1
|
Sharma A, Steger RF, Li JM, Baude JA, Heom KA, Dey SS, Stowers RS. Sp1 mechanotransduction regulates breast cancer cell invasion in response to multiple tumor-mimicking extracellular matrix cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643983. [PMID: 40166320 PMCID: PMC11957027 DOI: 10.1101/2025.03.18.643983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Breast cancer progression is marked by extracellular matrix (ECM) remodeling, including increased stiffness, faster stress relaxation, and elevated collagen levels. In vitro experiments have revealed a role for each of these factors to individually promote malignant behavior, but their combined effects remain unclear. To address this, we developed alginate-collagen hydrogels with independently tunable stiffness, stress relaxation, and collagen density. We show that these combined tumor-mimicking ECM cues reinforced invasive morphologies and promoted spheroid invasion in breast cancer and mammary epithelial cells. High stiffness and low collagen density in slow-relaxing matrices led to the greatest cell migration speed and displacement. RNA-seq revealed Sp1 target gene enrichment in response to both individual and combined ECM cues, with a greater enrichment observed under multiple cues. Notably, high expression of Sp1 target genes upregulated by fast stress relaxation correlated with poor patient survival. Mechanistically, we found that phosphorylated-Sp1 (T453) was increasingly located in the nucleus in stiff and/or fast relaxing matrices, which was regulated by PI3K and ERK1/2 signaling, as well as actomyosin contractility. This study emphasizes how multiple ECM cues in complex microenvironments reinforce malignant traits and supports an emerging role for Sp1 as a mechanoresponsive transcription factor.
Collapse
Affiliation(s)
- Abhishek Sharma
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Rowan F Steger
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jen M Li
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jane A Baude
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Kellie A Heom
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Siddharth S Dey
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
2
|
Marshall-Burghardt S, Migueles-Ramírez RA, Lin Q, El Baba N, Saada R, Umar M, Mavalwala K, Hayer A. Excitable Rho dynamics control cell shape and motility by sequentially activating ERM proteins and actomyosin contractility. SCIENCE ADVANCES 2024; 10:eadn6858. [PMID: 39241071 PMCID: PMC11378911 DOI: 10.1126/sciadv.adn6858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/31/2024] [Indexed: 09/08/2024]
Abstract
Migration of endothelial and many other cells requires spatiotemporal regulation of protrusive and contractile cytoskeletal rearrangements that drive local cell shape changes. Unexpectedly, the small GTPase Rho, a crucial regulator of cell movement, has been reported to be active in both local cell protrusions and retractions, raising the question of how Rho activity can coordinate cell migration. Here, we show that Rho activity is absent in local protrusions and active during retractions. During retractions, Rho rapidly activated ezrin-radixin-moesin proteins (ERMs) to increase actin-membrane attachment, and, with a delay, nonmuscle myosin 2 (NM2). Rho activity was excitable, with NM2 acting as a slow negative feedback regulator. Strikingly, inhibition of SLK/LOK kinases, through which Rho activates ERMs, caused elongated cell morphologies, impaired Rho-induced cell contractions, and reverted Rho-induced blebbing. Together, our study demonstrates that Rho activity drives retractions by sequentially enhancing ERM-mediated actin-membrane attachment for force transmission and NM2-dependent contractility.
Collapse
Affiliation(s)
- Seph Marshall-Burghardt
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- Graduate Program in Biology, McGill University, Montréal, Québec, Canada
| | - Rodrigo A Migueles-Ramírez
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- PhD Program in Quantitative Life Sciences, McGill University, Montréal, Québec, Canada
| | - Qiyao Lin
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- Graduate Program in Biology, McGill University, Montréal, Québec, Canada
| | - Nada El Baba
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
- Graduate Program in Biology, McGill University, Montréal, Québec, Canada
| | - Rayan Saada
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| | - Mustakim Umar
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| | - Kian Mavalwala
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| | - Arnold Hayer
- Department of Biology, Stewart Biology Building, McGill University, Montréal, Québec H3A 1B1, Canada
| |
Collapse
|
3
|
Ran Q, Li A, Tan Y, Zhang Y, Zhang Y, Chen H. Action and therapeutic targets of myosin light chain kinase, an important cardiovascular signaling mechanism. Pharmacol Res 2024; 206:107276. [PMID: 38944220 DOI: 10.1016/j.phrs.2024.107276] [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: 05/18/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
The global incidence of cardiac diseases is increasing, imposing a substantial socioeconomic burden on healthcare systems. The pathogenesis of cardiovascular disease is complex and not fully understood, and the physiological function of the heart is inextricably linked to well-regulated cardiac muscle movement. Myosin light chain kinase (MLCK) is essential for myocardial contraction and diastole, cardiac electrophysiological homeostasis, vasoconstriction of vascular nerves and blood pressure regulation. In this sense, MLCK appears to be an attractive therapeutic target for cardiac diseases. MLCK participates in myocardial cell movement and migration through diverse pathways, including regulation of calcium homeostasis, activation of myosin light chain phosphorylation, and stimulation of vascular smooth muscle cell contraction or relaxation. Recently, phosphorylation of myosin light chains has been shown to be closely associated with the activation of myocardial exercise signaling, and MLCK mediates systolic and diastolic functions of the heart through the interaction of myosin thick filaments and actin thin filaments. It works by upholding the integrity of the cytoskeleton, modifying the conformation of the myosin head, and modulating innervation. MLCK governs vasoconstriction and diastolic function and is associated with the activation of adrenergic and sympathetic nervous systems, extracellular transport, endothelial permeability, and the regulation of nitric oxide and angiotensin II. Additionally, MLCK plays a crucial role in the process of cardiac aging. Multiple natural products/phytochemicals and chemical compounds, such as quercetin, cyclosporin, and ML-7 hydrochloride, have been shown to regulate cardiomyocyte MLCK. The MLCK-modifying capacity of these compounds should be considered in designing novel therapeutic agents. This review summarizes the mechanism of action of MLCK in the cardiovascular system and the therapeutic potential of reported chemical compounds in cardiac diseases by modifying MLCK processes.
Collapse
Affiliation(s)
- Qingzhi Ran
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China
| | - Aoshuang Li
- Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100053, China
| | - Yuqing Tan
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China
| | - Yue Zhang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China.
| | - Yongkang Zhang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| | - Hengwen Chen
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China.
| |
Collapse
|
4
|
Wilson ZS, Raya-Sandino A, Miranda J, Fan S, Brazil JC, Quiros M, Garcia-Hernandez V, Liu Q, Kim CH, Hankenson KD, Nusrat A, Parkos CA. Critical role of thrombospondin-1 in promoting intestinal mucosal wound repair. JCI Insight 2024; 9:e180608. [PMID: 39078701 PMCID: PMC11385097 DOI: 10.1172/jci.insight.180608] [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: 02/23/2024] [Accepted: 07/18/2024] [Indexed: 09/11/2024] Open
Abstract
Thrombospondin-1 (TSP1) is a matricellular protein associated with the regulation of cell migration through direct binding interactions with integrin proteins and by associating with other receptors known to regulate integrin function, including CD47 and CD36. We previously demonstrated that deletion of an epithelial TSP1 receptor, CD47, attenuates epithelial wound repair following intestinal mucosal injury. However, the mechanisms by which TSP1 contributes to intestinal mucosal repair remain poorly understood. Our results show upregulated TSP1 expression in colonic mucosal wounds and impaired intestinal mucosal wound healing in vivo upon intestinal epithelium-specific loss of TSP1 (VillinCre/+ Thbs1fl/fl or Thbs1ΔIEC mice). We report that exposure to exogenous TSP1 enhanced migration of intestinal epithelial cells in a CD47- and TGF-β1-dependent manner and that deficiency of TSP1 in primary murine colonic epithelial cells resulted in impaired wound healing. Mechanistically, TSP1 modulated epithelial actin cytoskeletal dynamics through suppression of RhoA activity, activation of Rho family small GTPase (Rac1), and changes in filamentous-actin bundling. Overall, TSP1 was found to regulate intestinal mucosal wound healing via CD47 and TGF-β1, coordinate integrin-containing cell-matrix adhesion dynamics, and remodel the actin cytoskeleton in migrating epithelial cells to enhance cell motility and promote wound repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingyang Liu
- Department of Pathology
- Mary H. Weiser Food Allergy Center, and
| | - Chang H. Kim
- Department of Pathology
- Mary H. Weiser Food Allergy Center, and
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
5
|
Xing J, Wang Y, Peng A, Li J, Niu X, Zhang K. The role of actin cytoskeleton CFL1 and ADF/cofilin superfamily in inflammatory response. Front Mol Biosci 2024; 11:1408287. [PMID: 39114368 PMCID: PMC11303188 DOI: 10.3389/fmolb.2024.1408287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024] Open
Abstract
Actin remodeling proteins are important in immune diseases and regulate cell cytoskeletal responses. These responses play a pivotal role in maintaining the delicate balance of biological events, protecting against acute or chronic inflammation in a range of diseases. Cofilin (CFL) and actin depolymerization factor (ADF) are potent actin-binding proteins that cut and depolymerize actin filaments to generate actin cytoskeleton dynamics. Although the molecular mechanism by which actin induces actin cytoskeletal reconstitution has been studied for decades, the regulation of actin in the inflammatory process has only recently become apparent. In this paper, the functions of the actin cytoskeleton and ADF/cofilin superfamily members are briefly introduced, and then focus on the role of CFL1 in inflammatory response.
Collapse
Affiliation(s)
| | | | | | | | | | - Kaiming Zhang
- ShanXi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Taiyuan Central Hospital, Dong San Dao Xiang, Taiyuan, China
| |
Collapse
|
6
|
Tai Y, Goodrich R, Maldonado M, Ortiz J, Martinez J, Ico G, Ko A, Shih HP, Nam J. Nanofiber-microwell cell culture system for spatially patterned differentiation of pluripotent stem cells in 3D. Mater Today Bio 2024; 26:101109. [PMID: 38883422 PMCID: PMC11180340 DOI: 10.1016/j.mtbio.2024.101109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/04/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024] Open
Abstract
The intricate interplay between biochemical and physical cues dictates pluripotent stem cell (PSC) differentiation to form various tissues. While biochemical modulation has been extensively studied, the role of biophysical microenvironments in early lineage commitment remains elusive. Here, we introduce a novel 3D cell culture system combining electrospun nanofibers with microfabricated polydimethylsiloxane (PDMS) patterns. This system enables the controlled formation of semispherical human induced pluripotent stem cell (hiPSC) colonies, facilitating the investigation of local mechanical stem cell niches on mechano-responsive signaling and lineage specification. Our system unveiled spatially organized RhoA activity coupled with actin-myosin cable formation, suggesting mechano-dependent hiPSC behaviors. Nodal network analysis of RNA-seq data revealed RhoA downstream regulation of YAP signaling, DNA histone modifications, and patterned germ layer specification. Notably, altering colony morphology through controlled PDMS microwell shaping effectively modulated the spatial distribution of mechano-sensitive mediators and subsequent differentiation. This study provides a cell culture platform to decipher the role of biophysical cues in early embryogenesis, offering valuable insights for material design in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Youyi Tai
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Robyn Goodrich
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Maricela Maldonado
- Department of Biomedical Engineering, California State University Long Beach, CA, 90840, USA
| | - Jessica Ortiz
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Jeniree Martinez
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Gerardo Ico
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Angel Ko
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Hung Ping Shih
- Department of Translational Research and Cellular Therapeutics, City of Hope, CA, 91010, USA
| | - Jin Nam
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| |
Collapse
|
7
|
Wang J, Zhang B, Chen X, Xin Y, Li K, Zhang C, Tang K, Tan Y. Cell mechanics regulate the migration and invasion of hepatocellular carcinoma cells via JNK signaling. Acta Biomater 2024; 176:321-333. [PMID: 38272199 DOI: 10.1016/j.actbio.2024.01.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Hepatocellular carcinoma (HCC) cells, especially those with metastatic competence, show reduced stiffness compared to the non-malignant counterparts. However, it is still unclear whether and how the mechanics of HCC cells influence their migration and invasion. This study reports that HCC cells with enhanced motility show reduced mechanical stiffness and cytoskeleton, suggesting the inverse correlation between cellular stiffness and motility. Through pharmacologic and genetic approaches, inhibiting actomyosin activity reduces HCC cellular stiffness but promotes their migration and invasion, while activating it increases cell stiffness but impairs cell motility. Actomyosin regulates cell motility through the influence on cellular stiffness. Mechanistically, weakening/strengthening cells inhibits/promotes c-Jun N terminal kinase (JNK) phosphorylation, activation/inhibition of which rescues the effects of cell mechanics on their migration and invasion. Further, HCC cancer stem cells (CSCs) exhibit higher motility but lower stiffness than control cells. Increasing CSC stiffness weakens migration and invasion through the activation of JNK signaling. In conclusion, our findings unveil a new regulatory role of actomyosin-mediated cellular mechanics in tumor cell motility and present new evidence to support that tumor cell softening may be one driving force for HCC metastasis. STATEMENT OF SIGNIFICANCE: Tumor cells progressively become softened during metastasis and low cell stiffness is associated with high metastatic potential. However, it remains unclear whether tumor cell softening is a by-product of or a driving force for tumor progression. This work reports that the stiffness of hepatocellular carcinoma cells is linked to their migration and invasion. Importantly, tumor cell softening promotes migration and invasion, while cell stiffening impairs the mobility. Weakening/strengthening cells inhibits/promotes JNK phosphorylation, activation/inhibition of which rescues the effects of cell mechanics on their migration and invasion ability. Further, stiffening liver cancer stem cells attenuates their motility through activating JNK signaling. In summary, our study uncovers a previously unappreciated role of tumor cell mechanics in migration and invasion and implicates the therapeutic potential of cell mechanics in the mechanotargeting of metastasis.
Collapse
Affiliation(s)
- Junfan Wang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Bai Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Cunyu Zhang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Kai Tang
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| |
Collapse
|
8
|
Choo YY, Sakai T, Ikebe R, Jeffers A, Idell S, Tucker TA, Ikebe M. Role of ZIP kinase in development of myofibroblast differentiation from HPMCs. Am J Physiol Lung Cell Mol Physiol 2024; 326:L353-L366. [PMID: 38252666 PMCID: PMC11281797 DOI: 10.1152/ajplung.00251.2023] [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: 08/07/2023] [Revised: 12/14/2023] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
During the development of pleural fibrosis, pleural mesothelial cells (PMCs) undergo phenotypic switching from differentiated mesothelial cells to mesenchymal cells (MesoMT). Here, we investigated how external stimuli such as TGF-β induce HPMC-derived myofibroblast differentiation to facilitate the development of pleural fibrosis. TGF-β significantly increased di-phosphorylation but not mono-phosphorylation of myosin II regulatory light chain (RLC) in HPMCs. An increase in RLC di-phosphorylation was also found at the pleural layer of our carbon black bleomycin (CBB) pleural fibrosis mouse model, where it showed filamentous localization that coincided with alpha smooth muscle actin (αSMA) in the cells in the pleura. Among the protein kinases that can phosphorylate myosin II RLC, ZIPK (zipper-interacting kinase) protein expression was significantly augmented after TGF-β stimulation. Furthermore, ZIPK gene silencing attenuated RLC di-phosphorylation, suggesting that ZIPK is responsible for di-phosphorylation of myosin II in HPMCs. Although TGF-β significantly increased the expression of ZIP kinase protein, the change in ZIP kinase mRNA was marginal, suggesting a posttranscriptional mechanism for the regulation of ZIP kinase expression by TGF-β. ZIPK gene knockdown (KD) also significantly reduced TGF-β-induced upregulation of αSMA expression. This finding suggests that siZIPK attenuates myofibroblast differentiation of HPMCs. siZIPK diminished TGF-β-induced contractility of HPMCs consistent with siZIPK-induced decrease in the di-phosphorylation of myosin II RLC. The present results implicate ZIPK in the regulation of the contractility of HPMC-derived myofibroblasts, phenotype switching, and myofibroblast differentiation of HPMCs.NEW & NOTEWORTHY Here, we highlight that ZIP kinase is responsible for di-phosphorylation of myosin light chain, which facilitates stress fiber formation and actomyosin-based cell contraction during mesothelial to mesenchymal transition in human pleural mesothelial cells. This transition has a significant impact on tissue remodeling and subsequent stiffness of the pleura. This study provides insight into a new therapeutic strategy for the treatment of pleural fibrosis.
Collapse
Affiliation(s)
- Young-Yeon Choo
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Ann Jeffers
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, Texas, United States
| |
Collapse
|
9
|
Borjini N, Lun Y, Jang GF, Crabb J, Chen Y, Crabb J, Fox DA, Ivanov AI, Lin F. CD6 triggers actomyosin cytoskeleton remodeling after binding to its receptor complex. J Leukoc Biol 2024; 115:450-462. [PMID: 37820034 PMCID: PMC10890838 DOI: 10.1093/jleuko/qiad124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023] Open
Abstract
The T cell marker CD6 regulates both T cells and target cells during inflammatory responses by interacting with its receptors. However, only a few receptors binding to the extracellular domains of CD6 have been identified, and cellular events induced by CD6 engagement with its receptors in target cells remain poorly understood. In this study, we identified CD44 as a novel CD6 receptor by proximity labeling and confirmed the new CD6-CD44 interaction by biochemical and biophysical approaches. CD44 and the other 2 known CD6 receptors, CD166 and CDCP1, were distributed diffusely on resting retinal pigment epithelium (RPE) cells but clustered together to form a receptor complex upon CD6 binding. CD6 stimulation induced dramatic remodeling of the actomyosin cytoskeleton in RPE cells mediated by activation of RhoA, and Rho-associated kinase signaling, resulting in increased myosin II phosphorylation. Such actomyosin activation triggered the disassembly of tight junctions responsible for RPE barrier integrity in a process that required all components of the tripartite CD6 receptor complex. These data provided new insights into the mechanisms by which CD6 mediates T cell-driven disruption of tissue barriers during inflammation.
Collapse
Affiliation(s)
- Nozha Borjini
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, United States
| | - Yu Lun
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, United States
| | - Geen-Fu Jang
- Cole Eye Institute, Cleveland Clinic, 2042 E 102nd St, Cleveland, OH 44106, United States
| | - Jack Crabb
- Cole Eye Institute, Cleveland Clinic, 2042 E 102nd St, Cleveland, OH 44106, United States
| | - Yinghua Chen
- Department of Physiology and Biophysics, Case Western Reserve University, 2210 Circle Dr Robbins Building, Cleveland, OH 44106, United States
| | - John Crabb
- Cole Eye Institute, Cleveland Clinic, 2042 E 102nd St, Cleveland, OH 44106, United States
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109, United States
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, United States
| | - Feng Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, United States
- Cole Eye Institute, Cleveland Clinic, 2042 E 102nd St, Cleveland, OH 44106, United States
| |
Collapse
|
10
|
Wang Y, Ye Q, Cui Y, Wu Y, Cao S, Hu F. Impact and mechanisms of drag-reducing polymers on shear stress regulation in pulmonary hypertension. Clin Hemorheol Microcirc 2024; 88:247-261. [PMID: 38905038 DOI: 10.3233/ch-242281] [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: 06/23/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a refractory disease characterized by elevated pulmonary artery pressure and resistance. Drag-reducing polymers (DRPs) are blood-soluble macromolecules that reduce vascular resistance by altering the blood dynamics and rheology. Our previous work indicated that polyethylene oxide (PEO) can significantly reduce the medial wall thickness and vascular resistance of the pulmonary arteries, but the specific mechanism is still unclear. METHODS This study was designed to investigate the role and mechanism of PEO on intracellular calcium [Ca2 +] i and cytoskeletal proteins of endothelial cells (ECs) induced by low shear stress (LSS) in PH. Primary Pulmonary Artery Endothelial Cells (PAECs) were subjected to steady LSS (1 dyn/cm2) or physiological shear stress (SS) (10 dyn/cm2) for 20 h in a BioFlux 200 flow system. Calcium influx assays were conducted to evaluate the mechanisms of PEO on [Ca2 +] i. Subsequently, taking the key protein that induces cytoskeletal remodeling, the regulatory light chain (RLC) phosphorylation, as the breakthrough point, this study focused on the two key pathways of PEO that regulate phosphorylation of RLC: Myosin light chain kinase (MLCK) and Rho-associated kinase (ROCK) pathways. RESULTS Our current research revealed that PEO at LSS (1 dyn/cm2) significantly suppressed LSS-induced [Ca2 +] i and the expression level of transient receptor potential channel 1(TRPC1). In addition, ECs convert LSS stimuli into the upregulation of cytoskeletal proteins, including filamentous actin (F-actin), MLCK, ROCK, p-RLC, and pp-RLC. Further experiments using pharmacological inhibitors demonstrated that PEO at the LSS downregulated cytoskeleton-related proteins mainly through the ROCK and MLCK pathways. CONCLUSIONS This study considered intracellular calcium and cytoskeleton rearrangement as entry points to study the application of PEO in the biomedical field, which has important theoretical significance and practical application value for the treatment of PH.
Collapse
Affiliation(s)
- Yali Wang
- Department of Respiratory Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Qing Ye
- Department of Respiratory Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yongqi Cui
- Department of Respiratory Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yunjiang Wu
- Department of Thoracic Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Sipei Cao
- Department of Respiratory Medicine, The Third People's Hospital of Hefei, Hefei, China
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Feng Hu
- Department of Cardiology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
11
|
Cho Y, Kim J, Park J, Doh J. Surface nanotopography and cell shape modulate tumor cell susceptibility to NK cell cytotoxicity. MATERIALS HORIZONS 2023; 10:4532-4540. [PMID: 37559559 DOI: 10.1039/d3mh00367a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
Natural killer (NK) cells are innate cytotoxic lymphocytes exerting cytotoxicity against virally infected cells and tumor cells. NK cell cytotoxicity is primarily determined by biochemical signals received from ligands expressed on target cell surfaces, but it is also possible that biophysical environments of tumor cells, such as nanoscale surface topography typically existing on extracellular matrixes (ECMs) or cell morphology determined by ECM spaces or cell density, regulate NK cell cytotoxicity. In this study, micro/nanofabrication technology was applied to examine this possibility. Tumor cells were plated on flat or nanogrooved surfaces, or micropatterned into circular or elliptical geometries, and the effects of surface topography and tumor cell morphology on NK cell cytotoxicity were investigated. NK cells exhibited significantly higher cytotoxicity against tumor cells on nanogrooved surfaces or tumor cells in elliptical patterns than tumor cells on flat surfaces or tumor cells in circular patterns, respectively. The amounts of stress fiber formation in tumor cells positively correlated with NK cell cytotoxicity, indicating that increased cellular tension of tumor cells, either mediated by nanogrooved surfaces or elongated morphologies, was a key factor regulating NK cell cytotoxicity. These results may provide insight into the design of NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Yongbum Cho
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology, 77, Cheongam-ro, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - JangHyuk Kim
- Department of Materials Science and Engineering, Seoul National University, Seoul, South Korea.
| | - Jeehun Park
- SOFT Foundry Institute, Seoul National University, Seoul, South Korea.
| | - Junsang Doh
- Department of Materials Science and Engineering, Seoul National University, Seoul, South Korea.
- SOFT Foundry Institute, Seoul National University, Seoul, South Korea.
- Institute of Engineering Research, BioMAX, Seoul National University, Seoul, South Korea
| |
Collapse
|
12
|
Wang Y, Troughton LD, Xu F, Chatterjee A, Ding C, Zhao H, Cifuentes LP, Wagner RB, Wang T, Tan S, Chen J, Li L, Umulis D, Kuang S, Suter DM, Yuan C, Chan D, Huang F, Oakes PW, Deng Q. Atypical peripheral actin band formation via overactivation of RhoA and nonmuscle myosin II in mitofusin 2-deficient cells. eLife 2023; 12:e88828. [PMID: 37724949 PMCID: PMC10550287 DOI: 10.7554/elife.88828] [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: 04/21/2023] [Accepted: 09/19/2023] [Indexed: 09/21/2023] Open
Abstract
Cell spreading and migration play central roles in many physiological and pathophysiological processes. We have previously shown that MFN2 regulates the migration of human neutrophil-like cells via suppressing Rac activation. Here, we show that in mouse embryonic fibroblasts, MFN2 suppresses RhoA activation and supports cell polarization. After initial spreading, the wild-type cells polarize and migrate, whereas the Mfn2-/- cells maintain a circular shape. Increased cytosolic Ca2+ resulting from the loss of Mfn2 is directly responsible for this phenotype, which can be rescued by expressing an artificial tether to bring mitochondria and endoplasmic reticulum to close vicinity. Elevated cytosolic Ca2+ activates Ca2+/calmodulin-dependent protein kinase II, RhoA, and myosin light-chain kinase, causing an overactivation of nonmuscle myosin II, leading to a formation of a prominent F-actin ring at the cell periphery and increased cell contractility. The peripheral actin band alters cell physics and is dependent on substrate rigidity. Our results provide a novel molecular basis to understand how MFN2 regulates distinct signaling pathways in different cells and tissue environments, which is instrumental in understanding and treating MFN2-related diseases.
Collapse
Affiliation(s)
- Yueyang Wang
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Lee D Troughton
- Cell and Molecular Physiology, Loyola University ChicagoChicagoUnited States
| | - Fan Xu
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of TechnologyBeijingChina
| | - Aritra Chatterjee
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Chang Ding
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Han Zhao
- Davidson School of Chemical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Laura P Cifuentes
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Ryan B Wagner
- School of Mechanical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Tianqi Wang
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Shelly Tan
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Jingjuan Chen
- Department of Animal Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Linlin Li
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - David Umulis
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
- Department of Agricultural and Biological Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University West LafayetteWest LafayetteUnited States
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
- Purdue Institute for Integrative Neuroscience, Purdue University West LafayetteWest LafayetteUnited States
- Purdue Institute for Inflammation, Immunology & Infectious Disease, Purdue University West LafayetteWest LafayetteUnited States
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Deva Chan
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Fang Huang
- Weldon School of Biomedical Engineering, Purdue University West LafayetteWest LafayetteUnited States
| | - Patrick W Oakes
- Cell and Molecular Physiology, Loyola University ChicagoChicagoUnited States
| | - Qing Deng
- Department of Biological Sciences, Purdue University West LafayetteWest LafayetteUnited States
- Purdue Institute for Inflammation, Immunology & Infectious Disease, Purdue University West LafayetteWest LafayetteUnited States
- Purdue University Center for Cancer Research, Purdue University West LafayetteWest LafayetteUnited States
| |
Collapse
|
13
|
Jin Q, Pandey D, Thompson CB, Lewis S, Sung HW, Nguyen TD, Kuo S, Wilson KL, Gracias DH, Romer LH. Acute downregulation of emerin alters actomyosin cytoskeleton connectivity and function. Biophys J 2023; 122:3690-3703. [PMID: 37254483 PMCID: PMC10541481 DOI: 10.1016/j.bpj.2023.05.027] [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: 01/30/2023] [Revised: 04/30/2023] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
Fetal lung fibroblasts contribute dynamic infrastructure for the developing lung. These cells undergo dynamic mechanical transitions, including cyclic stretch and spreading, which are integral to lung growth in utero. We investigated the role of the nuclear envelope protein emerin in cellular responses to these dynamic mechanical transitions. In contrast to control cells, which briskly realigned their nuclei, actin cytoskeleton, and extracellular matrices in response to cyclic stretch, fibroblasts that were acutely downregulated for emerin showed incomplete reorientation of both nuclei and actin cytoskeleton. Emerin-downregulated fibroblasts were also aberrantly circular in contrast to the spindle-shaped controls and exhibited an altered pattern of filamentous actin organization that was disconnected from the nucleus. Emerin knockdown was also associated with reduced myosin light chain phosphorylation during cell spreading. Interestingly, emerin-downregulated fibroblasts also demonstrated reduced fibronectin fibrillogenesis and production. These findings indicate that nuclear-cytoskeletal coupling serves a role in the dynamic regulation of cytoskeletal structure and function and may also impact the transmission of traction force to the extracellular matrix microenvironment.
Collapse
Affiliation(s)
- Qianru Jin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Deepesh Pandey
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Carol B Thompson
- Biostatistics Center, Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Shawna Lewis
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Hyun Woo Sung
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Thao D Nguyen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Scot Kuo
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland; Microscope Facility, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Katherine L Wilson
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - David H Gracias
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland; Center for MicroPhysiological Systems, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Chemistry, Johns Hopkins University, Baltimore, Maryland; Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland
| | - Lewis H Romer
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland; Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
14
|
Baoyinna B, Miao J, Oliver PJ, Ye Q, Shaheen N, Kalin T, He J, Parinandi NL, Zhao Y, Zhao J. Non-Lethal Doses of RSL3 Impair Microvascular Endothelial Barrier through Degradation of Sphingosie-1-Phosphate Receptor 1 and Cytoskeletal Arrangement in A Ferroptosis-Independent Manner. Biomedicines 2023; 11:2451. [PMID: 37760892 PMCID: PMC10525432 DOI: 10.3390/biomedicines11092451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
The excess microvascular endothelial permeability is a hallmark of acute inflammatory diseases. Maintenance of microvascular integrity is critical to preventing leakage of vascular components into the surrounding tissues. Sphingosine-1-phosphate (S1P) is an active lysophospholipid that enhances the endothelial cell (EC) barrier via activation of its receptor S1PR1. Here, we delineate the effect of non-lethal doses of RSL3, an inhibitor of glutathione peroxidase 4 (GPX4), on EC barrier function. Low doses of RSL3 (50-100 nM) attenuated S1P-induced human lung microvascular barrier enhancement and the phosphorylation of AKT. To investigate the molecular mechanisms by which RSL3 attenuates S1P's effect, we examined the S1PR1 levels. RSL3 treatment reduced S1PR1 levels in 1 h, whereas the effect was attenuated by the proteasome and lysosome inhibitors as well as a lipid raft inhibitor. Immunofluorescence staining showed that RSL3 induced S1PR1 internalization from the plasma membrane into the cytoplasm. Furthermore, we found that RSL3 (100 and 200 nM) increased EC barrier permeability and cytoskeletal rearrangement without altering cell viability. Taken together, our data delineates that non-lethal doses of RSL3 impair EC barrier function via two mechanisms. RSL3 attenuates S1P1-induced EC barrier enhancement and disrupts EC barrier integrity through the generation of 4-hydroxynonena (4HNE). All these effects are independent of ferroptosis.
Collapse
Affiliation(s)
- Boina Baoyinna
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jiaxing Miao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Patrick J. Oliver
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Qinmao Ye
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Timothy Kalin
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jinshan He
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Shi X, Li H, Dan Z, Shu C, Zhu R, Yang Q, Wang Y, Zhu H. Melatonin Potentiates Sensitivity to 5-Fluorouracil in Gastric Cancer Cells by Upregulating Autophagy and Downregulating Myosin Light-Chain Kinase. J Cancer 2023; 14:2608-2618. [PMID: 37779875 PMCID: PMC10539390 DOI: 10.7150/jca.85353] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/23/2023] [Indexed: 10/03/2023] Open
Abstract
5-Fluorouracil is an effective chemotherapeutic drug for gastric cancer. However, the acquisition of chemotherapeutic resistance remains a challenge in treatment. Melatonin can enhance the therapeutic effect of 5-fluorouracil; however, the underlying mechanisms are not well understood. We investigated the effects of combinations of melatonin and 5-fluorouracil on the proliferation, migration and invasion of gastric cancer cells. Melatonin significantly potentiated the 5-fluorouracil-mediated inhibition of proliferation, migration and invasion in gastric cancer cells, which potentiates sensitivity to 5-FU by promoting the activation of Beclin-1-dependent autophagy and targeting the myosin light-chain kinase (MLCK) signaling pathway. Previous studies have shown that autophagy might be associated with the MLCK signaling pathway. The autophagy inhibitor, 3-methyladenine, effectively rescued the migratory and invasive capabilities of gastric cancer cells, while also reducing expression level of MLCK and the phosphorylation level of MLC. This indicates that autophagy is involved in tumor metastasis, which may be related to inhibition of the MLCK signaling pathway. Our findings indicate that melatonin can improve the effectiveness of 5-fluorouracil in gastric cancer and could be used as a supplemental agent in the treatment of gastric cancer with 5-fluorouracil.
Collapse
Affiliation(s)
- Xiaorui Shi
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Hongxia Li
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhangyong Dan
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Chuanlin Shu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Rumeng Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| | - Qingling Yang
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China
| | - Yi Wang
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
- Department of Biological Engineering, School of Life Sciences, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui 233030, China
| | - Huaqing Zhu
- Laboratory of Molecular Biology, Department of Biochemistry, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
16
|
Ruperti F, Becher I, Stokkermans A, Wang L, Marschlich N, Potel C, Maus E, Stein F, Drotleff B, Schippers K, Nickel M, Prevedel R, Musser JM, Savitski MM, Arendt D. Molecular profiling of sponge deflation reveals an ancient relaxant-inflammatory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551666. [PMID: 37577507 PMCID: PMC10418225 DOI: 10.1101/2023.08.02.551666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
A hallmark of animals is the coordination of whole-body movement. Neurons and muscles are central to this, yet coordinated movements also exist in sponges that lack these cell types. Sponges are sessile animals with a complex canal system for filter-feeding. They undergo whole-body movements resembling "contractions" that lead to canal closure and water expulsion. Here, we combine 3D optical coherence microscopy, pharmacology, and functional proteomics to elucidate anatomy, molecular physiology, and control of these movements. We find them driven by the relaxation of actomyosin stress fibers in epithelial canal cells, which leads to whole-body deflation via collapse of the incurrent and expansion of the excurrent system, controlled by an Akt/NO/PKG/A pathway. A concomitant increase in reactive oxygen species and secretion of proteinases and cytokines indicate an inflammation-like state reminiscent of vascular endothelial cells experiencing oscillatory shear stress. This suggests an ancient relaxant-inflammatory response of perturbed fluid-carrying systems in animals.
Collapse
Affiliation(s)
- Fabian Ruperti
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Collaboration for joint Ph.D. degree between EMBL and Heidelberg University, Faculty of Biosciences 69117 Heidelberg, Germany
| | - Isabelle Becher
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | | | - Ling Wang
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Nick Marschlich
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| | - Clement Potel
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Emanuel Maus
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Bernhard Drotleff
- Metabolomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Klaske Schippers
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Michael Nickel
- Bionic Consulting Dr. Michael Nickel, 71686 Remseck am Neckar, Germany
| | - Robert Prevedel
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Jacob M Musser
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Mikhail M Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Detlev Arendt
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
17
|
Fan JR, Chang SN, Chu CT, Chen HC. AKT2-mediated nuclear deformation leads to genome instability during epithelial-mesenchymal transition. iScience 2023; 26:106992. [PMID: 37378334 PMCID: PMC10291577 DOI: 10.1016/j.isci.2023.106992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/04/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Nuclear deformation has been observed in some cancer cells for decades, but its underlying mechanism and biological significance remain elusive. To address these questions, we employed human lung cancer A549 cell line as a model in context with transforming growth factor β (TGFβ)-induced epithelial-mesenchymal transition. Here, we report that nuclear deformation induced by TGFβ is concomitant with increased phosphorylation of lamin A at Ser390, defective nuclear lamina and genome instability. AKT2 and Smad3 serve as the downstream effectors for TGFβ to induce nuclear deformation. AKT2 directly phosphorylates lamin A at Ser390, whereas Smad3 is required for AKT2 activation upon TGFβ stimulation. Expression of the lamin A mutant with a substitution of Ser390 to Ala or suppression of AKT2 or Smad3 prevents nuclear deformation and genome instability induced by TGFβ. These findings reveal a molecular mechanism for TGFβ-induced nuclear deformation and establish a role of nuclear deformation in genome instability during epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Jia-Rong Fan
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Sung-Nian Chang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ching-Tung Chu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Hong-Chen Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
18
|
Serwe G, Kachaner D, Gagnon J, Plutoni C, Lajoie D, Duramé E, Sahmi M, Garrido D, Lefrançois M, Arseneault G, Saba-El-Leil MK, Meloche S, Emery G, Therrien M. CNK2 promotes cancer cell motility by mediating ARF6 activation downstream of AXL signalling. Nat Commun 2023; 14:3560. [PMID: 37322019 PMCID: PMC10272126 DOI: 10.1038/s41467-023-39281-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
Cell motility is a critical feature of invasive tumour cells that is governed by complex signal transduction events. Particularly, the underlying mechanisms that bridge extracellular stimuli to the molecular machinery driving motility remain partially understood. Here, we show that the scaffold protein CNK2 promotes cancer cell migration by coupling the pro-metastatic receptor tyrosine kinase AXL to downstream activation of ARF6 GTPase. Mechanistically, AXL signalling induces PI3K-dependent recruitment of CNK2 to the plasma membrane. In turn, CNK2 stimulates ARF6 by associating with cytohesin ARF GEFs and with a novel adaptor protein called SAMD12. ARF6-GTP then controls motile forces by coordinating the respective activation and inhibition of RAC1 and RHOA GTPases. Significantly, genetic ablation of CNK2 or SAMD12 reduces metastasis in a mouse xenograft model. Together, this work identifies CNK2 and its partner SAMD12 as key components of a novel pro-motility pathway in cancer cells, which could be targeted in metastasis.
Collapse
Affiliation(s)
- Guillaume Serwe
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - David Kachaner
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Jessica Gagnon
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Cédric Plutoni
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Driss Lajoie
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Eloïse Duramé
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Malha Sahmi
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Damien Garrido
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Martin Lefrançois
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Geneviève Arseneault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Marc K Saba-El-Leil
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Gregory Emery
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Marc Therrien
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada.
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
19
|
Kokate SB, Ciuba K, Tran VD, Kumari R, Tojkander S, Engel U, Kogan K, Kumar S, Lappalainen P. Caldesmon controls stress fiber force-balance through dynamic cross-linking of myosin II and actin-tropomyosin filaments. Nat Commun 2022; 13:6032. [PMID: 36229430 PMCID: PMC9561149 DOI: 10.1038/s41467-022-33688-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Contractile actomyosin bundles are key force-producing and mechanosensing elements in muscle and non-muscle tissues. Whereas the organization of muscle myofibrils and mechanism regulating their contractility are relatively well-established, the principles by which myosin-II activity and force-balance are regulated in non-muscle cells have remained elusive. We show that Caldesmon, an important component of smooth muscle and non-muscle cell actomyosin bundles, is an elongated protein that functions as a dynamic cross-linker between myosin-II and tropomyosin-actin filaments. Depletion of Caldesmon results in aberrant lateral movement of myosin-II filaments along actin bundles, leading to irregular myosin distribution within stress fibers. This manifests as defects in stress fiber network organization and contractility, and accompanied problems in cell morphogenesis, migration, invasion, and mechanosensing. These results identify Caldesmon as critical factor that ensures regular myosin-II spacing within non-muscle cell actomyosin bundles, and reveal how stress fiber networks are controlled through dynamic cross-linking of tropomyosin-actin and myosin filaments.
Collapse
Affiliation(s)
- Shrikant B Kokate
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Katarzyna Ciuba
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.,Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093, Warszawa, Poland
| | - Vivien D Tran
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sari Tojkander
- Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Building, E318, Arvo Ylpön katu 34, 33520, Tampere, Finland
| | - Ulrike Engel
- Nikon Imaging Center at Heidelberg University and Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 267, Heidelberg, 69120, Germany
| | - Konstantin Kogan
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland.
| |
Collapse
|
20
|
Hollósi A, Pászty K, Bunta BL, Bozó T, Kellermayer M, Debreczeni ML, Cervenak L, Baccarini M, Varga A. BRAF increases endothelial cell stiffness through reorganization of the actin cytoskeleton. FASEB J 2022; 36:e22478. [PMID: 35916021 DOI: 10.1096/fj.202200344r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/05/2022] [Accepted: 07/19/2022] [Indexed: 11/11/2022]
Abstract
The dynamics of the actin cytoskeleton and its connection to endothelial cell-cell junctions determine the barrier function of endothelial cells. The proper regulation of barrier opening/closing is necessary for the normal function of vessels, and its dysregulation can result in chronic and acute inflammation leading to edema formation. By using atomic force microscopy, we show here that thrombin-induced permeability of human umbilical vein endothelial cells, associated with actin stress fiber formation, stiffens the cell center. The depletion of the MEK/ERK kinase BRAF reduces thrombin-induced permeability prevents stress fiber formation and cell stiffening. The peripheral actin ring becomes stabilized by phosphorylated myosin light chain, while cofilin is excluded from the cell periphery. All these changes can be reverted by the inhibition of ROCK, but not of the MEK/ERK module. We propose that the balance between the binding of cofilin and myosin to F-actin in the cell periphery, which is regulated by the activity of ROCK, determines the local dynamics of actin reorganization, ultimately driving or preventing stress fiber formation.
Collapse
Affiliation(s)
- Anna Hollósi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Katalin Pászty
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Bálint Levente Bunta
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tamás Bozó
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Márta Lídia Debreczeni
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna, Austria
| | - Andrea Varga
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
21
|
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
|
22
|
Xiao X, Mao X, Chen D, Yu B, He J, Yan H, Wang J. miRNAs Can Affect Intestinal Epithelial Barrier in Inflammatory Bowel Disease. Front Immunol 2022; 13:868229. [PMID: 35493445 PMCID: PMC9043318 DOI: 10.3389/fimmu.2022.868229] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022] Open
Abstract
The most obvious pathological characterization of inflammatory bowel disease (IBD) is intestinal epithelium erosion and severe inflammation invasion. Micro-ribonucleic acids (miRNA or microRNA), single-stranded noncoding RNAs of ~22 nucleotides, have been considered as the potential therapeutic targets in the pathogenesis of IBD. Many previous studies have focused on the mechanisms that miRNAs use to regulate inflammation, immunity, and microorganisms in IBD. The review highlights in detail the findings of miRNAs in the intestinal epithelial barrier of IBD, and focuses on their gene targets, signaling pathways associated with IBD, and some potential therapies. It will be beneficial for the elucidation of the interaction between miRNAs and the intestinal epithelial barrier in IBD and provide a theoretical reference for preventing and treating IBD in the future.
Collapse
Affiliation(s)
- Xiangjun Xiao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| |
Collapse
|
23
|
Morris T, Sue E, Geniesse C, Brieher WM, Tang VW. Synaptopodin stress fiber and contractomere at the epithelial junction. J Cell Biol 2022; 221:e202011162. [PMID: 35416930 PMCID: PMC9011326 DOI: 10.1083/jcb.202011162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 10/07/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
The apical junction of epithelial cells can generate force to control cell geometry and perform contractile processes while maintaining barrier function and adhesion. Yet, the structural basis for force generation at the apical junction is not fully understood. Here, we describe two synaptopodin-dependent actomyosin structures that are spatially, temporally, and structurally distinct. The first structure is formed by the retrograde flow of synaptopodin initiated at the apical junction, creating a sarcomeric stress fiber that lies parallel to the apical junction. Contraction of the apical stress fiber is associated with either clustering of membrane components or shortening of junctional length. Upon junction maturation, apical stress fibers are disassembled. In mature epithelial monolayer, a motorized "contractomere" capable of "walking the junction" is formed at the junctional vertex. Actomyosin activities at the contractomere produce a compressive force evident by actin filament buckling and measurement with a new α-actinin-4 force sensor. The motility of contractomeres can adjust junctional length and change cell packing geometry during cell extrusion and intercellular movement. We propose a model of epithelial homeostasis that utilizes contractomere motility to support junction rearrangement while preserving the permeability barrier.
Collapse
Affiliation(s)
- Timothy Morris
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Eva Sue
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Caleb Geniesse
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Vivian W Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
24
|
Nawrot DA, Ozer LY, Al Haj Zen A. A Novel High Content Angiogenesis Assay Reveals That Lacidipine, L-Type Calcium Channel Blocker, Induces In Vitro Vascular Lumen Expansion. Int J Mol Sci 2022; 23:ijms23094891. [PMID: 35563280 PMCID: PMC9100973 DOI: 10.3390/ijms23094891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Angiogenesis is a critical cellular process toward establishing a functional circulatory system capable of delivering oxygen and nutrients to the tissue in demand. In vitro angiogenesis assays represent an important tool for elucidating the biology of blood vessel formation and for drug discovery applications. Herein, we developed a novel, high content 2D angiogenesis assay that captures endothelial morphogenesis’s cellular processes, including lumen formation. In this assay, endothelial cells form luminized vascular-like structures in 48 h. The assay was validated for its specificity and performance. Using the optimized assay, we conducted a phenotypic screen of a library containing 150 FDA-approved cardiovascular drugs to identify modulators of lumen formation. The screening resulted in several L-type calcium channel blockers being able to expand the lumen space compared to controls. Among these blockers, Lacidipine was selected for follow-up studies. We found that the endothelial cells treated with Lacidipine showed enhanced activity of caspase-3 in the luminal space. Pharmacological inhibition of caspase activity abolished the Lacidipine-enhancing effect on lumen formation, suggesting the involvement of apoptosis. Using a Ca2+ biosensor, we found that Lacipidine reduces the intracellular Ca2+ oscillations amplitude in the endothelial cells at the early stage, whereas Lacidipine blocks these Ca2+ oscillations completely at the late stage. The inhibition of MLCK exhibits a phenotype of lumen expansion similar to that of Lacidipine. In conclusion, this study describes a novel high-throughput phenotypic assay to study angiogenesis. Our findings suggest that calcium signalling plays an essential role during lumen morphogenesis. L-type Ca2+ channel blockers could be used for more efficient angiogenesis-mediated therapies.
Collapse
Affiliation(s)
- Dorota A. Nawrot
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- Alzheimer’s Research UK, Oxford Drug Discovery Institute, Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Lutfiye Yildiz Ozer
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha P.O. Box 34110, Qatar;
| | - Ayman Al Haj Zen
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha P.O. Box 34110, Qatar;
- Correspondence: ; Tel.: +974-4454-6352
| |
Collapse
|
25
|
Dobrokhotov O, Sunagawa M, Torii T, Mii S, Kawauchi K, Enomoto A, Sokabe M, Hirata H. Anti-Malignant Effect of Tensile Loading to Adherens Junctions in Cutaneous Squamous Cell Carcinoma Cells. Front Cell Dev Biol 2021; 9:728383. [PMID: 34858971 PMCID: PMC8632149 DOI: 10.3389/fcell.2021.728383] [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: 06/22/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Actomyosin contractility regulates various cellular processes including proliferation and differentiation while dysregulation of actomyosin activity contributes to cancer development and progression. Previously, we have reported that actomyosin-generated tension at adherens junctions is required for cell density-dependent inhibition of proliferation of normal skin keratinocytes. However, it remains unclear how actomyosin contractility affects the hyperproliferation ability of cutaneous squamous cell carcinoma (cSCC) cells. In this study, we find that actomyosin activity is impaired in cSCC cells both in vitro and in vivo. External application of tensile loads to adherens junctions by sustained mechanical stretch attenuates the proliferation of cSCC cells, which depends on intact adherens junctions. Forced activation of actomyosin of cSCC cells also inhibits their proliferation in a cell-cell contact-dependent manner. Furthermore, the cell cycle arrest induced by tensile loading to adherens junctions is accompanied by epidermal differentiation in cSCC cells. Our results show that the degree of malignant properties of cSCC cells can be reduced by applying tensile loads to adherens junctions, which implies that the mechanical status of adherens junctions may serve as a novel therapeutic target for cSCC.
Collapse
Affiliation(s)
- Oleg Dobrokhotov
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Sunagawa
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeru Torii
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keiko Kawauchi
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Hirata
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
26
|
Song Y, Yang Y, Zeng W, Loor JJ, Jiang Q, Peng Z, Li Y, Jiang S, Feng X, Du X, Li X, Liu G. β-Hydroxybutyrate impairs neutrophil migration distance through activation of a protein kinase C and myosin light chain 2 signaling pathway in ketotic cows. J Dairy Sci 2021; 105:761-771. [PMID: 34635355 DOI: 10.3168/jds.2021-20875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022]
Abstract
Ketosis in dairy cows often occurs in the peripartal period and is accompanied by immune dysfunction. High concentrations of β-hydroxybutyrate (BHB) in peripheral blood during ketosis are closely related to the impairment of polymorphonuclear neutrophil (PMN) chemotaxis and contribute to immune dysfunction. The specific effect of BHB on PMN chemotaxis in dairy cows and the underlying molecular mechanisms are unclear. Here, 30 multiparous cows (within 3 wk postpartum) classified based on serum BHB as control (n = 15, BHB <0.6 mM) or clinically ketotic (n = 15, BHB >3.0 mM) were used. Blood samples were collected before feeding, and the isolated PMN were treated with platelet-activating factor for 0.5 h to activate their migration. Scanning electron microscopy revealed a longer tail in PMN of ketotic cows. In addition, the phosphorylation and transcription levels of myosin light chain 2 (MLC2) increased in PMN of ketotic cows. Polymorphonuclear neutrophils from control dairy cows were incubated with 3.0 mM BHB for different times in vitro, and 6 h was selected as the proper duration of BHB stimulation according to its inhibition effect on PMN migration using an under-agarose PMN chemotaxis model. Similarly, BHB stimulation in vitro resulted in inhibition of migration distance and deviation of migration direction of PMN, as well as a longer tail in morphology in the scanning electron microscope data, suggesting that BHB-induced PMN migration inhibition may be mediated by impairing the trailing edge contraction. To confirm this hypothesis, sotrastaurin (Sotra)-a specific inhibitor of protein kinase C (PKC), which is the core regulator of cell contraction-was used with or without BHB treatment in vitro. Sotra was pretreated 0.5 h before BHB treatment. Accordingly, BHB treatment increased the phosphorylation level of PKC and MLC2, the protein abundance of RhoA and rho-kinase 1 (ROCK1), and the mRNA abundance of PRKCA, MYL2, RHOA, and ROCK1 in PMN. In contrast, these effects of BHB on PMN were dampened by Sotra. As demonstrated by immunofluorescence experiments in vitro, the BHB-induced inhibition of trailing edge contraction of PMN was relieved by Sotra. In addition, Sotra also dampened the effects of BHB on PMN migration in vitro. Furthermore, as verified by in vivo experiments, compared with the control cows, both abundance and activation of PKC signaling were enhanced in PMN of ketotic cows. Overall, the present study revealed that high concentrations of blood BHB impaired PMN migration distance through inhibition of the trailing edge contraction, mediated by enhancing the activation of PKC-MLC2 signaling. These findings help explain the dysfunctional immune state in ketotic cows and provide information on the pathogenesis of infectious diseases secondary to ketosis.
Collapse
Affiliation(s)
- Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Wen Zeng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yunfei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Shang Jiang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiancheng Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
27
|
Taneja N, Baillargeon SM, Burnette DT. Myosin light chain kinase-driven myosin II turnover regulates actin cortex contractility during mitosis. Mol Biol Cell 2021; 32:br3. [PMID: 34319762 PMCID: PMC8684764 DOI: 10.1091/mbc.e20-09-0608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 07/02/2021] [Accepted: 07/19/2021] [Indexed: 11/11/2022] Open
Abstract
Force generation by the molecular motor myosin II (MII) at the actin cortex is a universal feature of animal cells. Despite its central role in driving cell shape changes, the mechanisms underlying MII regulation at the actin cortex remain incompletely understood. Here we show that myosin light chain kinase (MLCK) promotes MII turnover at the mitotic cortex. Inhibition of MLCK resulted in an alteration of the relative levels of phosphorylated regulatory light chain (RLC), with MLCK preferentially creating a short-lived pRLC species and Rho-associated kinase (ROCK) preferentially creating a stable ppRLC species during metaphase. Slower turnover of MII and altered RLC homeostasis on MLCK inhibition correlated with increased cortex tension, driving increased membrane bleb initiation and growth, but reduced bleb retraction during mitosis. Taken together, we show that ROCK and MLCK play distinct roles at the actin cortex during mitosis; ROCK activity is required for recruitment of MII to the cortex, while MLCK activity promotes MII turnover. Our findings support the growing evidence that MII turnover is an essential dynamic process influencing the mechanical output of the actin cortex.
Collapse
Affiliation(s)
- Nilay Taneja
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Sophie M. Baillargeon
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| | - Dylan T. Burnette
- Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37212
| |
Collapse
|
28
|
The role of actomyosin in the regulation of syndecan-1 in hyperosmosis. Biochim Biophys Acta Gen Subj 2021; 1865:129975. [PMID: 34343643 DOI: 10.1016/j.bbagen.2021.129975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/14/2021] [Accepted: 07/28/2021] [Indexed: 11/21/2022]
Abstract
INTRODUCTION The endothelial glycocalyx is susceptible to high concentration of glucose and sodium in the blood. These challenges often involve an increase in osmotic pressure which may independently alters the glycocalyx components. The glycocalyx anchors on the cell membrane via core proteins that link with the actin cytoskeleton. This study aims to investigate the role of actomyosin in the osmoregulation of syndecan-1, a core protein that bears abundant sugar chains of the glycocalyx. METHODS Human umbilical vein endothelial cells were incubated with mannitol-based hyperosmotic medium up to 2 h. The surface expression of syndecan-1 and the actin cytoskeleton were analysed by confocal microscopy, either without or with cytoskeletal manipulation. RESULTS Syndecan-1 expression was compromised when hyperosmotic challenge was prolonged for 2 h, with the normalised intensity substantially dropped to 65.78 ± 2.07% at +200 mOsm. The reduction is associated with a sustained actin hyper-polymerisation, including significant increases in cortex coverage and cytoskeletal tension. Disassembling the cortex by cytochalasin D restores syndecan-1 in hyperosmosis. Inhibition of ROCK, rather than MLCK and myosin II ATPase activity, prevents the reduction of syndecan-1. CONCLUSION We have demonstrated that prolonged hyperosmotic stress disrupts the integrity of syndecan-1 through an aberrant cortex polymerisation. Our results provide new evidence in the interplay between the glycocalyx and the actin. It helps us better interpret the regulation of the glycocalyx, moving towards a goal of protecting and restoring the glycocalyx under healthy and diseased conditions.
Collapse
|
29
|
Porro C, Pennella A, Panaro MA, Trotta T. Functional Role of Non-Muscle Myosin II in Microglia: An Updated Review. Int J Mol Sci 2021; 22:ijms22136687. [PMID: 34206505 PMCID: PMC8267657 DOI: 10.3390/ijms22136687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/08/2023] Open
Abstract
Myosins are a remarkable superfamily of actin-based motor proteins that use the energy derived from ATP hydrolysis to translocate actin filaments and to produce force. Myosins are abundant in different types of tissues and involved in a large variety of cellular functions. Several classes of the myosin superfamily are expressed in the nervous system; among them, non-muscle myosin II (NM II) is expressed in both neurons and non-neuronal brain cells, such as astrocytes, oligodendrocytes, endothelial cells, and microglia. In the nervous system, NM II modulates a variety of functions, such as vesicle transport, phagocytosis, cell migration, cell adhesion and morphology, secretion, transcription, and cytokinesis, as well as playing key roles during brain development, inflammation, repair, and myelination functions. In this review, we will provide a brief overview of recent emerging roles of NM II in resting and activated microglia cells, the principal regulators of immune processes in the central nervous system (CNS) in both physiological and pathological conditions. When stimulated, microglial cells react and produce a number of mediators, such as pro-inflammatory cytokines, free radicals, and nitric oxide, that enhance inflammation and contribute to neurodegenerative diseases. Inhibition of NM II could be a new therapeutic target to treat or to prevent CNS diseases.
Collapse
Affiliation(s)
- Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Antonio Pennella
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70125 Bari, Italy;
| | - Teresa Trotta
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy; (C.P.); (A.P.)
- Correspondence:
| |
Collapse
|
30
|
Rajakylä EK, Lehtimäki JI, Acheva A, Schaible N, Lappalainen P, Krishnan R, Tojkander S. Assembly of Peripheral Actomyosin Bundles in Epithelial Cells Is Dependent on the CaMKK2/AMPK Pathway. Cell Rep 2021; 30:4266-4280.e4. [PMID: 32209483 DOI: 10.1016/j.celrep.2020.02.096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 12/02/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
Defects in the maintenance of intercellular junctions are associated with loss of epithelial barrier function and consequent pathological conditions, including invasive cancers. Epithelial integrity is dependent on actomyosin bundles at adherens junctions, but the origin of these junctional bundles is incompletely understood. Here we show that peripheral actomyosin bundles can be generated from a specific actin stress fiber subtype, transverse arcs, through their lateral fusion at cell-cell contacts. Importantly, we find that assembly and maintenance of peripheral actomyosin bundles are dependent on the mechanosensitive CaMKK2/AMPK signaling pathway and that inhibition of this route leads to disruption of tension-maintaining actomyosin bundles and re-growth of stress fiber precursors. This results in redistribution of cellular forces, defects in monolayer integrity, and loss of epithelial identity. These data provide evidence that the mechanosensitive CaMKK2/AMPK pathway is critical for the maintenance of peripheral actomyosin bundles and thus dictates cell-cell junctions through cellular force distribution.
Collapse
Affiliation(s)
- Eeva Kaisa Rajakylä
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Niccole Schaible
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
31
|
Wu F, Li H, Zhang H, Liao Y, Ren H, Wu J, Zheng D. Phosphorylated‑myosin light chain mediates the destruction of small intestinal epithelial tight junctions in mice with acute liver failure. Mol Med Rep 2021; 23:392. [PMID: 33760163 PMCID: PMC8008219 DOI: 10.3892/mmr.2021.12031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Tight junction dysregulation and epithelial damage contribute to intestinal barrier loss in patients with acute liver failure (ALF); however, the regulatory mechanisms of these processes remain poorly understood. The aim of the present study was to investigate the changes of intestinal tight junction and intestinal mucosa in mice with ALF and their mechanisms. In the present study, ALF was induced in mice through an intraperitoneal injection of D-galactosamine and lipopolysaccharide (D-GalN/LPS), and the morphological changes of the liver or small intestine were analyzed using hematoxylin and eosin staining, scanning electron microscopy (SEM) and transmission electron microscopy (TEM). The intestinal tissues and isolated serum were analyzed using western blotting, immunofluorescence staining and ELISA. D-GalN/LPS-induced mice exhibited signs of hepatocyte necrosis, alongside inflammatory cell infiltration into the liver tissue and partial microvilli detachment in the small intestinal mucosa. TEM demonstrated that the intestinal epithelial tight junctions were impaired, whereas SEM micrographs revealed the presence of abnormal microvilli in D-GalN/LPS-induced mice. In addition, the expression levels of phosphorylated (p)-myosin light chain (MLC), MLC kinase (MLCK) and Rho-associated kinase (ROCK) were significantly increased in the D-GalN/LPS-induced mice compared with those in the control mice, whereas the subsequent inhibition of MLCK or ROCK significantly reduced p-MLC expression levels. Conversely, the expression levels of occludin and zonula occludens-1 (ZO-1) were significantly decreased in the D-GalN/LPS-induced mice, and the inhibition of MLCK or ROCK significantly increased occludin and ZO-1 protein expression levels compared with those in the control group. Changes in the serum levels of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 were similar to the trend observed in p-MLC expression levels. In conclusion, the findings of the present study suggested that in a D-GalN/LPS-induced ALF model, TNF-α and IL-6 signaling may increase MLCK and ROCK expression levels, further mediate phosphorylation of MLC, which may result in tight junction dysregulation and intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Fan Wu
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Hui Li
- Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Heng Zhang
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yusheng Liao
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Huanping Ren
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jie Wu
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Dan Zheng
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
32
|
Naydenov NG, Lechuga S, Huang EH, Ivanov AI. Myosin Motors: Novel Regulators and Therapeutic Targets in Colorectal Cancer. Cancers (Basel) 2021; 13:741. [PMID: 33670106 PMCID: PMC7916823 DOI: 10.3390/cancers13040741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) remains the third most common cause of cancer and the second most common cause of cancer deaths worldwide. Clinicians are largely faced with advanced and metastatic disease for which few interventions are available. One poorly understood aspect of CRC involves altered organization of the actin cytoskeleton, especially at the metastatic stage of the disease. Myosin motors are crucial regulators of actin cytoskeletal architecture and remodeling. They act as mechanosensors of the tumor environments and control key cellular processes linked to oncogenesis, including cell division, extracellular matrix adhesion and tissue invasion. Different myosins play either oncogenic or tumor suppressor roles in breast, lung and prostate cancer; however, little is known about their functions in CRC. This review focuses on the functional roles of myosins in colon cancer development. We discuss the most studied class of myosins, class II (conventional) myosins, as well as several classes (I, V, VI, X and XVIII) of unconventional myosins that have been linked to CRC development. Altered expression and mutations of these motors in clinical tumor samples and their roles in CRC growth and metastasis are described. We also evaluate the potential of using small molecular modulators of myosin activity to develop novel anticancer therapies.
Collapse
Affiliation(s)
- Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Emina H. Huang
- Departments of Cancer Biology and Colorectal Surgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| |
Collapse
|
33
|
Ghosh I, Singh RK, Mishra M, Kapoor S, Jana SS. Switching between blebbing and lamellipodia depends on the degree of non-muscle myosin II activity. J Cell Sci 2021; 134:jcs.248732. [PMID: 33298514 DOI: 10.1242/jcs.248732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Cells can adopt both mesenchymal and amoeboid modes of migration through membrane protrusive activities, namely formation of lamellipodia and blebbing. How the molecular players control the transition between lamellipodia and blebs is yet to be explored. Here, we show that addition of the ROCK inhibitor Y27632 or low doses of blebbistatin, an inhibitor of non-muscle myosin II (NMII) ATPase activity and filament partitioning, induces blebbing to lamellipodia conversion (BLC), whereas addition of low doses of ML7, an inhibitor of myosin light chain kinase (MLCK), induces lamellipodia to blebbing conversion (LBC) in human MDA-MB-231 cells. Similarly, siRNA-mediated knockdown of ROCK and MLCK induces BLC and LBC, respectively. Interestingly, both blebs and lamellipodia membrane protrusions are able to maintain the ratio of phosphorylated to unphosphorylated regulatory light chain at cortices when MLCK and ROCK, respectively, are inhibited either pharmacologically or genetically, suggesting that MLCK and ROCK activities are interlinked in BLC and LBC. Such BLCs and LBCs are also inducible in other cell lines, including MCF7 and MCF10A. These studies reveal that the relative activity of ROCK and MLCK, which controls both the ATPase activity and filament-forming property of NMII, is a determining factor in whether a cell exhibits blebbing or lamellipodia.
Collapse
Affiliation(s)
- Indranil Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Raman K Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India.,Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Manjari Mishra
- Department of Chemistry, Indian Institute of Technology - Bombay, Mumbai 400076, India
| | - Shobhna Kapoor
- Department of Chemistry, Indian Institute of Technology - Bombay, Mumbai 400076, India
| | - Siddhartha S Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
34
|
Afrin S, Islam MS, Patzkowsky K, Malik M, Catherino WH, Segars JH, Borahay MA. Simvastatin ameliorates altered mechanotransduction in uterine leiomyoma cells. Am J Obstet Gynecol 2020; 223:733.e1-733.e14. [PMID: 32417359 DOI: 10.1016/j.ajog.2020.05.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 04/18/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress β1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies. OBJECTIVE This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells. STUDY DESIGN This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 μM, or control) for 48 hours. Protein and mRNA levels of β1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction. RESULTS We found that simvastatin significantly reduced the protein expression of β1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in β1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1. CONCLUSION The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials.
Collapse
|
35
|
Myosin light chain kinase is a potential target for hypopharyngeal cancer treatment. Biomed Pharmacother 2020; 131:110665. [PMID: 32920510 PMCID: PMC8122670 DOI: 10.1016/j.biopha.2020.110665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/08/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Hypopharyngeal cancer is squamous cell carcinoma (SCC) with the worst prognosis among the head and neck cancers. Overall, the 5-year survival rate remains poor although diagnostic imaging, radiation, chemotherapy, and surgical techniques have been improved. The mortality of patients with hypopharyngeal cancer is partly due to an increased likelihood of developing a second primary malignancy and metastasis. In this study, we found that MLCK expression, compared to healthy tissue, was up-regulated in hypopharyngeal tumor tissue. Of particular interest, a low 5-year survival rate was positively correlated with MLCK expression. We hypothesized that MLCK might be a target for hypopharyngeal cancer prognosis and treatment. In order to explore the function of MLCK in the development of cancer, we knockdown MLCK in hypopharyngeal cancer FaDu cells. The results showed that MLCK knockdown reduced the migration and invasion of FaDu cells. 4-amino-2-trifluoromethyl-phenyl retinate (ATPR) is the derivative of all-trans retinoic acid (ATRA), which was able to reduce both MLCK expression and activity in FaDu cells. ATPR induced FaDu cells apoptosis in a dose-dependent manner and also inhibited cell growth both in vivo and in vitro. Further experiments showed that overexpression of MLCK reduced ATPR induced-migration inhibition while increase of ATPR induced apoptosis, which suggested that MLCK was involved in ATPR's anti-cancer function. In conclusion, MLCK is a novel prognostic marker and therapeutic target for hypopharyngeal cancer. By targeting MLCK, ATPR exhibits its potential application in the treatment of this type of cancer.
Collapse
|
36
|
Huo Z, Su Y, Dong Y, Zheng Y, Zhang Q, Duan Y, Wang G. Rho-kinase inhibition by Fasudil promotes tumor maturation and apoptosis in small-cell lung cancer. Am J Transl Res 2020; 12:4354-4370. [PMID: 32913511 PMCID: PMC7476155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/17/2020] [Indexed: 06/11/2023]
Abstract
Small-cell lung cancer (SCLC) is a poorly differentiated neuroendocrine neoplasm with inadequate therapeutic options. Fasudil is a Rho-associated protein kinase 1 and 2 (ROCK1/2) inhibitor whose clinical indications remain limited in cardiocerebrovascular diseases. This study aimed to report a possible implication of Fasudil for SCLC. The expression and prognostic value of ROCK1/2 were investigated immunohistochemically in surgical specimens. The positive rates of ROCK1 (77/113, 68.1%) and ROCK2 (94/113, 83.2%) were distinctly higher in SCLC than in other lung neuroendocrine tumors. The high expression level of ROCK1 was related to the poor long-term survival of patients, especially in the classic SCLC subtype. In vitro, SCLC cell line treated with Fasudil exhibited synapse-like morphologic change, accompanied by a reduction in the expression levels of c-myc and cyclin D1. Cell cycle arrest was further demonstrated, accompanied by sensitivity to starvation-induced apoptosis, indicating tumor maturation. In addition, RNA-seq identified hundreds of differentially expressed genes involved in the positive regulation of neuron differentiation, stem cell differentiation, cell development, and nervous system development. Finally, Fasudil inhibited SCLC growth, promoted structural maturity, and induced apoptosis in BALB/c nude mice xenograft model. In conclusion, these results indicated a potential and novel application of Fasudil for SCLC treatment.
Collapse
Affiliation(s)
- Zitian Huo
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yinxia Su
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yuting Dong
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yiyun Zheng
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Qian Zhang
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Yaqi Duan
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| | - Guoping Wang
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
- Department of Pathology, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P. R. China
| |
Collapse
|
37
|
Yoshida GJ. Regulation of heterogeneous cancer-associated fibroblasts: the molecular pathology of activated signaling pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:112. [PMID: 32546182 PMCID: PMC7296768 DOI: 10.1186/s13046-020-01611-0] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/01/2020] [Indexed: 12/16/2022]
Abstract
Accumulating evidence indicates that intratumoral heterogeneity contributes to the development of resistance to anticancer therapeutics. Fibroblasts, which are components of the paraneoplastic stroma, play a crucial role in the wound-healing process. Activated fibroblasts accumulate in the wound and are involved in many aspects of the tissue remodeling cascade that initiates the repair process and prevents further tissue damage. The pathophysiological roles of cancer-associated fibroblasts (CAFs) in the heterogeneous tumor microenvironment have attracted increasing interest. CAFs play crucial roles in tumor progression and the response to chemotherapy. Several cytokines and chemokines are involved in the conversion of normal fibroblasts into CAFs, and some of these form a feedback loop between cancer cells and CAFs. In addition, the physical force between tumor cells and CAFs promotes cooperative invasion or co-migration of both types of cells. Pro-inflammatory cytokines, such as leukemia inhibitory factor (LIF) and interleukin-6 (IL-6), are secreted by both cancer cells and CAFs, and mediate the epigenetic modification of CAFs. This enhances the pro-tumorigenic function of CAFs mediated by promoting actomyosin contractility and extracellular matrix remodeling to form the tracks used for collective cancer cell migration. The concept of intra-tumoral CAF heterogeneity refers to the presence of inflammatory CAFs with low levels of α-smooth muscle actin (α-SMA) and high levels of IL-6 expression, which are in striking contrast to transforming growth factor-β (TGF-β)-dependent myofibroblastic CAFs with high α-SMA expression levels. CAF populations that suppress tumor growth and progression through stroma-specific Hedgehog (Hh) activation have been detected in different murine tumor models including those of the bladder, colon, and pancreas. A new therapeutic strategy targeting CAFs is the "stromal switch," in which tumor-promoting CAFs are changed into tumor-retarding CAFs with attenuated stromal stiffness. Several molecular mechanisms that can be exploited to design personalized anticancer therapies targeting CAFs remain to be elucidated. Strategies aimed at targeting the tumor stroma as well as tumor cells themselves have attracted academic attention for their application in precision medicine. This novel review discusses the role of the activation of EGFR, Wnt/β-catenin, Hippo, TGF-β, and JAK/STAT cascades in CAFs in relation to the chemoresistance and invasive/metastatic behavior of cancer cells. For instance, although activated EGFR signaling contributes to collective cell migration in cooperation with CAFs, an activated Hippo pathway is responsible for stromal stiffness resulting in the collapse of neoplastic blood vessels. Therefore, identifying the signaling pathways that are activated under specific conditions is crucial for precision medicine.
Collapse
Affiliation(s)
- Go J Yoshida
- Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
38
|
MLCK and ROCK mutualism in endothelial barrier dysfunction. Biochimie 2020; 168:83-91. [DOI: 10.1016/j.biochi.2019.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/22/2019] [Indexed: 01/15/2023]
|
39
|
Peng YS, Syu JP, Wang SD, Pan PC, Kung HN. BSA-bounded p-cresyl sulfate potentiates the malignancy of bladder carcinoma by triggering cell migration and EMT through the ROS/Src/FAK signaling pathway. Cell Biol Toxicol 2019; 36:287-300. [PMID: 31873818 DOI: 10.1007/s10565-019-09509-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Para-cresyl sulfate (P-CS), a major uremic toxin derived from the metabolites of tyrosine and phenylalanine through liver, existed in the blood of patients with chronic kidney disease (CKD). CKD increases the malignancy in bladder cancers; however, effects of P-CS on bladder cancers are not fully understood. P-CS is conjugated with BSA physiologically, and this study aims to investigate the effects and possible underlying mechanisms of BSA-bounded P-CS on human bladder cancer cells. With P-CS treatment, the intracellular ROS increased in bladder cancer cells. ROS then triggered epithelial-mesenchymal transition (EMT), stress fiber redistribution, and cell migration. With specific inhibitors, the key signals regulating P-CS-treated migration are Src and FAK. This study provided a clinical clue that patients with higher serum P-CS have a higher risk of malignant urothelial carcinomas, and a regulatory pathway of how P-CS regulates bladder cancer migration.
Collapse
Affiliation(s)
- Yu-Sen Peng
- Division of Nephrology, Department of Internal Medicine, Far Eastern Memorial Hospital, Taipei, Taiwan.,Department of Electrical Engineering, Yuan-Ze University, Taoyuan City, Taiwan
| | - Jhih-Pu Syu
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 1-1 Jen-Ai Road, 10051, Taipei, Taiwan
| | - Sheng-De Wang
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 1-1 Jen-Ai Road, 10051, Taipei, Taiwan
| | - Pie-Chun Pan
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 1-1 Jen-Ai Road, 10051, Taipei, Taiwan
| | - Hsiu-Ni Kung
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 1-1 Jen-Ai Road, 10051, Taipei, Taiwan.
| |
Collapse
|
40
|
Deng JT, Bhaidani S, Sutherland C, MacDonald JA, Walsh MP. Rho-associated kinase and zipper-interacting protein kinase, but not myosin light chain kinase, are involved in the regulation of myosin phosphorylation in serum-stimulated human arterial smooth muscle cells. PLoS One 2019; 14:e0226406. [PMID: 31834925 PMCID: PMC6910671 DOI: 10.1371/journal.pone.0226406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/26/2019] [Indexed: 01/09/2023] Open
Abstract
Myosin regulatory light chain (LC20) phosphorylation plays an important role in vascular smooth muscle contraction and cell migration. Ca2+/calmodulin-dependent myosin light chain kinase (MLCK) phosphorylates LC20 (its only known substrate) exclusively at S19. Rho-associated kinase (ROCK) and zipper-interacting protein kinase (ZIPK) have been implicated in the regulation of LC20 phosphorylation via direct phosphorylation of LC20 at T18 and S19 and indirectly via phosphorylation of MYPT1 (the myosin targeting subunit of myosin light chain phosphatase, MLCP) and Par-4 (prostate-apoptosis response-4). Phosphorylation of MYPT1 at T696 and T853 inhibits MLCP activity whereas phosphorylation of Par-4 at T163 disrupts its interaction with MYPT1, exposing the sites of phosphorylation in MYPT1 and leading to MLCP inhibition. To evaluate the roles of MLCK, ROCK and ZIPK in these phosphorylation events, we investigated the time courses of phosphorylation of LC20, MYPT1 and Par-4 in serum-stimulated human vascular smooth muscle cells (from coronary and umbilical arteries), and examined the effects of siRNA-mediated MLCK, ROCK and ZIPK knockdown and pharmacological inhibition on these phosphorylation events. Serum stimulation induced rapid phosphorylation of LC20 at T18 and S19, MYPT1 at T696 and T853, and Par-4 at T163, peaking within 30–120 s. MLCK knockdown or inhibition, or Ca2+ chelation with EGTA, had no effect on serum-induced LC20 phosphorylation. ROCK knockdown decreased the levels of phosphorylation of LC20 at T18 and S19, of MYPT1 at T696 and T853, and of Par-4 at T163, whereas ZIPK knockdown decreased LC20 diphosphorylation, but increased phosphorylation of MYPT1 at T696 and T853 and of Par-4 at T163. ROCK inhibition with GSK429286A reduced serum-induced phosphorylation of LC20 at T18 and S19, MYPT1 at T853 and Par-4 at T163, while ZIPK inhibition by HS38 reduced only LC20 diphosphorylation. We also demonstrated that serum stimulation induced phosphorylation (activation) of ZIPK, which was inhibited by ROCK and ZIPK down-regulation and inhibition. Finally, basal phosphorylation of LC20 in the absence of serum stimulation was unaffected by MLCK, ROCK or ZIPK knockdown or inhibition. We conclude that: (i) serum stimulation of cultured human arterial smooth muscle cells results in rapid phosphorylation of LC20, MYPT1, Par-4 and ZIPK, in contrast to the slower phosphorylation of kinases and other proteins involved in other signaling pathways (Akt, ERK1/2, p38 MAPK and HSP27), (ii) ROCK and ZIPK, but not MLCK, are involved in serum-induced phosphorylation of LC20, (iii) ROCK, but not ZIPK, directly phosphorylates MYPT1 at T853 and Par-4 at T163 in response to serum stimulation, (iv) ZIPK phosphorylation is enhanced by serum stimulation and involves phosphorylation by ROCK and autophosphorylation, and (v) basal phosphorylation of LC20 under serum-free conditions is not attributable to MLCK, ROCK or ZIPK.
Collapse
Affiliation(s)
- Jing-Ti Deng
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sabreena Bhaidani
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Cindy Sutherland
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Justin A. MacDonald
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael P. Walsh
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
41
|
Evans F, Hernández JA, Chifflet S. Signaling pathways in cytoskeletal responses to plasma membrane depolarization in corneal endothelial cells. J Cell Physiol 2019; 235:2947-2962. [PMID: 31535377 DOI: 10.1002/jcp.29200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 08/26/2019] [Indexed: 01/01/2023]
Abstract
In previous work, we reported that plasma membrane potential depolarization (PMPD) provokes cortical F-actin remodeling in bovine corneal endothelial (BCE) cells in culture, which eventually leads to the appearance of intercellular gaps. In kidney epithelial cells it has been shown that PMPD determines an extracellular-signal-regulated kinase (ERK)/Rho-dependent increase in diphosphorylated myosin light chain (ppMLC). The present study investigated the signaling pathways involved in the response of BCE cells to PMPD. Differently to renal epithelial cells, we observed that PMPD leads to a decrease in monophosphorylated MLC (pMLC) without affecting diphosphorylated MLC. Also, that the pMLC reduction is a consequence of cyclic adenosine 3',5'-monophosphate (cAMP)/protein kinase A (PKA) activation. In addition, we found evidence that the cAMP increase mostly depends on soluble adenylyl cyclase activity. Inhibition of this enzyme reduces the effect of PMPD on the cAMP rise, F-actin remodeling, and pMLC decrease. No changes in phosho-ERK were observed, although we could determine that RhoA undergoes activation. Our results suggested that active RhoA is not involved in the intercellular gap formation. Overall, the findings of this study support the view that, differently to renal epithelial cells, in BCE cells PMPD determines cytoskeletal reorganization via activation of the cAMP/PKA pathway.
Collapse
Affiliation(s)
- Frances Evans
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Julio A Hernández
- Sección Biofísica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Silvia Chifflet
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
42
|
Zhang Q, Narayanan V, Mui KL, O'Bryan CS, Anderson RH, Kc B, Cabe JI, Denis KB, Antoku S, Roux KJ, Dickinson RB, Angelini TE, Gundersen GG, Conway DE, Lele TP. Mechanical Stabilization of the Glandular Acinus by Linker of Nucleoskeleton and Cytoskeleton Complex. Curr Biol 2019; 29:2826-2839.e4. [PMID: 31402305 PMCID: PMC6736724 DOI: 10.1016/j.cub.2019.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/03/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022]
Abstract
The nucleoskeleton and cytoskeleton are important protein networks that govern cellular behavior and are connected together by the linker of nucleoskeleton and cytoskeleton (LINC) complex. Mutations in LINC complex components may be relevant to cancer, but how cell-level changes might translate into tissue-level malignancy is unclear. We used glandular epithelial cells in a three-dimensional culture model to investigate the effect of perturbations of the LINC complex on higher order cellular architecture. We show that inducible LINC complex disruption in human mammary epithelial MCF-10A cells and canine kidney epithelial MDCK II cells mechanically destabilizes the acinus. Lumenal collapse occurs because the acinus is unstable to increased mechanical tension that is caused by upregulation of Rho-kinase-dependent non-muscle myosin II motor activity. These findings provide a potential mechanistic explanation for how disruption of LINC complex may contribute to a loss of tissue structure in glandular epithelia.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Keeley L Mui
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Christopher S O'Bryan
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA
| | | | - Birendra Kc
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Jolene I Cabe
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Kevin B Denis
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Susumu Antoku
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Kyle J Roux
- Enabling Technologies Group, Sanford Research, Sioux Falls, SD 57104, USA; Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Richard B Dickinson
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Thomas E Angelini
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA.
| | - Tanmay P Lele
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
43
|
Li DT, Sun K, Huang P, Pan CS, Yan L, Ayan A, Liu YY, Fan JY, Fang WG, Han JY. Yiqifumai injection and its main ingredients attenuate lipopolysaccharide-induced cerebrovascular hyperpermeability through a multi-pathway mode. Microcirculation 2019; 26:e12553. [PMID: 31059171 DOI: 10.1111/micc.12553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/23/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Yiqifumai injection is a compound Chinese medicine used to treat microcirculatory disturbance-related diseases clinically. Our previous study proved that Yiqifumai injection pretreatment inhibited lipopolysaccharide-induced venular albumin leakage in rat mesentery. This study aimed to investigate whether Yiqifumai injection attenuated cerebral microvascular hyperpermeability and corresponding contribution of its main ingredients. METHODS Rats were challenged by lipopolysaccharide infusion (5 mg/kg/h) for 90 minutes. Yiqifumai injection (160 mg/kg/h), Rb1 (5 mg/kg/h), Sch (2.5 mg/kg/h), and Rb1 (5 mg/kg/h) + Sch (2.5 mg/kg/h) were infused 30 minutes before (pretreatment) or after (post-treatment) lipopolysaccharide administration. RESULTS Both pretreatment and post-treatment with Yiqifumai injection attenuated cerebral venular albumin leakage during lipopolysaccharide infusion and cerebrovascular hyperpermeability at 72 hours after lipopolysaccharide infusion. Yiqifumai injection restrained the decreased junction protein expression, adenosine triphosphate content, and mitochondria complex I, II, IV, and V activities. Moreover, Yiqifumai injection inhibited toll-like receptor-4 expression, Src phosphorylation, and caveolin-1 expression. Its main ingredients Rb1 and Sch alone worked differently, with Rb1 being more effective for enhancing energy metabolism, while Sch attenuating toll-like receptor-4 expression and Src activation. CONCLUSION Yiqifumai injection exerts a protective and ameliorated effect on cerebral microvascular hyperpermeability, which is more effective than any of its ingredients, possibly due to the interaction of its main ingredients through a multi-pathway mode.
Collapse
Affiliation(s)
- Dan-Tong Li
- Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China
| | - Kai Sun
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Ping Huang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Ayididaer Ayan
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Yu-Ying Liu
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| | - Wei-Gang Fang
- Department of Pathology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.,Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing, China.,State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing, China.,Beijing Microvascular Institute of Integration of Chinese and Western Medicine, Beijing, China
| |
Collapse
|
44
|
Zhovmer AS, Tabdanov ED, Miao H, Wen H, Chen J, Luo X, Ma X, Provenzano PP, Adelstein RS. The role of nonmuscle myosin 2A and 2B in the regulation of mesenchymal cell contact guidance. Mol Biol Cell 2019; 30:1961-1973. [PMID: 31318315 PMCID: PMC6727766 DOI: 10.1091/mbc.e19-01-0071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Contact guidance refers to the ability of cells to sense the geometrical features of the microenvironment and respond by changing their shape and adopting the appropriate orientation. Inhibition and ablation of nonmuscle myosin 2 (NM2) paralogues have demonstrated their importance for contact guidance. However, the specific roles of the NM2 paralogues have not been systematically studied. In this work we use micropatterned substrates to examine the roles of NM2A and NM2B and to elucidate the relationship of the microenvironment, actomyosin, and microtubules in contact guidance. We show that contact guidance is preserved following loss of NM2B and that expression of NM2A alone is sufficient to establish an appropriate orientation of the cells. Loss of NM2B and overexpression of NM2A result in a prominent cell polarization that is found to be linked to the increased alignment of microtubules with the actomyosin scaffold. Suppression of actomyosin with blebbistatin reduces cell polarity on a flat surface, but not on a surface with contact guidance cues. This indicates that the lost microtubule-actomyosin interactions are compensated for by microtubule-microenvironment interactions, which are sufficient to establish cell polarity through contact guidance.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Erdem D Tabdanov
- Laboratory for Engineering in Oncology, University of Minnesota, Minneapolis, MN 55455
| | - Houxun Miao
- Imaging Physics Laboratory, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Han Wen
- Imaging Physics Laboratory, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Jinqiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, Bethesda, MD 20892
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, Bethesda, MD 20892
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| | - Paolo P Provenzano
- Laboratory for Engineering in Oncology, University of Minnesota, Minneapolis, MN 55455
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, Bethesda, MD 20814
| |
Collapse
|
45
|
Hughes JH, Ewy JM, Chen J, Wong SY, Tharp KM, Stahl A, Kumar S. Transcriptomic analysis reveals that BMP4 sensitizes glioblastoma tumor-initiating cells to mechanical cues. Matrix Biol 2019; 85-86:112-127. [PMID: 31189077 DOI: 10.1016/j.matbio.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 12/15/2022]
Abstract
The poor prognosis of glioblastoma (GBM) is associated with a highly invasive stem-like subpopulation of tumor-initiating cells (TICs), which drive recurrence and contribute to intra-tumoral heterogeneity through differentiation. These TICs are better able to escape extracellular matrix-imposed mechanical restrictions on invasion than their more differentiated progeny, and sensitization of TICs to extracellular matrix mechanics extends survival in preclinical models of GBM. However, little is known about the molecular basis of the relationship between TIC differentiation and mechanotransduction. Here we explore this relationship through a combination of transcriptomic analysis and studies with defined-stiffness matrices. We show that TIC differentiation induced by bone morphogenetic protein 4 (BMP4) suppresses expression of proteins relevant to extracellular matrix signaling and sensitizes TIC spreading to matrix stiffness. Moreover, our findings point towards a previously unappreciated connection between BMP4-induced differentiation, mechanotransduction, and metabolism. Notably, stiffness and differentiation modulate oxygen consumption, and inhibition of oxidative phosphorylation influences cell spreading in a stiffness- and differentiation-dependent manner. Our work integrates bioinformatic analysis with targeted molecular measurements and perturbations to yield new insight into how morphogen-induced differentiation influences how GBM TICs process mechanical inputs.
Collapse
Affiliation(s)
- Jasmine H Hughes
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeanette M Ewy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph Chen
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sophie Y Wong
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kevin M Tharp
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California San Francisco, San Francisco, CA 94720, USA
| | - Andreas Stahl
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- UC Berkeley - UCSF Graduate Program in Bioengineering; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
46
|
Halder D, Saha S, Singh RK, Ghosh I, Mallick D, Dey SK, Ghosh A, Das BB, Ghosh S, Jana SS. Nonmuscle myosin IIA and IIB differentially modulate migration and alter gene expression in primary mouse tumorigenic cells. Mol Biol Cell 2019; 30:1463-1476. [PMID: 30995168 PMCID: PMC6724700 DOI: 10.1091/mbc.e18-12-0790] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/22/2022] Open
Abstract
Though many cancers are known to show up-regulation of nonmuscle myosin (NM) IIA and IIB, the mechanism by which NMIIs aid in cancer development remains unexplored. Here we demonstrate that tumor-generating, fibroblast-like cells isolated from 3-methylcholanthrene (3MC)-induced murine tumor exhibit distinct phospho-dependent localization of NMIIA and NMIIB at the perinuclear area and tip of the filopodia and affect cell migration differentially. While NMIIA-KD affects protrusion dynamics and increases cell directionality, NMIIB-KD lowers migration speed and increases filopodial branching. Strategically located NMIIs at the perinuclear area colocalize with the linker of nucleoskeleton and cytoskeleton (LINC) protein Nesprin2 and maintain the integrity of the nuclear-actin cap. Interestingly, knockdown of NMIIs results in altered expression of genes involved in epithelial-to-mesenchymal transition, angiogenesis, and cellular senescence. NMIIB-KD cells display down-regulation of Gsc and Serpinb2, which is strikingly similar to Nesprin2-KD cells as assessed by quantitative PCR analysis. Further gene network analysis predicts that NMIIA and NMIIB may act on similar pathways but through different regulators. Concomitantly, knockdown of NMIIA or NMIIB lowers the growth rate and tumor volume of 3MC-induced tumor in vivo. Altogether, these results open a new window to further investigate the effect of LINC-associated perinuclear actomyosin complex on mechanoresponsive gene expression in the growing tumor.
Collapse
Affiliation(s)
- Debdatta Halder
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Shekhar Saha
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908
| | - Raman K. Singh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610010, Israel
| | - Indranil Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Ditipriya Mallick
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Sumit K. Dey
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853
| | - Arijit Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Benu Brata Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | | | - Siddhartha S. Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
47
|
Matsui TS, Deguchi S. Spatially selective myosin regulatory light chain regulation is absent in dedifferentiated vascular smooth muscle cells but is partially induced by fibronectin and Klf4. Am J Physiol Cell Physiol 2019; 316:C509-C521. [DOI: 10.1152/ajpcell.00251.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The phosphorylation state of myosin regulatory light chain (MRLC) is central to the regulation of contractility that impacts cellular homeostasis and fate decisions. Rho-kinase (ROCK) and myosin light chain kinase (MLCK) are major kinases for MRLC documented to selectively regulate MRLC in a subcellular position-specific manner; specifically, MLCK in some nonmuscle cell types works in the cell periphery to promote migration, while ROCK does so at the central region to sustain contractility. However, it remains unclear whether or not the spatially selective regulation of the MRLC kinases is universally present in other cell types, including dedifferentiated vascular smooth muscle cells (SMCs). Here, we demonstrate the absence of the spatial regulation in dedifferentiated SMCs using both cell lines and primary cells. Thus, our work is distinct from previous reports on cells with migratory potential. We also observed that the spatial regulation is partly induced upon fibronectin stimulation and Krüppel-like factor 4 overexpression. To find clues to the mechanism, we reveal how the phosphorylation state of MRLC is determined within dedifferentiated A7r5 SMCs under the enzymatic competition among three major regulators ROCK, MLCK, and MRLC phosphatase (MLCP). We show that ROCK, but not MLCK, predominantly regulates the MRLC phosphorylation in a manner distinct from previous in vitro-based and in silico-based reports. In this ROCK-dominating cellular system, the contractility at physiological conditions was regulated at the level of MRLC diphosphorylation, because its monophosphorylation is already saturated. Thus, the present study provides insights into the molecular basis underlying the absence of spatial MRLC regulation in dedifferentiated SMCs.
Collapse
Affiliation(s)
- Tsubasa S. Matsui
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Shinji Deguchi
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| |
Collapse
|
48
|
Schwager SC, Taufalele PV, Reinhart-King CA. Cell-Cell Mechanical Communication in Cancer. Cell Mol Bioeng 2019; 12:1-14. [PMID: 31565083 PMCID: PMC6764766 DOI: 10.1007/s12195-018-00564-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Communication between cancer cells enables cancer progression and metastasis. While cell-cell communication in cancer has primarily been examined through chemical mechanisms, recent evidence suggests that mechanical communication through cell-cell junctions and cell-ECM linkages is also an important mediator of cancer progression. Cancer and stromal cells remodel the ECM through a variety of mechanisms, including matrix degradation, cross-linking, deposition, and physical remodeling. Cancer cells sense these mechanical environmental changes through cell-matrix adhesion complexes and subsequently alter their tension between both neighboring cells and the surrounding matrix, thereby altering the force landscape within the microenvironment. This communication not only allows cancer cells to communicate with each other, but allows stromal cells to communicate with cancer cells through matrix remodeling. Here, we review the mechanisms of intercellular force transmission, the subsequent matrix remodeling, and the implications of this mechanical communication on cancer progression.
Collapse
Affiliation(s)
- Samantha C. Schwager
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| | - Paul V. Taufalele
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, PMB 351631, Nashville, TN 37235 USA
| |
Collapse
|
49
|
Abstract
MYH9 was first discovered due to thrombocytopenia caused by MYH9 mutation-related abnormalities. In recent years, researchers have increasingly found that MYH9 plays an important role in cancer as a cytokine involved in cytoskeletal reorganization, cellular pseudopodia formation, and migration. MYH9 is closely related to the progress and poor prognosis of most solid tumors, and it is now accepted that MYH9 is a suppressor gene and plays an important role on the re-Rho pathway. Recent research has been limited to the study of tissues. However, it would be more direct and informative to be able to use hematology to assess tumor prognosis and changes in MYH9 levels and NMMHC-IIA. This article summarizes recent research on MYH9 and provides a reference for future clinical research.
Collapse
Affiliation(s)
- Yunmei Wang
- Shaanxi Provincial Cancer Hospital Affiliated to Medical School, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Shuguang Liu
- Hong Hui Hospital, The Affiliated Hospital, School of Medicine, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Yanjun Zhang
- Shaanxi Provincial Cancer Hospital Affiliated to Medical School, Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| | - Jin Yang
- First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
50
|
George Z, Omosun Y, Azenabor AA, Goldstein J, Partin J, Joseph K, Ellerson D, He Q, Eko F, McDonald MA, Reed M, Svoboda P, Stuchlik O, Pohl J, Lutter E, Bandea C, Black CM, Igietseme JU. The molecular mechanism of induction of unfolded protein response by Chlamydia. Biochem Biophys Res Commun 2019; 508:421-429. [PMID: 30503337 PMCID: PMC6343654 DOI: 10.1016/j.bbrc.2018.11.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 11/06/2018] [Indexed: 11/21/2022]
Abstract
The unfolded protein response (UPR) contributes to chlamydial pathogenesis, as a source of lipids and ATP during replication, and for establishing the initial anti-apoptotic state of host cell that ensures successful inclusion development. The molecular mechanism(s) of UPR induction by Chlamydia is unknown. Chlamydia use type III secretion system (T3SS) effector proteins (e.g, the Translocated Actin-Recruiting Phosphoprotein (Tarp) to stimulate host cell's cytoskeletal reorganization that facilitates invasion and inclusion development. We investigated the hypothesis that T3SS effector-mediated assembly of myosin-II complex produces activated non-muscle myosin heavy chain II (NMMHC-II), which then binds the UPR master regulator (BiP) and/or transducers to induce UPR. Our results revealed the interaction of the chlamydial effector proteins (CT228 and Tarp) with components of the myosin II complex and UPR regulator and transducer during infection. These interactions caused the activation and binding of NMMHC-II to BiP and IRE1α leading to UPR induction. In addition, specific inhibitors of myosin light chain kinase, Tarp oligomerization and myosin ATPase significantly reduced UPR activation and Chlamydia replication. Thus, Chlamydia induce UPR through T3SS effector-mediated activation of NMMHC-II components of the myosin complex to facilitate infectivity. The finding provides greater insights into chlamydial pathogenesis with the potential to identify therapeutic targets and formulations.
Collapse
Affiliation(s)
- Zenas George
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Yusuf Omosun
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA; Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Jason Goldstein
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - James Partin
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Kahaliah Joseph
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Debra Ellerson
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Qing He
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA; Morehouse School of Medicine, Atlanta, GA, USA
| | - Francis Eko
- Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Matthew Reed
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Pavel Svoboda
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Olga Stuchlik
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Jan Pohl
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | | | - Claudiu Bandea
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Carolyn M Black
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Joseph U Igietseme
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA; Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|