Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Bioessays ; 46(8): e2400034, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38798157

ABSTRACT

Sad1 and UNC84 (SUN) and Klarsicht, ANC-1, and Syne homology (KASH) proteins interact at the nuclear periphery to form the linker of nucleoskeleton and cytoskeleton (LINC) complex, spanning the nuclear envelope (NE) and connecting the cytoskeleton with the nuclear interior. It is now well-documented that several cellular functions depend on LINC complex formation, including cell differentiation and migration. Intriguingly, recent studies suggest that SUN proteins participate in cellular processes where their association with KASH proteins may not be required. Building on this recent research, we elaborate on the hypothesis that SUN proteins may perform LINC-independent functions and discuss the modalities that may allow SUN proteins to function at the INM when they are not forming LINC complex.


Subject(s)
Cytoskeleton , Nuclear Envelope , Nuclear Matrix , Nuclear Proteins , Humans , Cytoskeleton/metabolism , Animals , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Matrix/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 119(45): e2116167119, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36322767

ABSTRACT

How cells adjust their growth to the spatial and mechanical constraints of their surrounding environment is central to many aspects of biology. Here, we examined how extracellular matrix (ECM) rigidity affects cell division. We found that cells divide more rapidly when cultured on rigid substrates. While we observed no effect of ECM rigidity on rounding or postmitotic spreading duration, we found that changes in matrix stiffness impact mitosis progression. We noticed that ECM elasticity up-regulates the expression of the linker of nucleoskeleton and cytoskeleton (LINC) complex component SUN2, which in turn promotes metaphase-to-anaphase transition by acting on mitotic spindle formation, whereas when cells adhere to soft ECM, low levels of SUN2 expression perturb astral microtubule organization and delay the onset of anaphase.


Subject(s)
Cytoskeleton , Nuclear Matrix , Nuclear Matrix/metabolism , Cytoskeleton/metabolism , Microtubules/metabolism , Mitosis , Extracellular Matrix , Spindle Apparatus , Anaphase
3.
EMBO Rep ; 20(9): e48084, 2019 09.
Article in English | MEDLINE | ID: mdl-31368207

ABSTRACT

The shape of the cell nucleus can vary considerably during developmental and pathological processes; however, the impact of nuclear morphology on cell behavior is not known. Here, we observed that the nuclear envelope flattens as cells transit from G1 to S phase and inhibition of myosin II prevents nuclear flattening and impedes progression to S phase. Strikingly, we show that applying compressive force on the nucleus in the absence of myosin II-mediated tension is sufficient to restore G1 to S transition. Using a combination of tools to manipulate nuclear morphology, we observed that nuclear flattening activates a subset of transcription factors, including TEAD and AP1, leading to transcriptional induction of target genes that promote G1 to S transition. In addition, we found that nuclear flattening mediates TEAD and AP1 activation in response to ROCK-generated contractility or cell spreading. Our results reveal that the nuclear envelope can operate as a mechanical sensor whose deformation controls cell growth in response to tension.


Subject(s)
Cell Nucleus/metabolism , Mechanotransduction, Cellular/physiology , Nuclear Envelope/metabolism , Transcription Factors/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Division/genetics , Cell Division/physiology , Cell Line , Cell Nucleus/genetics , Flow Cytometry , G1 Phase/genetics , G1 Phase/physiology , HeLa Cells , Humans , Mechanotransduction, Cellular/genetics , Microscopy, Atomic Force , Nuclear Envelope/genetics , Plasmids/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S Phase/genetics , S Phase/physiology , Transcription Factors/genetics
4.
Eur Heart J ; 40(37): 3081-3094, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31114854

ABSTRACT

AIMS: The Brugada syndrome (BrS) is an inherited cardiac disorder predisposing to ventricular arrhythmias. Despite considerable efforts, its genetic basis and cellular mechanisms remain largely unknown. The objective of this study was to identify a new susceptibility gene for BrS through familial investigation. METHODS AND RESULTS: Whole-exome sequencing performed in a three-generation pedigree with five affected members allowed the identification of one rare non-synonymous substitution (p.R211H) in RRAD, the gene encoding the RAD GTPase, carried by all affected members of the family. Three additional rare missense variants were found in 3/186 unrelated index cases. We detected higher levels of RRAD transcripts in subepicardium than in subendocardium in human heart, and in the right ventricle outflow tract compared to the other cardiac compartments in mice. The p.R211H variant was then subjected to electrophysiological and structural investigations in human cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs). Cardiomyocytes derived from induced pluripotent stem cells from two affected family members exhibited reduced action potential upstroke velocity, prolonged action potentials and increased incidence of early afterdepolarizations, with decreased Na+ peak current amplitude and increased Na+ persistent current amplitude, as well as abnormal distribution of actin and less focal adhesions, compared with intra-familial control iPSC-CMs Insertion of p.R211H-RRAD variant in control iPSCs by genome editing confirmed these results. In addition, iPSC-CMs from affected patients exhibited a decreased L-type Ca2+ current amplitude. CONCLUSION: This study identified a potential new BrS-susceptibility gene, RRAD. Cardiomyocytes derived from induced pluripotent stem cells expressing RRAD variant recapitulated single-cell electrophysiological features of BrS, including altered Na+ current, as well as cytoskeleton disturbances.


Subject(s)
Brugada Syndrome/genetics , Mutation, Missense , Myocytes, Cardiac/pathology , ras Proteins/genetics , Action Potentials/genetics , Adult , Brugada Syndrome/pathology , Brugada Syndrome/physiopathology , Cytoskeleton/genetics , Cytoskeleton/pathology , Female , Genetic Markers , Genetic Predisposition to Disease , Humans , Male , Myocytes, Cardiac/physiology
5.
Exp Cell Res ; 343(1): 14-20, 2016 04 10.
Article in English | MEDLINE | ID: mdl-26519907

ABSTRACT

Stress fibers and focal adhesions are complex protein arrays that produce, transmit and sense mechanical tension. Evidence accumulated over many years led to the conclusion that mechanical tension generated within stress fibers contributes to the assembly of both stress fibers themselves and their associated focal adhesions. However, several lines of evidence have recently been presented against this model. Here we discuss the evidence for and against the role of mechanical tension in driving the assembly of these structures. We also consider how their assembly is influenced by the rigidity of the substratum to which cells are adhering. Finally, we discuss the recently identified connections between stress fibers and the nucleus, and the roles that these may play, both in cell migration and regulating nuclear function.


Subject(s)
Focal Adhesions/physiology , Stress Fibers/physiology , Stress, Mechanical , Animals , Humans , Models, Biological
6.
Nat Mater ; 19(6): 587-588, 2020 06.
Article in English | MEDLINE | ID: mdl-32461681
7.
J Immunol ; 192(7): 3390-8, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24585879

ABSTRACT

RhoA-mediated cytoskeletal rearrangements in endothelial cells (ECs) play an active role in leukocyte transendothelial cell migration (TEM), a normal physiological process in which leukocytes cross the endothelium to enter the underlying tissue. Although much has been learned about RhoA signaling pathways downstream from ICAM-1 in ECs, little is known about the consequences of the tractional forces that leukocytes generate on ECs as they migrate over the surface before TEM. We have found that after applying mechanical forces to ICAM-1 clusters, there is an increase in cellular stiffening and enhanced RhoA signaling compared with ICAM-1 clustering alone. We have identified that leukemia-associated Rho guanine nucleotide exchange factor (LARG), also known as Rho GEF 12 (ARHGEF12) acts downstream of clustered ICAM-1 to increase RhoA activity, and that this pathway is further enhanced by mechanical force on ICAM-1. Depletion of LARG decreases leukocyte crawling and inhibits TEM. To our knowledge, this is the first report of endothelial LARG regulating leukocyte behavior and EC stiffening in response to tractional forces generated by leukocytes.


Subject(s)
Endothelial Cells/immunology , Intercellular Adhesion Molecule-1/immunology , Mechanotransduction, Cellular/immunology , Rho Guanine Nucleotide Exchange Factors/immunology , Transendothelial and Transepithelial Migration/immunology , Blotting, Western , Cells, Cultured , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/immunology , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infant, Newborn , Intercellular Adhesion Molecule-1/metabolism , Microscopy, Fluorescence , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA Interference , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/immunology , Stress, Mechanical
8.
Stem Cells ; 31(11): 2528-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23836527

ABSTRACT

Mechanical strain provides an anti-adipogenic, pro-osteogenic stimulus to mesenchymal stem cells (MSC) through generating intracellular signals and via cytoskeletal restructuring. Recently, mTORC2 has been shown to be a novel mechanical target critical for the anti-adipogenic signal leading to preservation of ß-catenin. As mechanical activation of mTORC2 requires focal adhesions (FAs), we asked whether proximal signaling involved Src and FAK, which are early responders to integrin-FA engagement. Application of mechanical strain to marrow-derived MSCs was unable to activate mTORC2 when Src family kinases were inhibited. Fyn, but not Src, was specifically required for mechanical activation of mTORC2 and was recruited to FAs after strain. Activation of mTORC2 was further diminished following FAK inhibition, and as FAK phosphorylation (Tyr-397) required Fyn activity, provided evidence of Fyn/FAK cooperativity. Inhibition of Fyn also prevented mechanical activation of RhoA as well as mechanically induced actin stress fiber formation. We thus asked whether RhoA activation by strain was dependent on mTORC2 downstream of Fyn. Inhibition of mTORC2 or its downstream substrate, Akt, both prevented mechanical RhoA activation, indicating that Fyn/FAK affects cytoskeletal structure via mTORC2. We then sought to ascertain whether this Fyn-initiated signal pathway modulated MSC lineage decisions. siRNA knockdown of Fyn, but not Src, led to rapid attainment of adipogenic phenotype with significant increases in adipocyte protein 2, peroxisome proliferator-activated receptor gamma, adiponectin, and perilipin. As such, Fyn expression in mdMSCs contributes to basal cytoskeletal architecture and, when associated with FAs, functions as a proximal mechanical effector for environmental signals that influence MSC lineage allocation.


Subject(s)
Adipogenesis/physiology , Mesenchymal Stem Cells/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , TOR Serine-Threonine Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Culture Techniques , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 2 , Mesenchymal Stem Cells/cytology , Multiprotein Complexes/genetics , Phosphorylation , Proto-Oncogene Proteins c-fyn/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Transfection
9.
Blood ; 118(19): 5344-54, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21881052

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) is associated with 3 different human malignancies: Kaposi sarcoma (KS), primary effusion lymphoma, and multicentric Castleman disease. The KS lesion is driven by KSHV-infected endothelial cells and is highly dependent on autocrine and paracrine factors for survival and growth. We report that latent KSHV infection increases the vascular permeability of endothelial cells. Endothelial cells with latent KSHV infection display increased Rac1 activation and activation of its downstream modulator, p21-activated kinase 1 (PAK1). The KSHV-infected cells also exhibit increases in tyrosine phosphorylation of vascular endothelial (VE)-cadherin and ß-catenin, whereas total levels of these proteins remained unchanged, suggesting that latent infection disrupted endothelial cell junctions. Consistent with these findings, we found that KSHV-infected endothelial cells displayed increased permeability compared with uninfected endothelial cells. Knockdown of Rac1 and inhibition of reactive oxygen species (ROS) resulted in decreased permeability in the KSHV-infected endothelial cells. We further demonstrate that the KSHV K1 protein can activate Rac1. Rac1 was also highly activated in KSHV-infected endothelial cells and KS tumors. In conclusion, KSHV latent infection increases Rac1 and PAK1 activity in endothelial cells, resulting in the phosphorylation of VE-cadherin and ß-catenin and leading to the disassembly of cell junctions and to increased vascular permeability of the infected endothelial cells.


Subject(s)
Capillary Permeability , Herpesviridae Infections/physiopathology , Herpesvirus 8, Human/pathogenicity , Antigens, CD/metabolism , Base Sequence , Cadherins/metabolism , Endothelial Cells/physiology , Enzyme Activation , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Phosphorylation , RNA, Small Interfering/genetics , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , beta Catenin/metabolism , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology
10.
Elife ; 122023 03 29.
Article in English | MEDLINE | ID: mdl-36989130

ABSTRACT

Endothelial cells line all blood vessels, where they coordinate blood vessel formation and the blood-tissue barrier via regulation of cell-cell junctions. The nucleus also regulates endothelial cell behaviors, but it is unclear how the nucleus contributes to endothelial cell activities at the cell periphery. Here, we show that the nuclear-localized linker of the nucleoskeleton and cytoskeleton (LINC) complex protein SUN1 regulates vascular sprouting and endothelial cell-cell junction morphology and function. Loss of murine endothelial Sun1 impaired blood vessel formation and destabilized junctions, angiogenic sprouts formed but retracted in SUN1-depleted sprouts, and zebrafish vessels lacking Sun1b had aberrant junctions and defective cell-cell connections. At the cellular level, SUN1 stabilized endothelial cell-cell junctions, promoted junction function, and regulated contractility. Mechanistically, SUN1 depletion altered cell behaviors via the cytoskeleton without changing transcriptional profiles. Reduced peripheral microtubule density, fewer junction contacts, and increased catastrophes accompanied SUN1 loss, and microtubule depolymerization phenocopied effects on junctions. Depletion of GEF-H1, a microtubule-regulated Rho activator, or the LINC complex protein nesprin-1 rescued defective junctions of SUN1-depleted endothelial cells. Thus, endothelial SUN1 regulates peripheral cell-cell junctions from the nucleus via LINC complex-based microtubule interactions that affect peripheral microtubule dynamics and Rho-regulated contractility, and this long-range regulation is important for proper blood vessel sprouting and junction integrity.


Subject(s)
Endothelial Cells , Microtubule-Associated Proteins , Animals , Mice , Microtubule-Associated Proteins/metabolism , Endothelial Cells/metabolism , Zebrafish/metabolism , Nuclear Proteins/metabolism , Microtubules/metabolism , Intercellular Junctions/metabolism
11.
Biochemistry ; 51(38): 7420-32, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22931484

ABSTRACT

Throughout their lives, all cells constantly experience and respond to various mechanical forces. These frequently originate externally but can also arise internally as a result of the contractile actin cytoskeleton. Mechanical forces trigger multiple signaling pathways. Several converge and result in the activation of the GTPase RhoA. In this review, we focus on the pathways by which mechanical force leads to RhoA regulation, especially when force is transmitted via cell adhesion molecules that mediate either cell-matrix or cell-cell interactions. We discuss both the upstream signaling events that lead to activation of RhoA and the downstream consequences of this pathway. These include not only cytoskeletal reorganization and, in a positive feedback loop, increased myosin-generated contraction but also profound effects on gene expression and differentiation.


Subject(s)
rhoA GTP-Binding Protein/chemistry , Cell Adhesion Molecules/chemistry , Extracellular Matrix/chemistry , Focal Adhesions , Gene Expression , Humans , Neoplasms/pathology
12.
J Biol Chem ; 286(52): 45103-15, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22052910

ABSTRACT

Vinculin is an essential and highly conserved cell adhesion protein, found at both focal adhesions and adherens junctions, where it couples integrins or cadherins to the actin cytoskeleton. Vinculin is involved in controlling cell shape, motility, and cell survival, and has more recently been shown to play a role in force transduction. The tail domain of vinculin (Vt) contains determinants necessary for binding and bundling of actin filaments. Actin binding to Vt has been proposed to induce formation of a Vt dimer that is necessary for cross-linking actin filaments. Results from this study provide additional support for actin-induced Vt self-association. Moreover, the actin-induced Vt dimer appears distinct from the dimer formed in the absence of actin. To better characterize the role of the Vt strap and carboxyl terminus (CT) in actin binding, Vt self-association, and actin bundling, we employed smaller amino-terminal (NT) and CT deletions that do not perturb the structural integrity of Vt. Although both NT and CT deletions retain actin binding, removal of the CT hairpin (1061-1066) selectively impairs actin bundling in vitro. Moreover, expression of vinculin lacking the CT hairpin in vinculin knock-out murine embryonic fibroblasts affects the number of focal adhesions formed, cell spreading as well as cellular stiffening in response to mechanical force.


Subject(s)
Actins/metabolism , Avian Proteins/metabolism , Focal Adhesions/metabolism , Protein Multimerization/physiology , Vinculin/metabolism , Actins/genetics , Animals , Avian Proteins/genetics , Cells, Cultured , Chickens , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesions/genetics , Mice , Mice, Knockout , Protein Binding , Protein Structure, Tertiary , Vinculin/genetics
13.
Stem Cells ; 29(11): 1829-36, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21898699

ABSTRACT

The fate of pluripotent mesenchymal stem cells (MSC) is determined through integration of chemical, spatial, and physical signals. The suppression of MSC adipogenesis by mechanical stimuli, which requires Akt-induced inhibition of glycogen synthase kinase 3ß (GSK3ß) with ß-catenin activation, can be enhanced by repetitive dosing within a single day. Here, we demonstrate that reapplication of cyclic strain within a 24-hour period leads to amplification of both Akt activation and its subsequent inhibition of GSK3ß, such that total cycle number can be reduced while still inhibiting adipogenesis. Amplification of Akt signaling is facilitated by a dynamic restructuring of the cell in response to mechanical signals, as evidenced by a transient increase in focal adhesion (FA) number and increased RhoA activity. Preventing FA assembly or development of tension blocks activation of Akt by mechanical signals, but not by insulin. This indicates that the FA infrastructure is essential to the physical, but not necessarily the chemical, sensitivity, and responsiveness of the cell. Exploiting the transient nature of cytoskeletal remodeling may represent a process to enhance cell responsiveness to mechanical input and ultimately define the fate of MSCs with a minimal input.


Subject(s)
Adipocytes/cytology , Focal Adhesions/metabolism , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Stress, Mechanical , Adipocytes/metabolism , Animals , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mice , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
14.
Arterioscler Thromb Vasc Biol ; 31(11): 2634-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21852563

ABSTRACT

OBJECTIVE: Estradiol (E2) mediates numerous beneficial effects assigned to estrogens, but whereas mechanisms have been described at the endothelial level, direct effects on vascular smooth muscle cells (VSMC) are poorly documented. As evidence accumulates regarding the role of RhoA in vascular pathophysiology and the benefit of RhoA-Rho associated protein kinase (Rock) pathway inhibition, we analyzed if E2 could inhibit it in VSMC. METHODS AND RESULTS: We show that in VSMC, E2 inhibits the RhoA-Rock pathway in a time- and concentration-dependent manner. The inhibition of RhoA-Rock pathway results from E2-induced phosphorylation of the Ser188 of RhoA. Using pharmacological, transfection, and in vitro phosphorylation experiments, we demonstrate that AMP-activated protein kinase subunit alpha 1 (AMPKα1) is activated by estrogen receptor stimulation and catalyzes RhoA phosphorylation induced by E2. Ex vivo, ovariectomy leads to an increase in the amplitude of phenylephrine- or serotonine-induced contractions of aortic rings in wild-type mice but not in AMPKα1-knock-out mice or E2-supplemented animals. These functional effects were correlated with a reduced level of RhoA phosphorylation in the aorta of ovariectomized female, male, and AMPKα1 knock-out mice. CONCLUSION: Our work thus defines AMPKα1 as (1) a new kinase for RhoA and (2) a new mediator of the vasoprotective effects of estrogen.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Estradiol/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , rho GTP-Binding Proteins/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Knockout , Models, Animal , Muscle, Smooth, Vascular/cytology , Ovariectomy , Phosphorylation/drug effects , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Time Factors , Vasoconstriction/physiology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein
15.
Mol Biol Cell ; 32(16): 1409-1416, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34133212

ABSTRACT

A central challenge to the biology of development and disease is deciphering how individual cells process and respond to numerous biochemical and mechanical signals originating from the environment. Recent advances in genomic studies enabled the acquisition of information about population heterogeneity; however, these so far are poorly linked with the spatial heterogeneity of biochemical and mechanical cues. Whereas in vitro models offer superior control over spatiotemporal distribution of numerous mechanical parameters, researchers are limited by the lack of methods to select subpopulations of cells in order to understand how environmental heterogeneity directs the functional collective response. To circumvent these limitations, we present a method based on the use of photo convertible proteins, which when expressed within cells and activated with light, gives a stable fluorescence fingerprint enabling subsequent sorting and lysis for genomics analysis. Using this technique, we study the spatial distribution of genetic alterations on well-characterized local mechanical stimulation within the epithelial monolayer. Our method is an in vitro alternative to laser microdissection, which so far has found a broad application in ex vivo studies.


Subject(s)
Cytophotometry/methods , Genomics , Animals , Dogs , Flow Cytometry , Fluorescence , Gene Expression Profiling , Humans , Microfluidic Analytical Techniques , Sequence Analysis, RNA
16.
Dev Cell ; 56(23): 3222-3234.e6, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34875225

ABSTRACT

Cell shape dynamics during development is tightly regulated and coordinated with cell fate determination. Triggered by an interplay between biochemical and mechanical signals, epithelia form complex tissues by undergoing coordinated cell shape changes, but how such spatiotemporal coordination is controlled remains an open question. To dissect biochemical signaling from purely mechanical cues, we developed a microfluidic system that experimentally triggers epithelial folding to recapitulate stereotypic deformations observed in vivo. Using this system, we observe that the apical or basal direction of folding results in strikingly different mechanical states at the fold boundary, where the balance between tissue tension and torque (arising from the imposed curvature) controls the spread of folding-induced calcium waves at a short timescale and induces spatial patterns of gene expression at longer timescales. Our work uncovers that folding-associated gradients of cell shape and their resulting mechanical stresses direct spatially distinct biochemical responses within the monolayer.


Subject(s)
Cell Shape , Elasticity , Epithelial Cells/chemistry , Models, Biological , Stress, Mechanical , Animals , Biomechanical Phenomena , Dogs , Madin Darby Canine Kidney Cells
17.
Circ Res ; 102(10): 1265-74, 2008 May 23.
Article in English | MEDLINE | ID: mdl-18420945

ABSTRACT

The small G protein Rho signaling pathways are recognized as major regulators of cardiovascular functions, and activation of Rho proteins appears to be a common component for the pathogenesis of hypertension and vascular proliferative disorders. Recent evidence suggests that modulation of Rho protein signaling by phosphorylation of Rho proteins provides an additional simple mechanism for coordinating Rho protein functions. Phosphorylation of RhoA by cAMP- or cGMP-activated kinase on Ser188 induces cytosolic sequestration of RhoA through increased interaction with guanine dissociation inhibitor, thereby resulting in inhibition of RhoA-dependent functions. Here we show that stimulation of angiotensin II (Ang II) type 2 receptor (AT(2)R) in vascular smooth muscle cells induces Ser188 phosphorylation of RhoA independently of cAMP- or cGMP-activated kinase. We identify the Ser/Thr kinase Ste20-related kinase SLK as a new kinase phosphorylating RhoA on Ser188. Activation of the signaling cascade involving Src homology 2 domain-containing protein-tyrosine phosphatase 1, casein kinase II and SLK is responsible for RhoA phosphorylation and inhibition of RhoA-mediated arterial contraction induced by AT(2)R activation. These results thus identify the molecular mechanism linking AT(2)R to RhoA inhibition and vasodilation.


Subject(s)
Hypertension/metabolism , Muscle, Smooth, Vascular/enzymology , Protein Serine-Threonine Kinases/metabolism , Receptor, Angiotensin, Type 2/metabolism , Vasodilation/physiology , rhoA GTP-Binding Protein/metabolism , Angiotensin II/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/enzymology , Casein Kinase II/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Ethinyl Estradiol/metabolism , Male , Megestrol Acetate/metabolism , Muscle, Smooth, Vascular/cytology , Nitric Oxide Synthase/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/physiology , Vasoconstrictor Agents/pharmacology
18.
Am J Respir Crit Care Med ; 179(12): 1151-8, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19299501

ABSTRACT

RATIONALE: The complex and multifactorial pathogenesis of pulmonary hypertension (PH) involves constriction, remodeling, and in situ thrombosis of pulmonary vessels. Both serotonin (5-HT) and Rho kinase signaling may contribute to these alterations. OBJECTIVES: To investigate possible links between the 5-HT transporter (5-HTT) and RhoA/Rho kinase pathways, as well as their involvement in the progression of human and experimental PH. METHODS: Biochemical and functional analyses of lungs, platelets, and pulmonary artery smooth muscle cells (PA-SMCs) from patients with idiopathic PH (iPH) and 5-HTT overexpressing mice. MEASUREMENTS AND MAIN RESULTS: Lungs, platelets, and PA-SMCs from patients with iPH were characterized by marked elevation in RhoA and Rho kinase activities and a strong increase in 5-HT binding to RhoA indicating RhoA serotonylation. The 5-HTT inhibitor fluoxetine and the type 2 transglutaminase inhibitor monodansylcadaverin prevented 5-HT-induced RhoA serotonylation and RhoA/Rho kinase activation, as well as 5-HT-induced proliferation of PA-SMCs from iPH patients that was also inhibited by the Rho kinase inhibitor fasudil. Increased Rho kinase activity, RhoA activation, and RhoA serotonylation were also observed in lungs from SM22-5-HTT(+)mice, which overexpress 5-HTT in smooth muscle and spontaneously develop PH. Treatment of SM22-5-HTT(+) mice with either fasudil or fluoxetine limited PH progression and RhoA/Rho kinase activation. CONCLUSIONS: RhoA and Rho kinase activities are increased in iPH, in association with enhanced RhoA serotonylation. Direct involvement of the 5-HTT/RhoA/Rho kinase signaling pathway in 5-HT-mediated PA-SMC proliferation and platelet activation during PH progression identify RhoA/Rho kinase signaling as a promising target for new treatments against PH.


Subject(s)
Hypertension, Pulmonary/enzymology , Serotonin/metabolism , Signal Transduction/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Adolescent , Adult , Animals , Blood Platelets/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Female , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Male , Mice , Mice, Transgenic , Middle Aged , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Vasoconstriction/physiology , Young Adult
19.
Matrix Biol ; 88: 33-52, 2020 06.
Article in English | MEDLINE | ID: mdl-31759052

ABSTRACT

Lysyl oxidases are major actors of microenvironment and extracellular matrix (ECM) remodeling. These cross-linking enzymes are thus involved in many aspects of physiopathology, including tumor progression, fibrosis and cardiovascular diseases. We have already shown that Lysyl Oxidase-Like 2 (LOXL2) regulates collagen IV deposition by endothelial cells and angiogenesis. We here provide evidence that LOXL2 also affects deposition of other ECM components, including fibronectin, thus altering structural and mechanical properties of the matrix generated by endothelial cells. LOXL2 interacts intracellularly and directly with collagen IV and fibronectin before incorporation into ECM fibrillar structures upon exocytosis, as demonstrated by TIRF time-lapse microscopy. Furthermore, surface plasmon resonance experiments using recombinant scavenger receptor cysteine-rich (SRCR) domains truncated for the catalytic domain demonstrated their direct binding to collagen IV. We thus used directed mutagenesis to investigate the role of LOXL2 catalytic domain. Neither enzyme activity nor catalytic domain were necessary for collagen IV deposition and angiogenesis, whereas the SRCR domains were effective for these processes. Finally, surface coating with recombinant SRCR domains restored deposition of collagen IV by LOXL2-depleted cells. We thus propose that LOXL2 SRCR domains orchestrate scaffolding of the vascular basement membrane and angiogenesis through interactions with collagen IV and fibronectin, independently of the enzymatic cross-linking activity.


Subject(s)
Amino Acid Oxidoreductases/chemistry , Amino Acid Oxidoreductases/metabolism , Extracellular Matrix/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism , Amino Acid Oxidoreductases/genetics , Animals , Binding Sites , Cell Line , Collagen Type IV/metabolism , Dermis/cytology , Dermis/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibronectins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mutagenesis, Site-Directed , Neovascularization, Physiologic , Protein Domains , Zebrafish , Zebrafish Proteins/genetics
20.
Methods Mol Biol ; 1840: 73-80, 2018.
Article in English | MEDLINE | ID: mdl-30141039

ABSTRACT

The mechanical properties of the cellular microenvironment can impact many aspects of cell behavior, including molecular processes in the nucleus. Recent studies indicate that the LINC complex and its associated nuclear envelope transmit and transduce mechanical stress into biochemical pathways that ultimately regulate nuclear structure or gene expression. Here we describe a method to apply tensional forces to the LINC complex of isolated nuclei. Using magnetic beads and magnets, this technique can be used to explore the biochemical pathways that are activated in response to tension applied to the surface of isolated nuclei.


Subject(s)
Cytoskeleton/metabolism , Mechanotransduction, Cellular , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Cell Fractionation , Cell Nucleus/metabolism , HeLa Cells , Humans
SELECTION OF CITATIONS
SEARCH DETAIL