1,906 results on '"stress fibers"'
Search Results
2. Mechanically induced topological transition of spectrin regulates its distribution in the mammalian cell cortex.
- Author
-
Ghisleni, Andrea, Bonilla-Quintana, Mayte, Crestani, Michele, Lavagnino, Zeno, Galli, Camilla, Rangamani, Padmini, and Gauthier, Nils
- Subjects
Spectrin ,Animals ,Fibroblasts ,Actomyosin ,Mice ,Cytoskeleton ,Stress ,Mechanical ,Cell Membrane ,Cell Shape ,Actins ,Stress Fibers ,Humans - Abstract
The cell cortex is a dynamic assembly formed by the plasma membrane and underlying cytoskeleton. As the main determinant of cell shape, the cortex ensures its integrity during passive and active deformations by adapting cytoskeleton topologies through yet poorly understood mechanisms. The spectrin meshwork ensures such adaptation in erythrocytes and neurons by adopting different organizations. Erythrocytes rely on triangular-like lattices of spectrin tetramers, whereas in neurons they are organized in parallel, periodic arrays. Since spectrin is ubiquitously expressed, we exploited Expansion Microscopy to discover that, in fibroblasts, distinct meshwork densities co-exist. Through biophysical measurements and computational modeling, we show that the non-polarized spectrin meshwork, with the intervention of actomyosin, can dynamically transition into polarized clusters fenced by actin stress fibers that resemble periodic arrays as found in neurons. Clusters experience lower mechanical stress and turnover, despite displaying an extension close to the tetramer contour length. Our study sheds light on the adaptive properties of spectrin, which participates in the protection of the cell cortex by varying its densities in response to key mechanical features.
- Published
- 2024
3. Plakophilin 4 controls the spatio-temporal activity of RhoA at adherens junctions to promote cortical actin ring formation and tissue tension.
- Author
-
Müller, Lisa, Keil, René, Glaß, Markus, and Hatzfeld, Mechthild
- Subjects
- *
ADHERENS junctions , *ACTIN , *CELL junctions , *CELL communication , *ACTOMYOSIN - Abstract
Plakophilin 4 (PKP4) is a component of cell–cell junctions that regulates intercellular adhesion and Rho-signaling during cytokinesis with an unknown function during epidermal differentiation. Here we show that keratinocytes lacking PKP4 fail to develop a cortical actin ring, preventing adherens junction maturation and generation of tissue tension. Instead, PKP4-depleted cells display increased stress fibers. PKP4-dependent RhoA localization at AJs was required to activate a RhoA-ROCK2-MLCK-MLC2 axis and organize actin into a cortical ring. AJ-associated PKP4 provided a scaffold for the Rho activator ARHGEF2 and the RhoA effectors MLCK and MLC2, facilitating the spatio-temporal activation of RhoA signaling at cell junctions to allow cortical ring formation and actomyosin contraction. In contrast, association of PKP4 with the Rho suppressor ARHGAP23 reduced ARHGAP23 binding to RhoA which prevented RhoA activation in the cytoplasm and stress fiber formation. These data identify PKP4 as an AJ component that transduces mechanical signals into cytoskeletal organization. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. An Elementary Model of Focal Adhesion Detachment and Reattachment During Cell Reorientation Using Ideas from the Kinetics of Wiggly Energies.
- Author
-
Abeyaratne, Rohan, Puntel, Eric, and Tomassetti, Giuseppe
- Subjects
FOCAL adhesions ,FIBER orientation ,ADHESION ,ACTIVATION energy ,STRETCH (Physiology) - Abstract
A simple, transparent, two-dimensional, nonlinear model of cell reorientation is constructed in this paper. The cells are attached to a substrate by "focal adhesions" that transmit the deformation of the substrate to the "stress fibers" in the cell. When the substrate is subjected to a deformation, say an in-plane bi-axial deformation with stretches λ 1 and λ 2 , the stress fibers deform with it and change their length and orientation. In addition, the focal adhesions can detach from the substrate and reattach to it at new nearby locations, and this process of detachment and reattachment can happen many times. In this scenario the (varying) fiber angle Θ in the reference configuration plays the role of an internal variable. In addition to the elastic energy of the stress fibers, the energy associated with the focal adhesions is accounted for by a wiggly energy ϵ a cos Θ / ϵ , 0 < ϵ ≪ 1 . Each local minimum of this energy corresponds to a particular configuration of the focal adhesions. The small amplitude ϵ a indicates that the energy barrier between two neighboring configurations is relatively small, and the small distance 2 π ϵ between the local minima indicates that a focal adhesion does not have to move very far before it reattaches. The evolution of this system is studied using a gradient flow kinetic law, which is homogenized for ϵ → 0 using results from weak convergence. The results determine (a) a region of the λ 1 , λ 2 -plane in which the (referential) fiber orientation remains stuck at the angle Θ and does not evolve, and (b) the evolution of the orientation when the stretches move out of this region as the fibers seek to minimize energy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. LUZP1 regulates the maturation of contractile actomyosin bundles.
- Author
-
Wang, Liang, Tsang, Hoi Ying, Yan, Ziyi, Tojkander, Sari, Ciuba, Katarzyna, Kogan, Konstantin, Liu, Xiaonan, and Zhao, Hongxia
- Subjects
- *
ACTOMYOSIN , *MUSCLE contraction , *CELL migration , *MYOSIN , *LEUCINE zippers - Abstract
Contractile actomyosin bundles play crucial roles in various physiological processes, including cell migration, morphogenesis, and muscle contraction. The intricate assembly of actomyosin bundles involves the precise alignment and fusion of myosin II filaments, yet the underlying mechanisms and factors involved in these processes remain elusive. Our study reveals that LUZP1 plays a central role in orchestrating the maturation of thick actomyosin bundles. Loss of LUZP1 caused abnormal cell morphogenesis, migration, and the ability to exert forces on the environment. Importantly, knockout of LUZP1 results in significant defects in the concatenation and persistent association of myosin II filaments, severely impairing the assembly of myosin II stacks. The disruption of these processes in LUZP1 knockout cells provides mechanistic insights into the defective assembly of thick ventral stress fibers and the associated cellular contractility abnormalities. Overall, these results significantly contribute to our understanding of the molecular mechanism involved in actomyosin bundle formation and highlight the essential role of LUZP1 in this process. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Intracellular Macromolecular Crowding within Individual Stress Fibers Analyzed by Fluorescence Correlation Spectroscopy.
- Author
-
Buenaventura, Aria, Saito, Takumi, Kanao, Taiga, Matsunaga, Daiki, Matsui, Tsubasa S., and Deguchi, Shinji
- Subjects
- *
FLUORESCENCE spectroscopy , *GREEN fluorescent protein , *DIFFUSION coefficients , *CELL anatomy , *MACROMOLECULES - Abstract
Introduction: The diffusion of cell components such as proteins is crucial to the function of all living cells. The abundance of macromolecules in cells is likely to cause a state of macromolecular crowding, but its effects on the extent of diffusion remain poorly understood. Methods: Here we investigate the diffusion rate in three distinct locations in mesenchymal cell types, namely the open cytoplasm, the stress fibers in the open cytoplasm, and those below the nucleus using three kinds of biologically inert green fluorescent proteins (GFPs), namely a monomer, dimer, and trimer GFP. Fluorescence correlation spectroscopy (FCS) was used to determine the diffusion coefficients. Results: We show that diffusion tends to be lowered on average in stress fibers and is significantly lower in those located below the nucleus. Our data suggest that the diffusive properties of GFPs, and potentially other molecules as well, are hindered by macromolecular crowding. However, although the size dependence on protein diffusion was also studied for monomer, dimer, and trimer GFPs, there was no significant difference in the diffusion rates among the GFPs of these sizes. These results could be attributed to the lack of significant change in protein size among the selected GFP multimers. Conclusion: The data presented here would provide a basis for better understanding of the complex protein diffusion in the nonuniform cytoplasm, shedding light on cellular responses to mechanical stress, their local mechanical properties, and reduced turnover in senescent cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Are the class 18 myosins Myo18A and Myo18B specialist sarcomeric proteins?
- Author
-
Horsthemke, Markus, Arnaud, Charles-Adrien, and Hanley, Peter J.
- Subjects
MYOSIN ,MOLECULAR motor proteins ,STRIATED muscle ,MUSCULAR hypertrophy ,MUSCLE cells ,PROTEINS - Abstract
Initially, the two members of class 18 myosins, Myo18A and Myo18B, appeared to exhibit highly divergent functions, complicating the assignment of class-specific functions. However, the identification of a striated muscle-specific isoform of Myo18A, Myo18Aγ, suggests that class 18 myosins may have evolved to complement the functions of conventional class 2 myosins in sarcomeres. Indeed, both genes, Myo18a and Myo18b, are predominantly expressed in the heart and somites, precursors of skeletal muscle, of developing mouse embryos. Genetic deletion of either gene in mice is embryonic lethal and is associated with the disorganization of cardiac sarcomeres. Moreover, Myo18Aγ and Myo18B localize to sarcomeric A-bands, albeit the motor (head) domains of these unconventional myosins have been both deduced and biochemically demonstrated to exhibit negligible ATPase activity, a hallmark of motor proteins. Instead, Myo18Aγ and Myo18B presumably coassemble with thick filaments and provide structural integrity and/or internal resistance through interactions with F-actin and/or other proteins. In addition, Myo18Aγ and Myo18B may play distinct roles in the assembly of myofibrils, which may arise from actin stress fibers containing the α-isoform of Myo18A, Myo18Aα. The β- isoform of Myo18A, Myo18Aβ, is similar to Myo18Aα, except that it lacks the N-terminal extension, and may serve as a negative regulator through heterodimerization with either Myo18Aα or Myo18Aγ. In this review, we contend that Myo18Aγ and Myo18B are essential for myofibril structure and function in striated muscle cells, while α- and β-isoforms of Myo18A play diverse roles in nonmuscle cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Elevated expression levels of the protein kinase DYRK1B induce mesenchymal features in A549 lung cancer cells
- Author
-
Sester, Soraya, Wilms, Gerrit, Ahlburg, Joana, Babendreyer, Aaron, and Becker, Walter
- Published
- 2024
- Full Text
- View/download PDF
9. Inhibition of EphA4 reduces vasogenic edema after experimental stroke in mice by protecting the blood-brain barrier integrity.
- Author
-
Zhang, Shuai, Zhao, Jing, Sha, Wei-Meng, Zhang, Xin-Pei, Mai, Jing-Yuan, Bartlett, Perry F, and Hou, Sheng-Tao
- Abstract
Cerebral vasogenic edema, a severe complication of ischemic stroke, aggravates neurological deficits. However, therapeutics to reduce cerebral edema still represent a significant unmet medical need. Brain microvascular endothelial cells (BMECs), vital for maintaining the blood-brain barrier (BBB), represent the first defense barrier for vasogenic edema. Here, we analyzed the proteomic profiles of the cultured mouse BMECs during oxygen-glucose deprivation and reperfusion (OGD/R). Besides the extensively altered cytoskeletal proteins, ephrin type-A receptor 4 (EphA4) expressions and its activated phosphorylated form p-EphA4 were significantly increased. Blocking EphA4 using EphA4-Fc, a specific and well-tolerated inhibitor shown in our ongoing human phase I trial, effectively reduced OGD/R-induced BMECs contraction and tight junction damage. EphA4-Fc did not protect OGD/R-induced neuronal and astrocytic death. However, administration of EphA4-Fc, before or after the onset of transient middle cerebral artery occlusion (tMCAO), reduced brain edema by about 50%, leading to improved neurological function recovery. The BBB permeability test also confirmed that cerebral BBB integrity was well maintained in tMCAO brains treated with EphA4-Fc. Therefore, EphA4 was critical in signaling BMECs-mediated BBB breakdown and vasogenic edema during cerebral ischemia. EphA4-Fc is promising for the treatment of clinical post-stroke edema. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. The mechanical mechanism of angiotensin II induced activation of hepatic stellate cells promoting portal hypertension
- Author
-
Yiheng Zhang, Mulan Xing, Fansheng Meng, Ling Zhu, Qingchuan Huang, Tianle Ma, Huihua Fang, Xujing Gu, Suzhou Huang, Xinyu Wu, Gaohong Lv, Jun Guo, Li Wu, Xin Liu, and Zhipeng Chen
- Subjects
Hepatic stellate cell ,Portal hypertension ,α-SMA ,Microfilaments ,Stress fibers ,COL1A1 ,Cytology ,QH573-671 - Abstract
In the development of chronic liver disease, the hepatic stellate cell (HSC) plays a pivotal role in increasing intrahepatic vascular resistance (IHVR) and inducing portal hypertension (PH) in cirrhosis. Our research demonstrated that HSC contraction, prompted by angiotensin II (Ang II), significantly contributed to the elevation of type I collagen (COL1A1) expression. This increase was intimately associated with enhanced cell tension and YAP nuclear translocation, mediated through α-smooth muscle actin (α-SMA) expression, microfilaments (MF) polymerization, and stress fibers (SF) assembly. Further investigation revealed that the Rho/ROCK signaling pathway regulated MF polymerization and SF assembly by facilitating the phosphorylation of cofilin and MLC, while Ca2+ chiefly governed SF assembly via MLC. Inhibiting α-SMA-MF-SF assembly changed Ang II-induced cell contraction, YAP nuclear translocation, and COL1A1 expression, findings corroborated in cirrhotic mice models. Overall, our study offers insights into mitigating IHVR and PH through cell mechanics, heralding potential breakthroughs.
- Published
- 2024
- Full Text
- View/download PDF
11. Targeting F-actin stress fibers to suppress the dedifferentiated phenotype in chondrocytes
- Author
-
Mandy M. Schofield, Alissa T. Rzepski, Stephanie Richardson-Solorzano, Jonah Hammerstedt, Sohan Shah, Chloe E. Mirack, Marin Herrick, and Justin Parreno
- Subjects
Actin ,Chondrocytes ,CDC42 ,TPM3.1 ,MRTF ,Stress fibers ,Cytology ,QH573-671 - Abstract
Actin is a central mediator of the chondrocyte phenotype. Monolayer expansion of articular chondrocytes on tissue culture polystyrene, for cell-based repair therapies, leads to chondrocyte dedifferentiation. During dedifferentiation, chondrocytes spread and filamentous (F-)actin reorganizes from a cortical to a stress fiber arrangement causing a reduction in cartilage matrix expression and an increase in fibroblastic matrix and contractile molecule expression. While the downstream mechanisms regulating chondrocyte molecular expression by alterations in F-actin organization have become elucidated, the critical upstream regulators of F-actin networks in chondrocytes are not completely known. Tropomyosin (TPM) and the RhoGTPases are known regulators of F-actin networks. The main purpose of this study is to elucidate the regulation of passaged chondrocyte F-actin stress fiber networks and cell phenotype by the specific TPM, TPM3.1, and the RhoGTPase, CDC42. Our results demonstrated that TPM3.1 associates with cortical F-actin and stress fiber F-actin in primary and passaged chondrocytes, respectively. In passaged cells, we found that pharmacological TPM3.1 inhibition or siRNA knockdown causes F-actin reorganization from stress fibers back to cortical F-actin and causes an increase in G/F-actin. CDC42 inhibition also causes formation of cortical F-actin. However, pharmacological CDC42 inhibition, but not TPM3.1 inhibition, leads to the re-association of TPM3.1 with cortical F-actin. Both TPM3.1 and CDC42 inhibition, as well as TPM3.1 knockdown, reduces nuclear localization of myocardin related transcription factor, which suppresses dedifferentiated molecule expression. We confirmed that TPM3.1 or CDC42 inhibition partially redifferentiates passaged cells by reducing fibroblast matrix and contractile expression, and increasing chondrogenic SOX9 expression. A further understanding on the regulation of F-actin in passaged cells may lead into new insights to stimulate cartilage matrix expression in cells for regenerative therapies.
- Published
- 2024
- Full Text
- View/download PDF
12. Are the class 18 myosins Myo18A and Myo18B specialist sarcomeric proteins?
- Author
-
Markus Horsthemke, Charles-Adrien Arnaud, and Peter J. Hanley
- Subjects
unconventional myosins ,MYO18A ,MYO18B ,sarcomere ,stress fibers ,knockout (KO) mice ,Physiology ,QP1-981 - Abstract
Initially, the two members of class 18 myosins, Myo18A and Myo18B, appeared to exhibit highly divergent functions, complicating the assignment of class-specific functions. However, the identification of a striated muscle-specific isoform of Myo18A, Myo18Aγ, suggests that class 18 myosins may have evolved to complement the functions of conventional class 2 myosins in sarcomeres. Indeed, both genes, Myo18a and Myo18b, are predominantly expressed in the heart and somites, precursors of skeletal muscle, of developing mouse embryos. Genetic deletion of either gene in mice is embryonic lethal and is associated with the disorganization of cardiac sarcomeres. Moreover, Myo18Aγ and Myo18B localize to sarcomeric A-bands, albeit the motor (head) domains of these unconventional myosins have been both deduced and biochemically demonstrated to exhibit negligible ATPase activity, a hallmark of motor proteins. Instead, Myo18Aγ and Myo18B presumably coassemble with thick filaments and provide structural integrity and/or internal resistance through interactions with F-actin and/or other proteins. In addition, Myo18Aγ and Myo18B may play distinct roles in the assembly of myofibrils, which may arise from actin stress fibers containing the α-isoform of Myo18A, Myo18Aα. The β-isoform of Myo18A, Myo18Aβ, is similar to Myo18Aα, except that it lacks the N-terminal extension, and may serve as a negative regulator through heterodimerization with either Myo18Aα or Myo18Aγ. In this review, we contend that Myo18Aγ and Myo18B are essential for myofibril structure and function in striated muscle cells, while α- and β-isoforms of Myo18A play diverse roles in nonmuscle cells.
- Published
- 2024
- Full Text
- View/download PDF
13. Caldesmon controls stress fiber force-balance through dynamic cross-linking of myosin II and actin-tropomyosin filaments
- Author
-
Kokate, Shrikant B, Ciuba, Katarzyna, Tran, Vivien D, Kumari, Reena, Tojkander, Sari, Engel, Ulrike, Kogan, Konstantin, Kumar, Sanjay, and Lappalainen, Pekka
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Actin Cytoskeleton ,Actins ,Actomyosin ,Calmodulin-Binding Proteins ,Muscle ,Smooth ,Myosin Type II ,Myosins ,Stress Fibers ,Tropomyosin - 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.
- Published
- 2022
14. Analysis of the Tensioning Field Induced by Stress Fibers in Nanoindented Stem Cells Adhered to a Flat Substrate
- Author
-
Vaiani, Lorenzo, Uva, Antonio Emmanuele, Boccaccio, Antonio, Chaari, Fakher, Series Editor, Gherardini, Francesco, Series Editor, Ivanov, Vitalii, Series Editor, Cavas-Martínez, Francisco, Editorial Board Member, di Mare, Francesca, Editorial Board Member, Haddar, Mohamed, Editorial Board Member, Kwon, Young W., Editorial Board Member, Trojanowska, Justyna, Editorial Board Member, Gerbino, Salvatore, editor, Lanzotti, Antonio, editor, Martorelli, Massimo, editor, Mirálbes Buil, Ramón, editor, Rizzi, Caterina, editor, and Roucoules, Lionel, editor
- Published
- 2023
- Full Text
- View/download PDF
15. Characterizing the cellular architecture of dynamically remodeling vascular tissue using 3-D image analysis and virtual reconstruction
- Author
-
Valentine, Megan T
- Subjects
Epithelial mechanics ,Botryllus ,morphology ,blood vessels ,stress fibers ,cell shape ,image analysis - Abstract
Epithelial tubules form critical structures in lung, kidney and vascular tissues. However, the processes that control their morphogenesis and physiological expansion and contraction are not well understood. Here we examine the dynamic remodeling of epithelial tubes in vivo using a novel model system: the extracorporeal vasculature of Botryllus schlosseri, in which the disruption of the basement membrane triggers rapid, massive vascular retraction without loss of barrier function. We developed and implemented 3-D image analysis and virtual reconstruction tools to characterize the cellular morphology of the vascular wall in unmanipulated vessels and during retraction. In both control and regressed conditions, cells within the vascular wall were planar polarized, with an integrin- and curvature-dependent axial elongation of cells and a robust circumferential alignment of actin bundles. Surprisingly, we found no measurable differences in morphology between normal and retracting vessels under ECM disruption. However, inhibition of integrin signaling through FAK inhibition caused disruption of cellular actin organization. Our results demonstrate that epithelial tubes can maintain tissue organization even during extreme remodeling events, but that the robust response to mechanical signals – such as the response to loss of vascular tension after ECM disruption - requires functional force sensing machinery via integrin signaling.
- Published
- 2020
16. Direct evidence that tumor cells soften when navigating confined spaces
- Author
-
Rianna, Carmela, Radmacher, Manfred, and Kumar, Sanjay
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Women's Health ,Bioengineering ,Cancer ,Generic health relevance ,Adaptor Proteins ,Signal Transducing ,Cell Line ,Tumor ,Cell Movement ,Cell Nucleus ,Cytoplasm ,Elasticity ,Extracellular Matrix ,Hardness ,Humans ,Microscopy ,Atomic Force ,Neoplasms ,Stress Fibers ,Transcription Factors ,YAP-Signaling Proteins ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
The mechanical properties of cells strongly regulate many physiological and pathological processes. For example, in cancer, invasive and metastatic tumor cells have often been reported to be softer than nontumor cells, raising speculation that cancer cells might adaptively soften to facilitate migration through narrow tissue spaces. Despite growing interest in targeting cell softening to impede invasion and metastasis, it remains to be directly demonstrated that tumor cells soften as they migrate through confined spaces. Here, we address this open question by combining topographically patterned substrates with atomic force microscopy (AFM). Using a polydimethylsiloxane open-roof microdevice featuring tapered, fibronectin-coated channels, we followed the migration of U2OS cells through various stages of confinement while simultaneously performing AFM indentation. As cells progress from unconfined migration to fully confined migration, cells soften and exclude Yes-associated protein from the nucleus. Superresolution imaging reveals that confinement induces remodeling of actomyosin stress fiber architecture. Companion studies with flat one-dimensional microlines indicate that the changes in cytoarchitecture and mechanics are intrinsically driven by topographical confinement rather than changes in cellular aspect ratio. Our studies represent among the most direct evidence to date that tumor cells soften during confined migration and support cell softening as a mechanoadaptive mechanism during invasion.
- Published
- 2020
17. Toxoplasma gondii Dysregulates Barrier Function and Mechanotransduction Signaling in Human Endothelial Cells
- Author
-
Franklin-Murray, Armond L, Mallya, Sharmila, Jankeel, Allen, Sureshchandra, Suhas, Messaoudi, Ilhem, and Lodoen, Melissa B
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Vaccine Related ,Infectious Diseases ,Emerging Infectious Diseases ,Biodefense ,Prevention ,Foodborne Illness ,Aetiology ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Infection ,Actins ,Adaptor Proteins ,Signal Transducing ,Antigens ,CD ,Cadherins ,Cell Membrane Permeability ,Cell Polarity ,Cells ,Cultured ,Cytoskeleton ,Hippo Signaling Pathway ,Human Umbilical Vein Endothelial Cells ,Humans ,Mechanotransduction ,Cellular ,Protein Serine-Threonine Kinases ,RNA-Seq ,Stress Fibers ,Toxoplasma ,Transcription Factors ,Transcriptome ,YAP-Signaling Proteins ,beta Catenin ,Hippo signaling ,Toxoplasma gondii ,VE-cadherin ,actin ,endothelial cell ,mechanotransduction ,Toxoplasma gondii ,Microbiology - Abstract
Toxoplasma gondii can infect and replicate in vascular endothelial cells prior to entering host tissues. However, little is known about the molecular interactions at the parasite-endothelial cell interface. We demonstrate that T. gondii infection of primary human umbilical vein endothelial cells (HUVEC) altered cell morphology and dysregulated barrier function, increasing permeability to low-molecular-weight polymers. T. gondii disrupted vascular endothelial cadherin (VE-cadherin) and β-catenin localization to the cell periphery and reduced VE-cadherin protein expression. Notably, T. gondii infection led to reorganization of the host cytoskeleton by reducing filamentous actin (F-actin) stress fiber abundance under static and microfluidic shear stress conditions and by reducing planar cell polarity. RNA sequencing (RNA-Seq) comparing genome-wide transcriptional profiles of infected to uninfected endothelial cells revealed changes in gene expression associated with cell-cell adhesion, extracellular matrix reorganization, and cytokine-mediated signaling. In particular, genes downstream of Hippo signaling and the biomechanical sensor and transcriptional coactivator Yes-associated protein (YAP) were downregulated in infected endothelial cells. Interestingly, T. gondii infection activated Hippo signaling by increasing phosphorylation of LATS1, leading to cytoplasmic retention of YAP, and reducing YAP target gene expression. These findings suggest that T. gondii infection triggers Hippo signaling and YAP nuclear export, leading to an altered transcriptional profile of infected endothelial cells.IMPORTANCE Toxoplasma gondii is a foodborne parasite that infects virtually all warm-blooded animals and can cause severe disease in individuals with compromised or weakened immune systems. During dissemination in its infected hosts, T. gondii breaches endothelial barriers to enter tissues and establish the chronic infections underlying the most severe manifestations of toxoplasmosis. The research presented here examines how T. gondii infection of primary human endothelial cells induces changes in cell morphology, barrier function, gene expression, and mechanotransduction signaling under static conditions and under the physiological conditions of shear stress found in the bloodstream. Understanding the molecular interactions occurring at the interface between endothelial cells and T. gondii may provide insights into processes linked to parasite dissemination and pathogenesis.
- Published
- 2020
18. Functional differences in human aortic valve interstitial cells from patients with varying calcific aortic valve disease.
- Author
-
Tuscher, Robin, Khang, Alex, West, Toni M., Camillo, Chiara, Ferrari, Giovanni, and Sacks, Michael S.
- Subjects
AORTIC valve diseases ,AORTIC valve ,INTERSTITIAL cells ,MITRAL valve ,STRAINS & stresses (Mechanics) ,AORTIC valve insufficiency ,MITRAL valve prolapse - Abstract
Calcific aortic valve disease (CAVD) is characterized by progressive stiffening of aortic valve (AV) tissues, inducing stenosis and insufficiency. Bicuspid aortic valve (BAV) is a common congenital defect in which the AV has two leaflets rather than three, with BAV patients developing CAVD decades years earlier than in the general population. Current treatment for CAVD remains surgical replacement with its continued durability problems, as there are no pharmaceutical therapies or other alternative treatments available. Before such therapeutic approaches can be developed, a deeper understanding of CAVD disease mechanisms is clearly required. It is known that AV interstitial cells (AVICs) maintain the AV extracellular matrix and are typically quiescent in the normal state, transitioning into an activated, myofibroblast-like state during periods of growth or disease. One proposed mechanism of CAVD is the subsequent transition of AVICs into an osteoblast-like phenotype. A sensitive indicator of AVIC phenotypic state is enhanced basal contractility (tonus), so that AVICs from diseased AV will exhibit a higher basal tonus level. The goals of the present study were thus to assess the hypothesis that different human CAVD states lead to different biophysical AVIC states. To accomplish this, we characterized AVIC basal tonus behaviors from diseased human AV tissues embedded in 3D hydrogels. Established methods were utilized to track AVIC-induced gel displacements and shape changes after the application of Cytochalasin D (an actin polymerization inhibitor) to depolymerize the AVIC stress fibers. Results indicated that human diseased AVICs from the non-calcified region of TAVs were significantly more activated than AVICs from the corresponding calcified region. In addition, AVICs from the raphe region of BAVs were more activated than from the non-raphe region. Interestingly, we observed significantly greater basal tonus levels in females compared to males. Furthermore, the overall AVIC shape changes after Cytochalasin suggested that AVICs from TAVs and BAVs develop different stress fiber architectures. These findings are the first evidence of sex-specific differences in basal tonus state in human AVICs in varying disease states. Future studies are underway to quantify stress fiber mechanical behaviors to further elucidate CAVD disease mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. Three-dimensional analysis of hydrogel-imbedded aortic valve interstitial cell shape and its relation to contractile behavior.
- Author
-
Khang, Alex, Nguyen, Quan, Feng, Xinzeng, Howsmon, Daniel P., and Sacks, Michael S.
- Subjects
AORTIC valve ,INTERSTITIAL cells ,CELL morphology ,AORTIC valve diseases ,HEART valve diseases ,HEART valves - Abstract
Cell-shape is a conglomerate of mechanical, chemical, and biological mechanisms that reflects the cell biophysical state. In a specific application, we consider aortic valve interstitial cells (AVICs), which maintain the structure and function of aortic heart valve leaflets. Actomyosin stress fibers help determine AVIC shape and facilitate processes such as adhesion, contraction, and mechanosensing. However, detailed 3D assessment of stress fiber architecture and function is currently impractical. Herein, we assessed AVIC shape and contractile behaviors using hydrogel-based 3D traction force microscopy to intuit the orientation and behavior of AVIC stress fibers. We utilized spherical harmonics (SPHARM) to quantify AVIC geometries through three days of incubation, which demonstrated a shift from a spherical shape to forming substantial protrusions. Furthermore, we assessed changes in post-three day AVIC shape and contractile function within two testing regimes: (1) normal contractile level to relaxation (cytochalasin D), and (2) normal contractile level to hyper-contraction (endothelin-1). In both scenarios, AVICs underwent isovolumic shape changes and produced complex displacement fields within the hydrogel. AVICs were more elongated when relaxed and more spherical in hyper-contraction. Locally, AVIC protrusions contracted along their long axis and expanded in their circumferential direction, indicating predominately axially aligned stress fibers. Furthermore, the magnitude of protrusion displacements was correlated with protrusion length and approached a consistent displacement plateau at a similar critical length across all AVICs. This implied that stress fiber behavior is conserved, despite great variations in AVIC shapes. We anticipate our findings will bolster future investigations into AVIC stress fiber architecture and function. Within the aortic valve there exists a population of aortic valve interstitial cells, which orchestrate the turnover, secretion, and remodeling of its extracellular matrix, maintaining tissue integrity and ultimately sustaining the proper mechanical function. Alterations in these processes are thought to underlie diseases of the aortic valve, which affect hundreds of thousands domestically and world-wide. Yet, to date, there are no non-surgical treatments for aortic heart valve disease, in part due to our limited understanding of the underlying disease processes. In the present study, we built upon our previous study to include a full 3D analysis of aortic valve interstitial cell shapes at differing contractile levels. The resulting detailed shape and deformation analysis provided insight into the underlying stress-fiber structures and mechanical behaviors. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Registry Kinetics of Myosin Motor Stacks Driven by Mechanical Force-Induced Actin Turnover
- Author
-
Dasbiswas, Kinjal, Hu, Shiqiong, Bershadsky, Alexander D, and Safran, Samuel A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Bioengineering ,1.1 Normal biological development and functioning ,Actins ,Animals ,Biomechanical Phenomena ,Elasticity ,Kinetics ,Myosins ,Polymerization ,Rats ,Stress Fibers ,Physical Sciences ,Chemical Sciences ,Biophysics ,Biological sciences ,Chemical sciences ,Physical sciences - Abstract
Actin filaments associated with myosin motors constitute the cytoskeletal force-generating machinery for many types of adherent cells. These actomyosin units are structurally ordered in muscle cells and, in particular, may be spatially registered across neighboring actin bundles. Such registry or stacking of myosin filaments have been recently observed in ordered actin bundles of even fibroblasts with super-resolution microscopy techniques. We introduce here a model for the dynamics of stacking arising from long-range mechanical interactions between actomyosin units through mutual contractile deformations of the intervening cytoskeletal network. The dynamics of registry involve two key processes: 1) polymerization and depolymerization of actin filaments and 2) remodeling of cross-linker-rich actin adhesion zones, both of which are, in principle, mechanosensitive. By calculating the elastic forces that drive registry and their effect on actin polymerization rates, we estimate a characteristic timescale of tens of minutes for registry to be established, in agreement with experimentally observed timescales for individual kinetic processes involved in myosin stack formation, which we track and quantify. This model elucidates the role of actin turnover dynamics in myosin stacking and explains the loss of stacks seen when actin assembly or disassembly and cross-linking is experimentally disrupted in fibroblasts.
- Published
- 2019
21. Extracellular Matrix Geometry and Initial Adhesive Position Determine Stress Fiber Network Organization during Cell Spreading
- Author
-
Kassianidou, Elena, Probst, Dimitri, Jäger, Julia, Lee, Stacey, Roguet, Anne-Lou, Schwarz, Ulrich Sebastian, and Kumar, Sanjay
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Actin Cytoskeleton ,Cell Adhesion ,Cell Line ,Tumor ,Cell Movement ,Collagen ,Computer Simulation ,Extracellular Matrix ,Half-Life ,Heterocyclic Compounds ,4 or More Rings ,Humans ,Kinetics ,Models ,Biological ,Pseudopodia ,Stress Fibers ,Time Factors ,actin cytoskeleton ,cell memory ,cell migration ,cell shape ,cell spreading ,cell-matrix adhesion ,cellular Potts model ,mathematical modeling ,mechanobiology ,stress fibers ,Medical Physiology ,Biological sciences - Abstract
Three-dimensional matrices often contain highly structured adhesive tracks that require cells to turn corners and bridge non-adhesive areas. Here, we investigate these complex processes using micropatterned cell adhesive frames. Spreading kinetics on these matrices depend strongly on initial adhesive position and are predicted by a cellular Potts model (CPM), which reflects a balance between adhesion and intracellular tension. As cells spread, new stress fibers (SFs) assemble periodically and parallel to the leading edge, with spatial intervals of ∼2.5 μm, temporal intervals of ∼15 min, and characteristic lifetimes of ∼50 min. By incorporating these rules into the CPM, we can successfully predict SF network architecture. Moreover, we observe broadly similar behavior when we culture cells on arrays of discrete collagen fibers. Our findings show that ECM geometry and initial cell position strongly determine cell spreading and that cells encode a memory of their spreading history through SF network organization.
- Published
- 2019
22. IL-17A Recruits Rab35 to IL-17R to Mediate PKCα-Dependent Stress Fiber Formation and Airway Smooth Muscle Contractility
- Author
-
Bulek, Katarzyna, Chen, Xing, Parron, Vandy, Sundaram, Aparna, Herjan, Tomasz, Ouyang, Suidong, Liu, Caini, Majors, Alana, Zepp, Jarod, Gao, Ji, Dongre, Ashok, Bodaszewska-Lubas, Malgorzata, Echard, Arnaud, Aronica, Mark, Carman, Julie, Garantziotis, Stavros, Sheppard, Dean, and Li, Xiaoxia
- Subjects
Lung ,Asthma ,Aetiology ,2.1 Biological and endogenous factors ,Respiratory ,Animals ,Interleukin-17 ,Mice ,Mice ,Knockout ,Muscle Contraction ,Muscle ,Smooth ,Protein Kinase C-alpha ,Protein Kinase Inhibitors ,Receptors ,Interleukin-17 ,Stress Fibers ,rab GTP-Binding Proteins ,Immunology - Abstract
IL-17A is a critical proinflammatory cytokine for the pathogenesis of asthma including neutrophilic pulmonary inflammation and airway hyperresponsiveness. In this study, by cell type-specific deletion of IL-17R and adaptor Act1, we demonstrated that IL-17R/Act1 exerts a direct impact on the contraction of airway smooth muscle cells (ASMCs). Mechanistically, IL-17A induced the recruitment of Rab35 (a small monomeric GTPase) and DennD1C (guanine nucleotide exchange factor [GEF]) to the IL-17R/Act1 complex in ASMCs, resulting in activation of Rab35. Rab35 knockdown showed that IL-17A-induced Rab35 activation was essential for protein kinase Cα (PKCα) activation and phosphorylation of fascin at Ser39 in ASMCs, allowing F-actin to interact with myosin to form stress fibers and enhance the contraction induced by methacholine. PKCα inhibitor or Rab35 knockdown indeed substantially reduced IL-17A-induced stress fiber formation in ASMCs and attenuated IL-17A-enhanced, methacholine-induced contraction of airway smooth muscle. Taken together, these data indicate that IL-17A promotes airway smooth muscle contraction via direct recruitment of Rab35 to IL-17R, followed by PKCα activation and stress fiber formation.
- Published
- 2019
23. Functional differences in human aortic valve interstitial cells from patients with varying calcific aortic valve disease
- Author
-
Robin Tuscher, Alex Khang, Toni M. West, Chiara Camillo, Giovanni Ferrari, and Michael S. Sacks
- Subjects
aortic valve ,bicuspid aortic valve ,aortic valve interstitial cells ,contractility ,stress fibers ,calcific aortic valve disease (CAVD) ,Physiology ,QP1-981 - Abstract
Calcific aortic valve disease (CAVD) is characterized by progressive stiffening of aortic valve (AV) tissues, inducing stenosis and insufficiency. Bicuspid aortic valve (BAV) is a common congenital defect in which the AV has two leaflets rather than three, with BAV patients developing CAVD decades years earlier than in the general population. Current treatment for CAVD remains surgical replacement with its continued durability problems, as there are no pharmaceutical therapies or other alternative treatments available. Before such therapeutic approaches can be developed, a deeper understanding of CAVD disease mechanisms is clearly required. It is known that AV interstitial cells (AVICs) maintain the AV extracellular matrix and are typically quiescent in the normal state, transitioning into an activated, myofibroblast-like state during periods of growth or disease. One proposed mechanism of CAVD is the subsequent transition of AVICs into an osteoblast-like phenotype. A sensitive indicator of AVIC phenotypic state is enhanced basal contractility (tonus), so that AVICs from diseased AV will exhibit a higher basal tonus level. The goals of the present study were thus to assess the hypothesis that different human CAVD states lead to different biophysical AVIC states. To accomplish this, we characterized AVIC basal tonus behaviors from diseased human AV tissues embedded in 3D hydrogels. Established methods were utilized to track AVIC-induced gel displacements and shape changes after the application of Cytochalasin D (an actin polymerization inhibitor) to depolymerize the AVIC stress fibers. Results indicated that human diseased AVICs from the non-calcified region of TAVs were significantly more activated than AVICs from the corresponding calcified region. In addition, AVICs from the raphe region of BAVs were more activated than from the non-raphe region. Interestingly, we observed significantly greater basal tonus levels in females compared to males. Furthermore, the overall AVIC shape changes after Cytochalasin suggested that AVICs from TAVs and BAVs develop different stress fiber architectures. These findings are the first evidence of sex-specific differences in basal tonus state in human AVICs in varying disease states. Future studies are underway to quantify stress fiber mechanical behaviors to further elucidate CAVD disease mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
24. Rat Hepatic Stellate Cell Line CFSC-2G: Genetic Markers and Short Tandem Repeat Profile Useful for Cell Line Authentication.
- Author
-
Nanda, Indrajit, Schröder, Sarah K., Steinlein, Claus, Haaf, Thomas, Buhl, Eva M., Grimm, Domink G., and Weiskirchen, Ralf
- Subjects
- *
MICROSATELLITE repeats , *LIVER cells , *TANDEM repeats , *CELL lines , *GENETIC markers , *EXTRACELLULAR matrix , *KARYOTYPES , *FIBROBLASTS - Abstract
Hepatic stellate cells (HSCs) are also known as lipocytes, fat-storing cells, perisinusoidal cells, or Ito cells. These liver-specific mesenchymal cells represent about 5% to 8% of all liver cells, playing a key role in maintaining the microenvironment of the hepatic sinusoid. Upon chronic liver injury or in primary culture, these cells become activated and transdifferentiate into a contractile phenotype, i.e., the myofibroblast, capable of producing and secreting large quantities of extracellular matrix compounds. Based on their central role in the initiation and progression of chronic liver diseases, cultured HSCs are valuable in vitro tools to study molecular and cellular aspects of liver diseases. However, the isolation of these cells requires special equipment, trained personnel, and in some cases needs approval from respective authorities. To overcome these limitations, several immortalized HSC lines were established. One of these cell lines is CFSC, which was originally established from cirrhotic rat livers induced by carbon tetrachloride. First introduced in 1991, this cell line and derivatives thereof (i.e., CFSC-2G, CFSC-3H, CFSC-5H, and CFSC-8B) are now used in many laboratories as an established in vitro HSC model. We here describe molecular features that are suitable for cell authentication. Importantly, chromosome banding and multicolor spectral karyotyping (SKY) analysis demonstrate that the CFSC-2G genome has accumulated extensive chromosome rearrangements and most chromosomes exist in multiple copies producing a pseudo-triploid karyotype. Furthermore, our study documents a defined short tandem repeat (STR) profile including 31 species-specific markers, and a list of genes expressed in CFSC-2G established by bulk mRNA next-generation sequencing (NGS). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Polarized light retardation analysis allows for the evaluation of tension in individual stress fibers.
- Author
-
Sugita, Shukei, Hozaki, Masatoshi, Matsui, Tsubasa S., Nagayama, Kazuaki, Deguchi, Shinji, and Nakamura, Masanori
- Subjects
- *
VASCULAR smooth muscle , *ELECTRON microscopes , *FIBERS , *OPTICAL microscopes , *OPTICAL images , *PHOTOELASTICITY , *PROTEIN synthesis - Abstract
The tension in the stress fibers (SFs) of cells plays a pivotal role in determining biological processes such as cell migration, morphological formation, and protein synthesis. Our previous research developed a method to evaluate the cellular contraction force generated in SFs based on photoelasticity-associated retardation of polarized light; however, we employed live cells, which could have caused an increase in retardation and not contraction force. Therefore, the present study aimed to confirm that polarized light retardation increases inherently due to contraction, regardless of cell activity. We also explored the reason why retardation increased with SF contractions. We used SFs physically isolated from vascular smooth muscle cells to stop cell activity. The retardation of SFs was measured after ATP administration, responsible for contracting SFs. The SFs were imaged under optical and electron microscopes to measure SF length, width, and retardation. The retardation of isolated SFs after ATP administration was significantly higher than before. Thus, we confirmed that retardation increased with elevated tension in individual SFs. Furthermore, the SF diameter decreased while the SF length remained almost constant. Thus, we conclude that a contraction force-driven increase in the density of SFs is the main factor for the rise in polarized light retardation. • Polarized light retardation (PLR) of stress fibers (SFs) increases intrinsically with tension. • PLR can be used for evaluating tension in individual SFs. • The lateral width of individual SFs decreases with tension. • PLR of SFs is caused by the tension-induced increase in the structural density. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. The formin FMNL2 plays a role in the response of melanoma cells to substrate stiffness.
- Subjects
INTRACELLULAR space ,CELL morphology ,MORPHOLOGY ,CYTOLOGY ,BIOCHEMICAL substrates - Abstract
The article explores the impact of substrate stiffness on the morphology and motility of A2058 human melanoma cells, focusing on the role of the formin protein FMNL2 in regulating these responses. The study found that increasing substrate stiffness led to more elongated cell morphologies and increased actin stress fiber alignment, with FMNL2 depletion amplifying these effects. Additionally, substrate stiffness influenced cell motility, with an optimal stiffness maximizing motility before a decrease in distance traveled on stiffer substrates. The research suggests that FMNL2 is crucial for maintaining cell motility and adaptability under varying stiffness conditions, highlighting its role as a mechanosensitive effector in melanoma cells. [Extracted from the article]
- Published
- 2025
27. Inhibition of stress fiber formation preserves blood–brain barrier after intracerebral hemorrhage in mice
- Author
-
Manaenko, Anatol, Yang, Peng, Nowrangi, Derek, Budbazar, Enkhjargal, Hartman, Richard E, Obenaus, Andre, Pearce, William J, Zhang, John H, and Tang, Jiping
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Brain Disorders ,Stroke ,Neurosciences ,Animals ,Blood-Brain Barrier ,Capillary Permeability ,Intracranial Hemorrhages ,Male ,Mice ,Stress Fibers ,Cortactin ,intracerebral hemorrhage ,LIM kinase ,PDGFR-beta ,stress fibers ,PDGFR-β ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Neurology & Neurosurgery ,Clinical sciences - Abstract
Intracerebral hemorrhage (ICH) represents the deadliest subtype of all strokes. The development of brain edema, a consequence of blood-brain barrier (BBB) disruption, is the most life-threatening event after ICH. Pathophysiological conditions activate the endothelium, one of the components of BBB, inducing rearrangement of the actin cytoskeleton. Upon activation, globular actin assembles into a filamentous actin resulting in the formation of contractile actin bundles, stress fibers. The contraction of stress fibers leads to the formation of intercellular gaps between endothelial cells increasing the permeability of BBB. In the present study, we investigated the effect of ICH on stress fiber formation in CD1 mice. We hypothesized that ICH-induced formation of stress fiber is triggered by the activation of PDGFR-β and mediated by the cortactin/RhoA/LIMK pathway. We demonstrated that ICH induces formation of stress fibers. Furthermore, we demonstrated that the inhibition of PDGFR-β and its downstream reduced the number of stress fibers, preserving BBB and resulting in the amelioration of brain edema and improvement of neurological functions in mice after ICH.
- Published
- 2018
28. Study Data from Nagoya Institute of Technology Update Knowledge of Hypertension (Stress fiber strain is zero in normal aortic smooth muscle, elevated in hypertensive stretch, and minimal in wall thickening rats).
- Subjects
INTRACELLULAR space ,THORACIC aorta ,BLOOD pressure ,REPORTERS & reporting ,CARDIOVASCULAR diseases - Abstract
Researchers from the Nagoya Institute of Technology conducted a study on hypertension, focusing on stress fiber (SF) tension in aortic smooth muscle cells. The study compared SF strain in normal rats, rats with initial hypertension, and rats simulating after wall thickening. The findings suggest that SFs function as mechanosensors in response to hypertension. For more information, the full article can be accessed in Scientific Reports. [Extracted from the article]
- Published
- 2024
29. Activation of ROCK and MLCK tunes regional stress fiber formation and mechanics via preferential myosin light chain phosphorylation
- Author
-
Kassianidou, Elena, Hughes, Jasmine H, and Kumar, Sanjay
- Subjects
1.1 Normal biological development and functioning ,Underpinning research ,Cell Line ,Tumor ,Humans ,Muscle ,Smooth ,Myosin Light Chains ,Myosin-Light-Chain Kinase ,Phosphorylation ,Stress Fibers ,rho-Associated Kinases ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
The assembly and mechanics of actomyosin stress fibers (SFs) depend on myosin regulatory light chain (RLC) phosphorylation, which is driven by myosin light chain kinase (MLCK) and Rho-associated kinase (ROCK). Although previous work suggests that MLCK and ROCK control distinct pools of cellular SFs, it remains unclear how these kinases differ in their regulation of RLC phosphorylation or how phosphorylation influences individual SF mechanics. Here, we combine genetic approaches with biophysical tools to explore relationships between kinase activity, RLC phosphorylation, SF localization, and SF mechanics. We show that graded MLCK overexpression increases RLC monophosphorylation (p-RLC) in a graded manner and that this p-RLC localizes to peripheral SFs. Conversely, graded ROCK overexpression preferentially increases RLC diphosphorylation (pp-RLC), with pp-RLC localizing to central SFs. Interrogation of single SFs with subcellular laser ablation reveals that MLCK and ROCK quantitatively regulate the viscoelastic properties of peripheral and central SFs, respectively. The effects of MLCK and ROCK on single-SF mechanics may be correspondingly phenocopied by overexpression of mono- and diphosphomimetic RLC mutants. Our results point to a model in which MLCK and ROCK regulate peripheral and central SF viscoelastic properties through mono- and diphosphorylation of RLC, offering new quantitative connections between kinase activity, RLC phosphorylation, and SF viscoelasticity.
- Published
- 2017
30. Geometry and network connectivity govern the mechanics of stress fibers
- Author
-
Kassianidou, Elena, Brand, Christoph A, Schwarz, Ulrich S, and Kumar, Sanjay
- Subjects
Actomyosin ,Cell Movement ,Cell Polarity ,Cytoskeleton ,Models ,Theoretical ,Single-Cell Analysis ,Stress Fibers ,Stress ,Mechanical ,cytoskeleton ,active cable model ,actin ,myosin ,cell migration - Abstract
Actomyosin stress fibers (SFs) play key roles in driving polarized motility and generating traction forces, yet little is known about how tension borne by an individual SF is governed by SF geometry and its connectivity to other cytoskeletal elements. We now address this question by combining single-cell micropatterning with subcellular laser ablation to probe the mechanics of single, geometrically defined SFs. The retraction length of geometrically isolated SFs after cutting depends strongly on SF length, demonstrating that longer SFs dissipate more energy upon incision. Furthermore, when cell geometry and adhesive spacing are fixed, cell-to-cell heterogeneities in SF dissipated elastic energy can be predicted from varying degrees of physical integration with the surrounding network. We apply genetic, pharmacological, and computational approaches to demonstrate a causal and quantitative relationship between SF connectivity and mechanics for patterned cells and show that similar relationships hold for nonpatterned cells allowed to form cell-cell contacts in monolayer culture. Remarkably, dissipation of a single SF within a monolayer induces cytoskeletal rearrangements in cells long distances away. Finally, stimulation of cell migration leads to characteristic changes in network connectivity that promote SF bundling at the cell rear. Our findings demonstrate that SFs influence and are influenced by the networks in which they reside. Such higher order network interactions contribute in unexpected ways to cell mechanics and motility.
- Published
- 2017
31. Myofibroblast transdifferentiation of keratocytes results in slower migration and lower sensitivity to mesoscale curvatures
- Author
-
Cas van der Putten, Daniëlle van den Broek, and Nicholas A. Kurniawan
- Subjects
cell migration ,substrate curvature ,corneal keratocyte ,myofibroblast transdifferentiation ,stress fibers ,Biology (General) ,QH301-705.5 - Abstract
Functional tissue repair after injury or disease is governed by the regenerative or fibrotic response by cells within the tissue. In the case of corneal damage, keratocytes are a key cell type that determine the outcome of the remodeling response by either adapting to a fibroblast or myofibroblast phenotype. Although a growing body of literature indicates that geometrical cues in the environment can influence Myo(fibroblast) phenotype, there is a lack of knowledge on whether and how differentiated keratocyte phenotype is affected by the curved tissue geometry in the cornea. To address this gap, in this study we characterized the phenotype of fibroblastic and transforming growth factor β (TGFβ)-induced myofibroblastic keratocytes and studied their migration behavior on curved culture substrates with varying curvatures. Immunofluorescence staining and quantification of cell morphological parameters showed that, generally, fibroblastic keratocytes were more likely to elongate, whereas myofibroblastic keratocytes expressed more pronounced α smooth muscle actin (α-SMA) and actin stress fibers as well as more mature focal adhesions. Interestingly, keratocyte adhesion on convex structures was weak and unstable, whereas they adhered normally on flat and concave structures. On concave cylinders, fibroblastic keratocytes migrated faster and with higher persistence along the longitudinal direction compared to myofibroblastic keratocytes. Moreover, this behavior became more pronounced on smaller cylinders (i.e., higher curvatures). Taken together, both keratocyte phenotypes can sense and respond to the sign and magnitude of substrate curvatures, however, myofibroblastic keratocytes exhibit weaker curvature sensing and slower migration on curved substrates compared to fibroblastic keratocytes. These findings provide fundamental insights into keratocyte phenotype after injury, but also exemplify the potential of tuning the physical cell environments in tissue engineering settings to steer towards a favorable regeneration response.
- Published
- 2022
- Full Text
- View/download PDF
32. Cells Dynamically Adapt to Surface Geometry by Remodeling Their Focal Adhesions and Actin Cytoskeleton
- Author
-
Aysegul Dede Eren, Amy W. A. Lucassen, Urandelger Tuvshindorj, Roman Truckenmüller, Stefan Giselbrecht, E. Deniz Eren, Mehmet Orhan Tas, Phanikrishna Sudarsanam, and Jan de Boer
- Subjects
mechanotransduction ,focal adhesion ,tenocytes ,cell shape ,stress fibers ,Biology (General) ,QH301-705.5 - Abstract
Cells probe their environment and adapt their shape accordingly via the organization of focal adhesions and the actin cytoskeleton. In an earlier publication, we described the relationship between cell shape and physiology, for example, shape-induced differentiation, metabolism, and proliferation in mesenchymal stem cells and tenocytes. In this study, we investigated how these cells organize their adhesive machinery over time when exposed to microfabricated surfaces of different topographies and adhesive island geometries. We further examined the reciprocal interaction between stress fiber and focal adhesion formation by pharmacological perturbations. Our results confirm the current literature that spatial organization of adhesive sites determines the ability to form focal adhesions and stress fibers. Therefore, cells on roughened surfaces have smaller focal adhesion and fewer stress fibers. Our results further highlight the importance of integrin-mediated adhesion in the adaptive properties of cells and provide clear links to the development of bioactive materials.
- Published
- 2022
- Full Text
- View/download PDF
33. MAGI1 localizes to mature focal adhesion and modulates endothelial cell adhesion, migration and angiogenesis
- Author
-
Begoña Alday-Parejo, Kedar Ghimire, Oriana Coquoz, Gioele W. Albisetti, Luca Tamò, Jelena Zaric, Jimmy Stalin, and Curzio Rüegg
- Subjects
cell adhesion ,migration ,signaling ,extracellular matrix ,cytoskeleton ,stress fibers ,Cytology ,QH573-671 - Abstract
MAGI1 is an intracellular adaptor protein that stabilizes cell junctions and regulates epithelial and endothelial integrity. Here, we report that that in endothelial cells MAGI1 colocalizes with paxillin, β3-integrin, talin 1, tensin 3 and α-4-actinin at mature focal adhesions and actin stress fibers, and regulates their dynamics. Downregulation of MAGI1 reduces focal adhesion formation and maturation, cell spreading, actin stress fiber formation and RhoA/Rac1 activation. MAGI1 silencing increases phosphorylation of paxillin at Y118, an indicator of focal adhesion turnover. MAGI1 promotes integrin-dependent endothelial cells adhesion to ECM, reduces invasion and tubulogenesisin vitro and suppresses angiogenesis in vivo. Our results identify MAGI1 as anovel component of focal adhesions, and regulator of focal adhesion dynamics, cell adhesion, invasion and angiogenesis.
- Published
- 2021
- Full Text
- View/download PDF
34. Circulating Fibroblast Growth Factor-2, HIV-Tat, and Vascular Endothelial Cell Growth Factor-A in HIV-Infected Children with Renal Disease Activate Rho-A and Src in Cultured Renal Endothelial Cells.
- Author
-
Das, Jharna, Gutkind, J, and Ray, Patricio
- Subjects
Cell Line ,Cells ,Cultured ,Endothelial Cells ,Enzyme Activation ,Fibroblast Growth Factor 2 ,HIV Infections ,HIV-1 ,Heparin ,Humans ,Kidney Diseases ,Permeability ,Podocytes ,Signal Transduction ,Stress Fibers ,Vascular Endothelial Growth Factor A ,rhoA GTP-Binding Protein ,src-Family Kinases ,tat Gene Products ,Human Immunodeficiency Virus - Abstract
Renal endothelial cells (REc) are the first target of HIV-1 in the kidney. The integrity of REc is maintained at least partially by heparin binding growth factors that bind to heparan sulfate proteoglycans located on their cell surface. However, previous studies showed that the accumulation of two heparin-binding growth factors, Vascular Endothelial Cell Growth Factor-A (VEGF-A) and Fibroblast Growth Factor-2 (FGF-2), in combination with the viral protein Tat, can precipitate the progression of HIV-renal diseases. Nonetheless, very little is known about how these factors affect the behavior of REc in HIV+ children. We carried out this study to determine how VEGF-A, FGF-2, and HIV-Tat, modulate the cytoskeletal structure and permeability of cultured REc, identify key signaling pathways involved in this process, and develop a functional REc assay to detect HIV+ children affected by these changes. We found that VEGF-A and FGF-2, acting in synergy with HIV-Tat and heparin, affected the cytoskeletal structure and permeability of REc through changes in Rho-A, Src, and Rac-1 activity. Furthermore, urine samples from HIV+ children with renal diseases, showed high levels of VEGF-A and FGF-2, and induced similar changes in cultured REc and podocytes. These findings suggest that FGF-2, VEGF-A, and HIV-Tat, may affect the glomerular filtration barrier in HIV+ children through the induction of synergistic changes in Rho-A and Src activity. Further studies are needed to define the clinical value of the REc assay described in this study to identify HIV+ children exposed to circulating factors that may induce glomerular injury through similar mechanisms.
- Published
- 2016
35. Mechanotransduction: use the force(s)
- Author
-
Paluch, Ewa K, Nelson, Celeste M, Biais, Nicolas, Fabry, Ben, Moeller, Jens, Pruitt, Beth L, Wollnik, Carina, Kudryasheva, Galina, Rehfeldt, Florian, and Federle, Walter
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Actomyosin ,Animals ,Biomechanical Phenomena ,Cell Adhesion ,Focal Adhesions ,Humans ,Kinetics ,Locomotion ,Mechanotransduction ,Cellular ,Mesenchymal Stem Cells ,Stress Fibers ,Developmental Biology ,Biological sciences - Abstract
Mechanotransduction - how cells sense physical forces and translate them into biochemical and biological responses - is a vibrant and rapidly-progressing field, and is important for a broad range of biological phenomena. This forum explores the role of mechanotransduction in a variety of cellular activities and highlights intriguing questions that deserve further attention.
- Published
- 2015
36. A biomechanical perspective on stress fiber structure and function
- Author
-
Kassianidou, Elena and Kumar, Sanjay
- Subjects
Bioengineering ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Animals ,Cell Adhesion ,Cell Movement ,Humans ,Mechanotransduction ,Cellular ,Stress Fibers ,Stress fiber ,Biomechanical property ,Mechanobiology ,Subcellular laser ablation ,Physical Sciences ,Biological Sciences - Abstract
Stress fibers are actomyosin-based bundles whose structural and contractile properties underlie numerous cellular processes including adhesion, motility and mechanosensing. Recent advances in high-resolution live-cell imaging and single-cell force measurement have dramatically sharpened our understanding of the assembly, connectivity, and evolution of various specialized stress fiber subpopulations. This in turn has motivated interest in understanding how individual stress fibers generate tension and support cellular structure and force generation. In this review, we discuss approaches for measuring the mechanical properties of single stress fibers. We begin by discussing studies conducted in cell-free settings, including strategies based on isolation of intact stress fibers and reconstitution of stress fiber-like structures from purified components. We then discuss measurements obtained in living cells based both on inference of stress fiber properties from whole-cell mechanical measurements (e.g., atomic force microscopy) and on direct interrogation of single stress fibers (e.g., subcellular laser nanosurgery). We conclude by reviewing various mathematical models of stress fiber function that have been developed based on these experimental measurements. An important future challenge in this area will be the integration of these sophisticated biophysical measurements with the field's increasingly detailed molecular understanding of stress fiber assembly, dynamics, and signal transduction. This article is part of a Special Issue entitled: Mechanobiology.
- Published
- 2015
37. Differential Contributions of Nonmuscle Myosin II Isoforms and Functional Domains to Stress Fiber Mechanics.
- Author
-
Chang, Ching-Wei and Kumar, Sanjay
- Subjects
Muscle ,Skeletal ,Stress Fibers ,Humans ,Myosin Type II ,Myosin-Light-Chain Kinase ,Protein Isoforms ,Amino Acid Sequence ,Protein Structure ,Tertiary ,Structure-Activity Relationship ,Viscosity ,Stress ,Mechanical ,Molecular Sequence Data ,Molecular Motor Proteins ,rho-Associated Kinases ,Elastic Modulus ,Generic Health Relevance ,Muscle ,Skeletal ,Protein Structure ,Tertiary ,Stress ,Mechanical ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
While is widely acknowledged that nonmuscle myosin II (NMMII) enables stress fibers (SFs) to generate traction forces against the extracellular matrix, little is known about how specific NMMII isoforms and functional domains contribute to SF mechanics. Here we combine biophotonic and genetic approaches to address these open questions. First, we suppress the NMMII isoforms MIIA and MIIB and apply femtosecond laser nanosurgery to ablate and investigate the viscoelastic retraction of individual SFs. SF retraction dynamics associated with MIIA and MIIB suppression qualitatively phenocopy our earlier measurements in the setting of Rho kinase (ROCK) and myosin light chain kinase (MLCK) inhibition, respectively. Furthermore, fluorescence imaging and photobleaching recovery reveal that MIIA and MIIB are enriched in and more stably localize to ROCK- and MLCK-controlled central and peripheral SFs, respectively. Additional domain-mapping studies surprisingly reveal that deletion of the head domain speeds SF retraction, which we ascribe to reduced drag from actomyosin crosslinking and frictional losses. We propose a model in which ROCK/MIIA and MLCK/MIIB functionally regulate common pools of SFs, with MIIA crosslinking and motor functions jointly contributing to SF retraction dynamics and cellular traction forces.
- Published
- 2015
38. The intrinsic stiffness of human trabecular meshwork cells increases with senescence
- Author
-
Morgan, Joshua T, Raghunathan, Vijay Krishna, Chang, Yow-Ren, Murphy, Christopher J, and Russell, Paul
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Neurodegenerative ,Neurosciences ,Aging ,2.1 Biological and endogenous factors ,Cells ,Cultured ,Cellular Senescence ,Disease Progression ,Glaucoma ,Humans ,Intercellular Signaling Peptides and Proteins ,Membrane Proteins ,Microscopy ,Atomic Force ,Real-Time Polymerase Chain Reaction ,Stress Fibers ,Stress ,Mechanical ,Trabecular Meshwork ,Vimentin ,trabecular meshwork ,senescence ,mechanobiology ,cytoskeleton ,Oncology and carcinogenesis - Abstract
Dysfunction of the human trabecular meshwork (HTM) plays a central role in the age-associated disease glaucoma, a leading cause of irreversible blindness. The etiology remains poorly understood but cellular senescence, increased stiffness of the tissue, and the expression of Wnt antagonists such as secreted frizzled related protein-1 (SFRP1) have been implicated. However, it is not known if senescence is causally linked to either stiffness or SFRP1 expression. In this study, we utilized in vitro HTM senescence to determine the effect on cellular stiffening and SFRP1 expression. Stiffness of cultured cells was measured using atomic force microscopy and the morphology of the cytoskeleton was determined using immunofluorescent analysis. SFRP1 expression was measured using qPCR and immunofluorescent analysis. Senescent cell stiffness increased 1.88±0.14 or 2.57±0.14 fold in the presence or absence of serum, respectively. This was accompanied by increased vimentin expression, stress fiber formation, and SFRP1 expression. In aggregate, these data demonstrate that senescence may be a causal factor in HTM stiffening and elevated SFRP1 expression, and contribute towards disease progression. These findings provide insight into the etiology of glaucoma and, more broadly, suggest a causal link between senescence and altered tissue biomechanics in aging-associated diseases.
- Published
- 2015
39. Exceptional aggressiveness of cerebral cavernous malformation disease associated with PDCD10 mutations
- Author
-
Shenkar, Robert, Shi, Changbin, Rebeiz, Tania, Stockton, Rebecca A, McDonald, David A, Mikati, Abdul Ghani, Zhang, Lingjiao, Austin, Cecilia, Akers, Amy L, Gallione, Carol J, Rorrer, Autumn, Gunel, Murat, Min, Wang, Marcondes de Souza, Jorge, Lee, Connie, Marchuk, Douglas A, and Awad, Issam A
- Subjects
Neurosciences ,Rare Diseases ,Brain Disorders ,Development of treatments and therapeutic interventions ,Aetiology ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine ,Adolescent ,Adult ,Animals ,Apoptosis Regulatory Proteins ,Carrier Proteins ,Cells ,Cultured ,Central Nervous System Neoplasms ,Child ,Child ,Preschool ,Disease Models ,Animal ,Hemangioma ,Cavernous ,Central Nervous System ,Human Umbilical Vein Endothelial Cells ,Humans ,Infant ,Intracellular Signaling Peptides and Proteins ,Keratin-1 ,Membrane Proteins ,Mice ,Middle Aged ,Mutation ,Prospective Studies ,Proto-Oncogene Proteins ,Stress Fibers ,Young Adult ,rho-Associated Kinases ,CCM3 ,cerebral cavernous malformation ,PDCD10 ,Rho kinase ,vascular malformations ,Genetics ,Clinical Sciences ,Genetics & Heredity - Abstract
PurposeThe phenotypic manifestations of cerebral cavernous malformation disease caused by rare PDCD10 mutations have not been systematically examined, and a mechanistic link to Rho kinase-mediated hyperpermeability, a potential therapeutic target, has not been established.MethodsWe analyzed PDCD10 small interfering RNA-treated endothelial cells for stress fibers, Rho kinase activity, and permeability. Rho kinase activity was assessed in cerebral cavernous malformation lesions. Brain permeability and cerebral cavernous malformation lesion burden were quantified, and clinical manifestations were assessed in prospectively enrolled subjects with PDCD10 mutations.ResultsWe determined that PDCD10 protein suppresses endothelial stress fibers, Rho kinase activity, and permeability in vitro. Pdcd10 heterozygous mice have greater lesion burden than other Ccm genotypes. We demonstrated robust Rho kinase activity in murine and human cerebral cavernous malformation vasculature and increased brain vascular permeability in humans with PDCD10 mutation. Clinical phenotype is exceptionally aggressive compared with the more common KRIT1 and CCM2 familial and sporadic cerebral cavernous malformation, with greater lesion burden and more frequent hemorrhages earlier in life. We first report other phenotypic features, including scoliosis, cognitive disability, and skin lesions, unrelated to lesion burden or bleeding.ConclusionThese findings define a unique cerebral cavernous malformation disease with exceptional aggressiveness, and they inform preclinical therapeutic testing, clinical counseling, and the design of trials.Genet Med 17 3, 188-196.
- Published
- 2015
40. Hyaluronidase inhibits TGF-β-mediated rat periodontal ligament fibroblast expression of collagen and myofibroblast markers: An in vitro exploration of periodontal tissue remodeling.
- Author
-
Li, Junlin, Chen, Chen, Zeng, Yunting, Lu, Jiaqi, and Xiao, Liwei
- Subjects
- *
PERIODONTAL ligament , *TISSUE remodeling , *GENE expression , *HYALURONIDASES , *FIBROBLASTS - Abstract
To determine the effect of hyaluronic acid (HA) degradation by hyaluronidase (HYAL) in inhibiting collagen fiber production by rat periodontal ligament cells (rPDLCs). Primary rPDLCs were isolated from the euthanized rats and used for in vitro experiments. The appropriate HYAL concentration was determined through CCK-8 testing for cytotoxicity detection and Alizarin red staining for mineralization detection. RT-qPCR and western blot assays were conducted to assess the effect of HYAL, with or without TGF-β, on generation of collagen fiber constituents and expression of actin alpha 2, smooth muscle (ACTA2) of rPDLCs. Neither cell proliferation nor mineralization were significantly affected by treatment with 4 U/mL HYAL. HYAL (4 U/mL) alone downregulated type I collagen fiber (Col1a1 and Col1a2) and Acta2 mRNA expression; however, ACTA2 and COL1 protein levels were only downregulated by HYAL treatment after TGF-β induction. Treatment of rPDLCs with HYAL can inhibit TGF-β-induced collagen matrix formation and myofibroblast transformation. • We found that 4 U/mL (or lower) has almost no significant effect on cell activity and mineralization function on rPDLCs. • This study found the mechanism of increasing tissue flexibility by hyaluronidase. • Hyaluronidase was safety under non TGF-β-induced conditions, providing a reliable drug for future animal experiments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Substrates with Patterned Extracellular Matrix and Subcellular Stiffness Gradients Reveal Local Biomechanical Responses
- Author
-
Tseng, Peter and Di Carlo, Dino
- Subjects
Bioengineering ,Generic health relevance ,3T3 Cells ,Acrylamide ,Actins ,Animals ,Cell Adhesion ,Cell Culture Techniques ,Cell Shape ,Culture Media ,Cytoskeleton ,Dimethylpolysiloxanes ,Elastic Modulus ,Elasticity ,Epoxy Resins ,Extracellular Matrix ,Fibroblasts ,Heterocyclic Compounds ,4 or More Rings ,Mechanotransduction ,Cellular ,Mice ,Microtechnology ,Myosin Type II ,mechanotransduction ,BioMEMS ,cell control ,biomechanics ,stress fibers ,Physical Sciences ,Chemical Sciences ,Engineering ,Nanoscience & Nanotechnology - Abstract
A substrate fabrication process is developed to pattern both the extracellular matrix (ECM) and rigidity at sub-cellular spatial resolution. When growing cells on these substrates, it is found that cells respond locally in their cytoskeleton assembly. The presented method allows unique insight into the biological interpretation of mechanical signals, whereas photolithography-based fabrication is amenable to integration with complex microfabricated substructures.
- Published
- 2014
42. Biomechanics and Modeling of Tissue-Engineered Heart Valves
- Author
-
Ristori, T., van Kelle, A. J., Baaijens, F. P. T., Loerakker, S., Sacks, Michael S., editor, and Liao, Jun, editor
- Published
- 2018
- Full Text
- View/download PDF
43. Mechanosensitive myosin II but not cofilin primarily contributes to cyclic cell stretch-induced selective disassembly of actin stress fibers.
- Author
-
Wenjing Huang, Matsui, Tsubasa S., Takumi Saito, Masahiro Kuragano, Masayuki Takahashi, Tomohiro Kawahara, Masaaki Sato, and Shinji Deguchi
- Subjects
- *
VASCULAR smooth muscle , *MYOSIN , *ACTIN , *MUSCLE cells , *ACTOMYOSIN , *FIBERS - Abstract
Cells adapt to applied cyclic stretch (CS) to circumvent chronic activation of proinflammatory signaling. Currently, the molecular mechanism of the selective disassembly of actin stress fibers (SFs) in the stretch direction, which occurs at the early stage of the cellular response to CS, remains controversial. Here, we suggest that the mechanosensitive behavior of myosin II, a major crosslinker of SFs, primarily contributes to the directional disassembly of the actomyosin complex SFs in bovine vascular smooth muscle cells and human U2OS osteosarcoma cells. First, we identified that CS with a shortening phase that exceeds in speed the inherent contractile rate of individual SFs leads to the disassembly. To understand the biological basis, we investigated the effect of expressing myosin regulatory light-chain mutants and found that SFs with less actomyosin activities disassemble more promptly upon CS. We consequently created a minimal mathematical model that recapitulates the salient features of the direction-selective and threshold-triggered disassembly of SFs to show that disassembly or, more specifically, unbundling of the actomyosin bundle SFs is enhanced with sufficiently fast cell shortening. We further demonstrated that similar disassembly of SFs is inducible in the presence of an active LIM-kinase-1 mutant that deactivates cofilin, suggesting that cofilin is dispensable as opposed to a previously proposed mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
44. Tumor Suppressors TSC1 and TSC2 Differentially Modulate Actin Cytoskeleton and Motility of Mouse Embryonic Fibroblasts
- Author
-
Goncharova, Elena A, James, Melane L, Kudryashova, Tatiana V, Goncharov, Dmitry A, and Krymskaya, Vera P
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Brain Disorders ,Tuberous Sclerosis ,Rare Diseases ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Actin Cytoskeleton ,Animals ,Carrier Proteins ,Cell Line ,Cell Movement ,Embryo ,Mammalian ,Fibroblasts ,Gene Knockout Techniques ,Mechanistic Target of Rapamycin Complex 1 ,Mechanistic Target of Rapamycin Complex 2 ,Mice ,Multiprotein Complexes ,RNA ,Small Interfering ,Rapamycin-Insensitive Companion of mTOR Protein ,Stress Fibers ,TOR Serine-Threonine Kinases ,Tuberous Sclerosis Complex 1 Protein ,Tuberous Sclerosis Complex 2 Protein ,Tumor Suppressor Proteins ,General Science & Technology - Abstract
TSC1 and TSC2 mutations cause neoplasms in rare disease pulmonary LAM and neuronal pathfinding in hamartoma syndrome TSC. The specific roles of TSC1 and TSC2 in actin remodeling and the modulation of cell motility, however, are not well understood. Previously, we demonstrated that TSC1 and TSC2 regulate the activity of small GTPases RhoA and Rac1, stress fiber formation and cell adhesion in a reciprocal manner. Here, we show that Tsc1(-/-) MEFs have decreased migration compared to littermate-derived Tsc1(+/+) MEFs. Migration of Tsc1(-/-) MEFs with re-expressed TSC1 was comparable to Tsc1(+/+) MEF migration. In contrast, Tsc2(-/-) MEFs showed an increased migration compared to Tsc2(+/+) MEFs that were abrogated by TSC2 re-expression. Depletion of TSC1 and TSC2 using specific siRNAs in wild type MEFs and NIH 3T3 fibroblasts also showed that TSC1 loss attenuates cell migration while TSC2 loss promotes cell migration. Morphological and immunochemical analysis demonstrated that Tsc1(-/-) MEFs have a thin protracted shape with a few stress fibers; in contrast, Tsc2(-/-) MEFs showed a rounded morphology and abundant stress fibers. Expression of TSC1 in either Tsc1(-/-) or Tsc2(-/-) MEFs promoted stress fiber formation, while TSC2 re-expression induced stress fiber disassembly and the formation of cortical actin. To assess the mechanism(s) by which TSC2 loss promotes actin re-arrangement and cell migration, we explored the role of known downstream effectors of TSC2, mTORC1 and mTORC2. Increased migration of Tsc2(-/-) MEFs is inhibited by siRNA mTOR and siRNA Rictor, but not siRNA Raptor. siRNA mTOR or siRNA Rictor promoted stress fiber disassembly in TSC2-null cells, while siRNA Raptor had little effect. Overexpression of kinase-dead mTOR induced actin stress fiber disassembly and suppressed TSC2-deficient cell migration. Our data demonstrate that TSC1 and TSC2 differentially regulate actin stress fiber formation and cell migration, and that only TSC2 loss promotes mTOR- and mTORC2-dependent pro-migratory cell phenotype.
- Published
- 2014
45. A nonlinear elastic description of cell preferential orientations over a stretched substrate.
- Author
-
Lucci, Giulio and Preziosi, Luigi
- Subjects
- *
STRAIN tensors , *TISSUE engineering , *NONLINEAR equations , *ELASTICITY - Abstract
The active response of cells to mechanical cues due to their interaction with the environment has been of increasing interest, since it is involved in many physiological phenomena, pathologies, and in tissue engineering. In particular, several experiments have shown that, if a substrate with overlying cells is cyclically stretched, they will reorient to reach a well-defined angle between their major axis and the main stretching direction. Recent experimental findings, also supported by a linear elastic model, indicated that the minimization of an elastic energy might drive this reorientation process. Motivated by the fact that a similar behaviour is observed even for high strains, in this paper we address the problem in the framework of finite elasticity, in order to study the presence of nonlinear effects. We find that, for a very large class of constitutive orthotropic models and with very general assumptions, there is a single linear relationship between a parameter describing the biaxial deformation and cos 2 θ eq , where θ eq is the orientation angle of the cell, with the slope of the line depending on a specific combination of four parameters that characterize the nonlinear constitutive equation. We also study the effect of introducing a further dependence of the energy on the anisotropic invariants related to the square of the Cauchy–Green strain tensor. This leads to departures from the linear relationship mentioned above, that are again critically compared with experimental data. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
46. What factors determine the number of nonmuscle myosin II in the sarcomeric unit of stress fibers?
- Author
-
Saito, Takumi, Huang, Wenjing, Matsui, Tsubasa S., Kuragano, Masahiro, Takahashi, Masayuki, and Deguchi, Shinji
- Subjects
- *
MYOSIN , *STRAINS & stresses (Mechanics) , *FIBERS , *MYOFIBRILS , *ACTIN - Abstract
Actin stress fibers (SFs), a contractile apparatus in nonmuscle cells, possess a contractile unit that is apparently similar to the sarcomere of myofibrils in muscles. The function of SFs has thus often been addressed based on well-characterized properties of muscles. However, unlike the fixed number of myosin molecules in myofibrils, the number of nonmuscle myosin II (NMII) within the contractile sarcomeric unit in SFs is quite low and variable for some reason. Here we address what factors may determine the specific number of NMII in SFs. We suggest with a theoretical model that the number lies just in between the function of SFs for bearing cellular tension under static conditions and for promptly disintegrating upon forced cell shortening. We monitored shortening-induced disintegration of SFs in human osteosarcoma U2OS cells expressing mutants of myosin regulatory light chain that virtually regulates the interaction of NMII with actin filaments, and the behaviors observed were indeed consistent with the theoretical consequences. This situation-specific nature of SFs may allow nonmuscle cells to respond adaptively to mechanical stress to circumvent activation of pro-inflammatory signals as previously indicated, i.e., a behavior distinct from that of muscles that are basically specialized for exhibiting contractile activity. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
47. Characterization of the endothelial cell cytoskeleton following HLA class I ligation.
- Author
-
Ziegler, Mary E, Souda, Puneet, Jin, Yi-Ping, Whitelegge, Julian P, and Reed, Elaine F
- Subjects
Aorta ,Endothelium ,Vascular ,Cells ,Cultured ,Cytoskeleton ,Stress Fibers ,Humans ,Actins ,Immunoglobulin G ,Histocompatibility Antigens Class I ,Blotting ,Western ,Chromatography ,Liquid ,Proteomics ,Paxillin ,Tandem Mass Spectrometry ,Biomarkers ,Endothelium ,Vascular ,Cells ,Cultured ,Blotting ,Western ,Chromatography ,Liquid ,General Science & Technology - Abstract
BackgroundVascular endothelial cells (ECs) are a target of antibody-mediated allograft rejection. In vitro, when the HLA class I molecules on the surface of ECs are ligated by anti-HLA class I antibodies, cell proliferation and survival pathways are activated and this is thought to contribute to the development of antibody-mediated rejection. Crosslinking of HLA class I molecules by anti-HLA antibodies also triggers reorganization of the cytoskeleton, which induces the formation of F-actin stress fibers. HLA class I induced stress fiber formation is not well understood.Methodology and principal findingsThe present study examines the protein composition of the cytoskeleton fraction of ECs treated with HLA class I antibodies and compares it to other agonists known to induce alterations of the cytoskeleton in endothelial cells. Analysis by tandem mass spectrometry revealed unique cytoskeleton proteomes for each treatment group. Using annotation tools a candidate list was created that revealed 12 proteins, which were unique to the HLA class I stimulated group. Eleven of the candidate proteins were phosphoproteins and exploration of their predicted kinases provided clues as to how these proteins may contribute to the understanding of HLA class I induced antibody-mediated rejection. Three of the candidates, eukaryotic initiation factor 4A1 (eIF4A1), Tropomyosin alpha 4-chain (TPM4) and DDX3X, were further characterized by Western blot and found to be associated with the cytoskeleton. Confocal microscopy analysis showed that class I ligation stimulated increased eIF4A1 co-localization with F-actin and paxillin.Conclusions/significanceColocalization of eIF4A1 with F-actin and paxillin following HLA class I ligation suggests that this candidate protein could be a target for understanding the mechanism(s) of class I mediated antibody-mediated rejection. This proteomic approach for analyzing the cytoskeleton of ECs can be applied to other agonists and various cells types as a method for uncovering novel regulators of cytoskeleton changes.
- Published
- 2012
48. Concentration Independent Modulation of Local Micromechanics in a Fibrin Gel
- Author
-
Kotlarchyk, Maxwell A., Shreim, Samir G., Alvarez-Elizondo, Martha B., Estrada, Laura C., Singh, Rahul, Valdevit, Lorenzo, Kniazeva, Ekaterina, Gratton, Enrico, Putnam, Andrew J., and Botvinick, Elliot L.
- Subjects
extracellular-matrix mimics ,mechanical-properties ,endothelial-cells ,particle tracking ,stress fibers ,mesh size ,hydrogels ,elasticity ,migration ,behavior - Abstract
Methods for tuning extracellular matrix (ECM) mechanics in 3D cell culture that rely on increasing the concentration of either protein or cross-linking molecules fail to control important parameters such as pore size, ligand density, and molecular diffusivity. Alternatively, ECM stiffness can be modulated independently from protein concentration by mechanically loading the ECM. We have developed a novel device for generating stiffness gradients in naturally derived ECMs, where stiffness is tuned by inducing strain, while local mechanical properties are directly determined by laser tweezers based active microrheology (AMR). Hydrogel substrates polymerized within 35 mm diameter Petri dishes are strained non-uniformly by the precise rotation of an embedded cylindrical post, and exhibit a position-dependent stiffness with little to no modulation of local mesh geometry. Here we present the device in the context of fibrin hydrogels. First AMR is used to directly measure local micromechanics in unstrained hydrogels of increasing fibrin concentration. Changes in stiffness are then mapped within our device, where fibrin concentration is held constant. Fluorescence confocal imaging and orbital particle tracking are used to quantify structural changes in fibrin on the micro and nano levels respectively. The micromechanical strain stiffening measured by microrheology is not accompanied by ECM microstructural changes under our applied loads, as measured by confocal microscopy. However, super-resolution orbital tracking reveals nanostructural straightening, lengthening, and reduced movement of fibrin fibers. Furthermore, we show that aortic smooth muscle cells cultured within our device are morphologically sensitive to the induced mechanical gradient. Our results demonstrate a powerful cell culture tool that can be used in the study of mechanical effects on cellular physiology in naturally derived 3D ECM tissues.
- Published
- 2011
49. Cellular Contact Guidance Emerges from Gap Avoidance
- Author
-
Antonetta B.C. Buskermolen, Tommaso Ristori, Dylan Mostert, Mark C. van Turnhout, Siamak S. Shishvan, Sandra Loerakker, Nicholas A. Kurniawan, Vikram S. Deshpande, and Carlijn V.C. Bouten
- Subjects
cell alignment ,microcontact printing ,statistical mechanics ,cell adhesion ,stress fibers ,substrate anisotropy ,Physics ,QC1-999 - Abstract
Summary: In the presence of anisotropic biochemical or topographical patterns, cells tend to align in the direction of these cues—a widely reported phenomenon known as “contact guidance.” To investigate the origins of contact guidance, here, we created substrates micropatterned with parallel lines of fibronectin with dimensions spanning multiple orders of magnitude. Quantitative morphometric analysis of our experimental data reveals two regimes of contact guidance governed by the length scale of the cues that cannot be explained by enforced alignment of focal adhesions. Adopting computational simulations of cell remodeling on inhomogeneous substrates based on a statistical mechanics framework for living cells, we show that contact guidance emerges from anisotropic cell shape fluctuation and “gap avoidance,” i.e., the energetic penalty of cell adhesions on non-adhesive gaps. Our findings therefore point to general biophysical mechanisms underlying cellular contact guidance, without the necessity of invoking specific molecular pathways.
- Published
- 2020
- Full Text
- View/download PDF
50. The eukaryotic translation elongation factor 1A regulation of actin stress fibers is important for infectious RSV production
- Author
-
Natale Snape, Dongsheng Li, Ting Wei, Hongping Jin, Mary Lor, Daniel J. Rawle, Kirsten M. Spann, and David Harrich
- Subjects
Respiratory syncytial virus ,Eukaryotic translation elongation factor 1A ,Didemnin B ,Stress fibers ,Virus replication ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Cellular protein eukaryotic translation elongation factor 1A (eEF1A) is an actin binding protein that plays a role in the formation of filamentous actin (F-actin) bundles. F-Actin regulates multiple stages of respiratory syncytial virus (RSV) replication including assembly and budding. Our previous study demonstrated that eEF1A knock-down significantly reduced RSV replication. Here we investigated if the eEF1A function in actin bundle formation was important for RSV replication and release. To investigate this, eEF1A function was impaired in HEp-2 cells by either knock-down of eEF1A with siRNA, or treatment with an eEF1A inhibitor, didemnin B (Did B). Cell staining and confocal microscopy analysis showed that both eEF1A knock-down and treatment with Did B resulted in disruption of cellular stress fiber formation and elevated accumulation of F-actin near the plasma membrane. When treated cells were then infected with RSV, there was also reduced formation of virus-induced cellular filopodia. Did B treatment, similarly to eEF1A knock-down, reduced the release of infectious RSV, but unlike eEF1A knock-down, did not significantly affect RSV genome replication. The lower infectious virus production in Did B treated cells also reduced RSV-induced cell death. In conclusion, the cellular factor eEF1A plays an important role in the regulation of F-actin stress fiber formation required for RSV assembly and release.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.