeleton organization, cell morphology and migration, gene expression, and impaired tumour growth in an orthotopic in vivo breast cancer model, suggesting that pharmacological MICAL1 inhibition could have therapeutic benefits for cancer patients.
Collapse
Affiliation(s)
- David J McGarry
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Garett Armstrong
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Giovanni Castino
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Susan Mason
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Robin Shaw
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Lynn McGarry
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Michael F Olson
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
39
|
Gahankari A, Dong C, Bartoletti G, Galazo M, He F. Deregulated Rac1 Activity in Neural Crest Controls Cell Proliferation, Migration and Differentiation During Midbrain Development. Front Cell Dev Biol 2021; 9:704769. [PMID: 34557485 PMCID: PMC8452869 DOI: 10.3389/fcell.2021.704769] [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: 05/03/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Mutations in RAC1 allele are implicated in multiple brain tumors, indicating a rigorous control of Rac1 activity is required for neural tissue normal development and homeostasis. To understand how elevated Rac1 activity affects neural crest cells (NCCs) development, we have generated Rac1 CA ;Wnt1-Cre2 mice, in which a constitutively active Rac1 G12V mutant is expressed specifically in NCCs derivatives. Our results revealed that augmented Rac1 activity leads to enlarged midbrain and altered cell density, accompanied by increased NCCs proliferation rate and misrouted cell migration. Interestingly, our experimental data also showed that elevated Rac1 activity in NCCs disrupts regionalization of dopaminergic neuron progenitors in the ventral midbrain and impairs their differentiation. These findings shed light on the mechanisms of RAC1 mutation correlated brain tumor at the cellular and molecular level.
Collapse
Affiliation(s)
- Apurva Gahankari
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Chunmin Dong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Garrett Bartoletti
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Maria Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States.,Tulane Brain Institute, Tulane University, New Orleans, LA, United States
| | - Fenglei He
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
40
|
Cui D, Jiang X, Chen M, Sheng H, Shao D, Yang L, Guo X, Wang Y, Lai B, Zheng P. Activation of Rac1 Has an Opposing Effect on Induction and Maintenance of Long-Term Potentiation in Hippocampus by Acting on Different Kinases. Front Mol Neurosci 2021; 14:720371. [PMID: 34531724 PMCID: PMC8438208 DOI: 10.3389/fnmol.2021.720371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/27/2021] [Indexed: 11/21/2022] Open
Abstract
Rac1 is a small GTPase of the Rho family. A previous study showed that the activation of Rac1 had an opposing effect on induction and maintenance of long-term potentiation (LTP) in the hippocampus. However, the molecular mechanism underlying this opposing effect remains to be addressed. In the present work, we find that the activation of Rac1 during the induction of LTP leads to an activation of PKCι/λ by phosphatidylinositol-3-kinase (PI3K), whereas the activation of Rac1 during the maintenance of LTP leads to the inhibition of PKMζ by LIM_kinase (LIMK) in the hippocampus. This result suggests that during different stages of LTP, the activation of Rac1 can modulate different signaling pathways, which leads to an opposing effect on the induction and maintenance of LTP in the hippocampus.
Collapse
Affiliation(s)
- Dongyang Cui
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaodong Jiang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ming Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Huan Sheng
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Da Shao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xinli Guo
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yingqi Wang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bin Lai
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ping Zheng
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Center for Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of Neurology of Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pharmacology of Medical College of China Three Gorges University, Yichang, China
| |
Collapse
|
41
|
Qin L, Cao X, Kaneko T, Voss C, Liu X, Wang G, Li SSC. Dynamic interplay of two molecular switches enabled by the MEK1/2-ERK1/2 and IL-6-STAT3 signaling axes controls epithelial cell migration in response to growth factors. J Biol Chem 2021; 297:101161. [PMID: 34480897 PMCID: PMC8477194 DOI: 10.1016/j.jbc.2021.101161] [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: 03/01/2021] [Revised: 08/10/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Cell migration is an essential physiological process, and aberrant migration of epithelial cells underlies many pathological conditions. However, the molecular mechanisms governing cell migration are not fully understood. We report here that growth factor–induced epithelial cell migration is critically dependent on the crosstalk of two molecular switches, namely phosphorylation switch (P-switch) and transcriptional switch (T-switch). P-switch refers to dynamic interactions of deleted in liver cancer 1 (DLC1) and PI3K with tensin-3 (TNS3), phosphatase and tensin homolog (PTEN), C-terminal tension, and vav guanine nucleotide exchange factor 2 (VAV2) that are dictated by mitogen-activated protein kinase kinase 1/2–extracellular signal–regulated protein kinase 1/2–dependent phosphorylation of TNS3, PTEN, and VAV2. Phosphorylation of TNS3 and PTEN on specific Thr residues led to the switch of DLC1–TNS3 and PI3K–PTEN complexes to DLC1–PTEN and PI3K–TNS3 complexes, whereas Ser phosphorylation of VAV2 promotes the transition of the PI3K–TNS3/PTEN complexes to PI3K–VAV2 complex. T-switch denotes an increase in C-terminal tension transcription/expression regulated by both extracellular signal–regulated protein kinase 1/2 and signal transducer and activator of transcription 3 (STAT3) via interleukin-6–Janus kinase–STAT3 signaling pathway. We have found that, the P-switch is indispensable for both the initiation and continuation of cell migration induced by growth factors, whereas the T-switch is only required to sustain cell migration. The interplay of the two switches facilitated by the interleukin-6–Janus kinase–STAT3 pathway governs a sequence of dynamic protein–protein interactions for sustained cell migration. That a similar mechanism is employed by both normal and tumorigenic epithelial cells to drive their respective migration suggests that the P-switch and T-switch are general regulators of epithelial cell migration and potential therapeutic targets.
Collapse
Affiliation(s)
- Lyugao Qin
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Xuan Cao
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tomonori Kaneko
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Courtney Voss
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Xuguang Liu
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Guoping Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shawn S-C Li
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
42
|
Liang L, Liang X, Jiang P, Zhou L, Zhong L, Wang M, Lin S, Guo Z, Yu J, Yang C, Chen Y, Zhuo C, Chen P, Wang Y. Metastasis suppressor 1 interacts with α-actinin 4 to affect its localization and regulate formation of membrane ruffling. Cytoskeleton (Hoboken) 2021; 78:337-348. [PMID: 34435464 DOI: 10.1002/cm.21686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 01/21/2023]
Abstract
Membrane ruffling plays an important role in the directed cell migration and escape of tumor cells from the monolayer. Metastasis suppressor 1 (MTSS1), also known as missing in metastasis, has been implicated in cell morphology, motility, metastasis, and development. Here, the dynamic interaction proteins associated with MTSS1 and involved in membrane ruffling were determined by cross-linking and mass spectrometry analysis. We identified α-actinin 4 (ACTN4) as an interacting protein and confirmed a direct interaction between MTSS1 and ACTN4. Moreover, co-expression of MTSS1 in fibroblasts recruited cytoplasmic ACTN4 to the cell periphery, at which point ruffling became thick and rigid. In MCF-7 cells, MTSS1 knockdown did not show an obvious effect on the cell shape or the distribution of endogenous ACTN4; however, ACTN4 overexpression transformed cell morphology from an epidermal- to a fibroblast-like shape, and further MTSS1 depletion significantly increased the ratio of fibroblast cells exhibiting prominent ruffling. Furthermore, biochemical data suggested that MTSS1 cross-linking with ACTN4 induced the formation of actin fiber bundles into more organized structures in vitro. These data indicated that MTSS1 might recruit cytoplasmic ACTN4 to the cell periphery and regulate cytoskeleton dynamics to restrict its performance in membrane ruffling.
Collapse
Affiliation(s)
- Lijun Liang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaoping Liang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Peng Jiang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Lu Zhou
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Luanluan Zhong
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Mei Wang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Shuyun Lin
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Zhen Guo
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Juan Yu
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Changcheng Yang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yu Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Chengjie Zhuo
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ping Chen
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Ying Wang
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
43
|
Arts JJG, Mahlandt EK, Grönloh MLB, Schimmel L, Noordstra I, Gordon E, van Steen ACI, Tol S, Walzog B, van Rijssel J, Nolte MA, Postma M, Khuon S, Heddleston JM, Wait E, Chew TL, Winter M, Montanez E, Goedhart J, van Buul JD. Endothelial junctional membrane protrusions serve as hotspots for neutrophil transmigration. eLife 2021; 10:66074. [PMID: 34431475 PMCID: PMC8437435 DOI: 10.7554/elife.66074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/22/2021] [Indexed: 12/27/2022] Open
Abstract
Upon inflammation, leukocytes rapidly transmigrate across the endothelium to enter the inflamed tissue. Evidence accumulates that leukocytes use preferred exit sites, alhough it is not yet clear how these hotspots in the endothelium are defined and how they are recognized by the leukocyte. Using lattice light sheet microscopy, we discovered that leukocytes prefer endothelial membrane protrusions at cell junctions for transmigration. Phenotypically, these junctional membrane protrusions are present in an asymmetric manner, meaning that one endothelial cell shows the protrusion and the adjacent one does not. Consequently, leukocytes cross the junction by migrating underneath the protruding endothelial cell. These protrusions depend on Rac1 activity and by using a photo-activatable Rac1 probe, we could artificially generate local exit-sites for leukocytes. Overall, we have discovered a new mechanism that uses local induced junctional membrane protrusions to facilitate/steer the leukocyte escape/exit from inflamed vessel walls.
Collapse
Affiliation(s)
- Janine JG Arts
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Eike K Mahlandt
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Max LB Grönloh
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Lilian Schimmel
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Ivar Noordstra
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Emma Gordon
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of QueenslandBrisbaneQLDAustralia
| | - Abraham CI van Steen
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Simon Tol
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Barbara Walzog
- Department of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Biomedical Center, Ludwig-Maximilians-Universität MünchenPlanegg-MartinsriedGermany
| | - Jos van Rijssel
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Martijn A Nolte
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Marten Postma
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Satya Khuon
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - John M Heddleston
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- Microscopy Facility at the Cleveland Clinic Florida Research and Innovation CenterPort St. LucieUnited States
| | - Eric Wait
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Teng Leong Chew
- Advanced Imaging Center at Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Mark Winter
- Zuckerman Postdoctoral Fellow, Department of Marine Sciences, University of HaifaHaifaIsrael
| | - Eloi Montanez
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of BarcelonaBarcelonaSpain
| | - Joachim Goedhart
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
- Leeuwenhoek Centre for Advanced Microscopy (LCAM), section Molecular Cytology at Swammerdam Institute for Life Sciences (SILS) at University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
44
|
Martín-Cámara O, Cores Á, López-Alvarado P, Menéndez JC. Emerging targets in drug discovery against neurodegenerative diseases: Control of synapsis disfunction by the RhoA/ROCK pathway. Eur J Med Chem 2021; 225:113742. [PMID: 34388381 DOI: 10.1016/j.ejmech.2021.113742] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 01/11/2023]
Abstract
Synaptic spine morphology is controlled by the activity of Rac1, Cdc42 and RhoA, which need to be finely balanced, and in particular RhoA/ROCK prevents the formation of new protrusions by stabilizing actin formation. These processes are crucial to the maturation process, slowing the de novo generation of new spines. The RhoA/ROCK also influences plasticity processes, and selective modulation by ROCK1 of MLC-dependent actin dynamics leads to neurite retraction, but not to spine retraction. ROCK1 is also responsible for the reduction of the readily releasable pool of synaptic vesicles. These and other evidences suggest that ROCK1 is the main isoform acting on the presynaptic neuron. On the other hand, ROCK2 seems to have broad effects on LIMK/cofilin-dependent plasticity processes such as cofilin-dependent PSD changes. The RhoA/ROCK pathway is an important factor in several different brain-related pathologies via both downstream and upstream pathways. In the aggregate, these evidences show that the RhoA/ROCK pathway has a central role in the etiopathogenesis of a large group of CNS diseases, which underscores the importance of the pharmacological modulation of RhoA/ROCK as an important pathway to drug discovery in the neurodegenerative disease area. This article aims at providing the first review of the role of compounds acting on the RhoA/ROCK pathway in the control of synaptic disfunction.
Collapse
Affiliation(s)
- Olmo Martín-Cámara
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas. Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas. Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Pilar López-Alvarado
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas. Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - J Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas. Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain.
| |
Collapse
|
45
|
Keller-Pinter A, Gyulai-Nagy S, Becsky D, Dux L, Rovo L. Syndecan-4 in Tumor Cell Motility. Cancers (Basel) 2021; 13:cancers13133322. [PMID: 34282767 PMCID: PMC8268284 DOI: 10.3390/cancers13133322] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cell migration is crucial fReaor metastasis formation and a hallmark of malignancy. The primary cause of high mortality among oncology patients is the ability of cancer cells to metastasize. To form metastasis, primary tumor cells must be intrinsically able to move. The transmembrane, heparan sulfate proteoglycan syndecan-4 (SDC4) exhibits multiple functions in signal transduction by regulating Rac1 GTPase activity and consequently actin remodeling, as well as regulating focal adhesion kinase, protein kinase C-alpha and the level of intracellular calcium. By affecting several signaling pathways and biological processes, SDC4 is involved in cell migration under physiological and pathological conditions as well. In this review, we discuss the SDC4-mediated cell migration focusing on the role of SDC4 in tumor cell movement. Abstract Syndecan-4 (SDC4) is a ubiquitously expressed, transmembrane proteoglycan bearing heparan sulfate chains. SDC4 is involved in numerous inside-out and outside-in signaling processes, such as binding and sequestration of growth factors and extracellular matrix components, regulation of the activity of the small GTPase Rac1, protein kinase C-alpha, the level of intracellular calcium, or the phosphorylation of focal adhesion kinase. The ability of this proteoglycan to link the extracellular matrix and actin cytoskeleton enables SDC4 to contribute to biological functions like cell adhesion and migration, cell proliferation, cytokinesis, cellular polarity, or mechanotransduction. The multiple roles of SDC4 in tumor pathogenesis and progression has already been demonstrated; therefore, the expression and signaling of SDC4 was investigated in several tumor types. SDC4 influences tumor progression by regulating cell proliferation as well as cell migration by affecting cell-matrix adhesion and several signaling pathways. Here, we summarize the general role of SDC4 in cell migration and tumor cell motility.
Collapse
Affiliation(s)
- Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
- Correspondence:
| | - Szuzina Gyulai-Nagy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Daniel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Laszlo Rovo
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, H-6725 Szeged, Hungary;
| |
Collapse
|
46
|
Sahu R, Mishra R, Kumar R, Salahuddin, Majee C, Mazumder A, Kumar A. Pyridine moiety: An insight into recent advances in treatment of cancer. Mini Rev Med Chem 2021; 22:248-272. [PMID: 34126914 DOI: 10.2174/1389557521666210614162031] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/01/2021] [Accepted: 04/15/2021] [Indexed: 11/22/2022]
Abstract
The incidence of cancer is increasing worldwide, affecting a vast majority of the human population. As new different anticancer agents are being developed now, the requirement is to deal somehow with them and evaluate their safety. Among them, pyridine based drugs are contributing a lot, as it is one of the imperative pharmacophores occurring synthetically as well as naturally in heterocyclic compounds, and having a wide range of therapeutic applications in the area of drug discovery, thereby offering many chances for further improvement in antitumor agents via acting onto numerous receptors of extreme prominence. Many pyridine derivatives have been reported to inhibit enzymes, receptors and many other targets for controlling and curing the global health issue of cancer. Nowadays, in combination with other moieties, researchers are focusing on the development of pyridine-based new derivatives for cancer treatment. Therefore, this review sheds light on the recent therapeutic expansions of pyridine together with its molecular docking, structure-activity-relationship, availability in the market, and a summary of recently patented and published research works that shall jointly help the scientists to produce effective drugs with the desired pharmacological activity.
Collapse
Affiliation(s)
- Rakesh Sahu
- Department of Pharmacy, School of Medical & Allied Sciences, Galgotias University, Greater Noida-201310, India
| | - Rakhi Mishra
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Greater Noida-201306, India
| | - Rajnish Kumar
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Greater Noida-201306, India
| | - Salahuddin
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Greater Noida-201306, India
| | - Chandana Majee
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Greater Noida-201306, India
| | - Avijit Mazumder
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Greater Noida-201306, India
| | - Ajay Kumar
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Greater Noida-201306, India
| |
Collapse
|
47
|
Parallel Rap1>RalGEF>Ral and Ras signals sculpt the C. elegans nervous system. Dev Biol 2021; 477:37-48. [PMID: 33991533 DOI: 10.1016/j.ydbio.2021.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 11/23/2022]
Abstract
Ras is the most commonly mutated oncogene in humans and uses three oncogenic effectors: Raf, PI3K, and RalGEF activation of Ral. Understanding the importance of RalGEF>Ral signaling in cancer is hampered by the paucity of knowledge about their function in animal development, particularly in cell movements. We found that mutations that disrupt function of RalGEF or Ral enhance migration phenotypes of mutants for genes with established roles in cell migration. We used as a model the migration of the canal associated neurons (CANs), and validated our results in HSN cell migration, neurite guidance, and general animal locomotion. These functions of RalGEF and Ral are specific to their control of Ral signaling output rather than other published functions of these proteins. In this capacity Ral functions cell autonomously as a permissive developmental signal. In contrast, we observed Ras, the canonical activator of RalGEF>Ral signaling in cancer, to function as an instructive signal. Furthermore, we unexpectedly identified a function for the close Ras relative, Rap1, consistent with activation of RalGEF>Ral. These studies define functions of RalGEF>Ral, Rap1 and Ras signaling in morphogenetic processes that fashion the nervous system. We have also defined a model for studying how small GTPases partner with downstream effectors. Taken together, this analysis defines novel molecules and relationships in signaling networks that control cell movements during development of the nervous system.
Collapse
|
48
|
Wen YT, Huang CW, Liu CP, Chen CH, Tu CM, Hwang CS, Chen YH, Chen WR, Lin KL, Ho YC, Chen TC, Tsai RK. Inhibition of Retinal Ganglion Cell Loss By a Novel ROCK Inhibitor (E212) in Ischemic Optic Nerve Injury Via Antioxidative and Anti-Inflammatory Actions. Invest Ophthalmol Vis Sci 2021; 62:21. [PMID: 34015079 PMCID: PMC8142697 DOI: 10.1167/iovs.62.6.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/24/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose This study investigated the neuroprotective effects of administration of ROCK inhibitor E212 on ischemic optic neuropathy. Methods Rats received an intravitreal injection of either E212 or PBS immediately after optic nerve infarct. The oxidative stress in the retina was detected by performing superoxide dismutase activity and CellROX assays. The integrity of retinal pigment epithelium was determined by staining of zona occludens 1. The visual function, retinal ganglion cell (RGC) density, and RGC apoptosis were determined by using flash visual-evoked potential analysis, retrograde FluoroGold labeling, and TdT-dUTP nick end-labeling assay. Macrophage infiltration was detected by staining for ED1. The protein levels of TNF-α, p-CRMP, p-AKT1, p-STAT3, and CD206 were evaluated using Western blotting. Results Administration of E212 resulted in a 1.23-fold increase in the superoxide dismutase activity of the retina and 2.28-fold decrease in RGC-produced reactive oxygen species as compared to the levels observed upon treatment with PBS (P < 0.05). Moreover, E212 prevented the disruption of the blood-retinal barrier (BRB) in contrast to PBS. The P1-N2 amplitude and RGC density in the E212-treated group were 1.75- and 2.05-fold higher, respectively, than those in the PBS-treated group (P < 0.05). The numbers of apoptotic RGCs and macrophages were reduced by 2.93- and 2.54-fold, respectively, in the E212-treated group compared with those in the PBS-treated group (P < 0.05). The levels of p-AKT1, p-STAT3, and CD206 were increased, whereas those of p-PTEN, p-CRMP2, and TNF-α were decreased after treatment with E212 (P < 0.05). Conclusions Treatment with E212 suppresses oxidative stress, BRB disruption, and neuroinflammation to protect the visual function in ischemic optic neuropathy.
Collapse
Affiliation(s)
- Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ching-Wen Huang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Peng Liu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chih-Hung Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chia-Mu Tu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chrong-Shiong Hwang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Yi-Hsun Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wan-Ru Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Keh-Liang Lin
- Department of Medical laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chieh Ho
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Ta-Ching Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien, Taiwan
| |
Collapse
|
49
|
Ostrowska-Podhorodecka Z, Ding I, Lee W, Tanic J, Abbasi S, Arora PD, Liu RS, Patteson AE, Janmey PA, McCulloch CA. Vimentin tunes cell migration on collagen by controlling β1 integrin activation and clustering. J Cell Sci 2021; 134:jcs.254359. [PMID: 33558312 DOI: 10.1242/jcs.254359] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Vimentin is a structural protein that is required for mesenchymal cell migration and directly interacts with actin, β1 integrin and paxillin. We examined how these interactions enable vimentin to regulate cell migration on collagen. In fibroblasts, depletion of vimentin increased talin-dependent activation of β1 integrin by more than 2-fold. Loss of vimentin was associated with reduction of β1 integrin clustering by 50% and inhibition of paxillin recruitment to focal adhesions by more than 60%, which was restored by vimentin expression. This reduction of paxillin was associated with 65% lower Cdc42 activation, a 60% reduction of cell extension formation and a greater than 35% decrease in cell migration on collagen. The activation of PAK1, a downstream effector of Cdc42, was required for vimentin phosphorylation and filament maturation. We propose that vimentin tunes cell migration through collagen by acting as an adaptor protein for focal adhesion proteins, thereby regulating β1 integrin activation, resulting in well-organized, mature integrin clusters.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Isabel Ding
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Wilson Lee
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Jelena Tanic
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Sevil Abbasi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Pamma D Arora
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Richard S Liu
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Alison E Patteson
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA.,Physics Department, Syracuse University, Syracuse, NY 13244, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104-6393, USA
| | | |
Collapse
|
50
|
Fibronectin in development and wound healing. Adv Drug Deliv Rev 2021; 170:353-368. [PMID: 32961203 DOI: 10.1016/j.addr.2020.09.005] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 09/15/2020] [Indexed: 01/15/2023]
Abstract
Fibronectin structure and composition regulate contextual cell signaling. Recent advances have been made in understanding fibronectin and its role in tissue organization and repair. This review outlines fibronectin splice variants and their functions, evaluates potential therapeutic strategies targeting or utilizing fibronectin, and concludes by discussing potential future directions to modulate fibronectin function in development and wound healing.
Collapse
|