Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 429
Filter
1.
J Biomed Mater Res A ; 111(5): 701-713, 2023 05.
Article in English | MEDLINE | ID: mdl-36807502

ABSTRACT

In vitro vascular wall bilayer models for drug testing and disease modeling must emulate the physical and biological properties of healthy vascular tissue and its endothelial barrier function. Both endothelial cell (EC)-vascular smooth muscle cell (SMC) interaction across the internal elastic lamina (IEL) and blood vessel stiffness impact endothelial barrier integrity. Polymeric porous track-etched membranes (TEM) typically represent the IEL in laboratory vascular bilayer models. However, TEM stiffness exceeds that of diseased blood vessels, and the membrane pore architecture limits EC-SMC interaction. The mechanical properties of compliant honeycomb film (HCF) membranes better simulate the Young's modulus of healthy blood vessels, and HCFs are thinner (4 vs. 10 µm) and more porous (57 vs. 6.5%) than TEMs. We compared endothelial barrier integrity in vascular wall bilayer models with human ECs and SMCs statically cultured on opposite sides of HCFs and TEMs (5 µm pores) for up to 12 days. Highly segregated localization of tight junction (ZO-1) and adherens junction (VE-cadherin) proteins and quiescent F-actin cytoskeletons demonstrated superior and earlier maturation of interendothelial junctions. Quantifying barrier integrity based on transendothelial electrical resistance (TEER), membranes showed only minor but significant TEER differences despite enhanced junctional protein localization on HCF. Elongated ECs on HCF likely experienced greater paracellular diffusion than blocky ECs on TEM. Also, larger populations of plaques of connexin 43 subunit-containing gap junctions suggested enhanced EC-SMC communication across the more porous, thinner HCF. Compared with standard TEMs, engineered vascular wall bilayers cultured on HCFs better replicate physiologic endothelial barrier integrity.


Subject(s)
Endothelial Cells , Endothelium, Vascular , Humans , Porosity , Endothelial Cells/metabolism , Cell Communication , Tight Junctions/physiology , Cells, Cultured , Adherens Junctions/physiology
2.
Nat Commun ; 13(1): 3520, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35725726

ABSTRACT

Actin-mediated mechanical forces are central drivers of cellular dynamics. They generate protrusive and contractile dynamics, the latter of which are induced in concert with myosin II bundled at the site of contraction. These dynamics emerge concomitantly in tissues and even each cell; thus, the tight regulation of such bidirectional forces is important for proper cellular deformation. Here, we show that contractile dynamics can eventually disturb cell-cell junction contraction in the absence of p21-activated kinase 3 (Pak3). Upon Pak3 depletion, contractility induces the formation of abnormal actin protrusions at the shortening junctions, which causes decrease in E-cadherin levels at the adherens junctions and mislocalization of myosin II at the junctions before they enough shorten, compromising completion of junction shortening. Overexpressing E-cadherin restores myosin II distribution closely placed at the junctions and junction contraction. Our results suggest that contractility both induces and perturbs junction contraction and that the attenuation of such perturbations by Pak3 facilitates persistent junction shortening.


Subject(s)
Actins , p21-Activated Kinases , Actins/physiology , Adherens Junctions/physiology , Cadherins , Epithelial Cells , Feedback , Intercellular Junctions , Myosin Type II , Tight Junctions , p21-Activated Kinases/genetics
3.
Curr Biol ; 32(9): 1986-2000.e5, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35381185

ABSTRACT

Tissue morphogenesis arises from the culmination of changes in cell-cell junction length. Mechanochemical signaling in the form of RhoA underlies these ratcheted contractions, which occur asymmetrically. The underlying mechanisms of asymmetry remain unknown. We use optogenetically controlled RhoA in model epithelia together with biophysical modeling to uncover the mechanism lending to asymmetric vertex motion. Using optogenetic and pharmacological approaches, we find that both local and global RhoA activation can drive asymmetric junction contraction in the absence of tissue-scale patterning. We find that standard vertex models with homogeneous junction properties are insufficient to recapitulate the observed junction dynamics. Furthermore, these experiments reveal a local coupling of RhoA activation with E-cadherin accumulation. This motivates a coupling of RhoA-mediated increases in tension and E-cadherin-mediated adhesion strengthening. We then demonstrate that incorporating this force-sensitive adhesion strengthening into a continuum model is successful in capturing the observed junction dynamics. Thus, we find that a force-dependent intercellular "clutch" at tricellular vertices stabilizes vertex motion under increasing tension and is sufficient to generate asymmetries in junction contraction.


Subject(s)
Adherens Junctions , Epithelial Cells , Adherens Junctions/physiology , Cadherins/genetics , Cell Adhesion , Epithelium , Morphogenesis
4.
PLoS Comput Biol ; 18(1): e1009812, 2022 01.
Article in English | MEDLINE | ID: mdl-35089922

ABSTRACT

Cell intercalation is a key cell behaviour of morphogenesis and wound healing, where local cell neighbour exchanges can cause dramatic tissue deformations such as body axis extension. Substantial experimental work has identified the key molecular players facilitating intercalation, but there remains a lack of consensus and understanding of their physical roles. Existing biophysical models that represent cell-cell contacts with single edges cannot study cell neighbour exchange as a continuous process, where neighbouring cell cortices must uncouple. Here, we develop an Apposed-Cortex Adhesion Model (ACAM) to understand active cell intercalation behaviours in the context of a 2D epithelial tissue. The junctional actomyosin cortex of every cell is modelled as a continuous viscoelastic rope-loop, explicitly representing cortices facing each other at bicellular junctions and the adhesion molecules that couple them. The model parameters relate directly to the properties of the key subcellular players that drive dynamics, providing a multi-scale understanding of cell behaviours. We show that active cell neighbour exchanges can be driven by purely junctional mechanisms. Active contractility and cortical turnover in a single bicellular junction are sufficient to shrink and remove a junction. Next, a new, orthogonal junction extends passively. The ACAM reveals how the turnover of adhesion molecules regulates tension transmission and junction deformation rates by controlling slippage between apposed cell cortices. The model additionally predicts that rosettes, which form when a vertex becomes common to many cells, are more likely to occur in actively intercalating tissues with strong friction from adhesion molecules.


Subject(s)
Actomyosin , Adherens Junctions , Actomyosin/metabolism , Adherens Junctions/physiology , Cell Adhesion , Cell Adhesion Molecules/metabolism , Epithelium/metabolism , Morphogenesis
5.
Mol Biol Cell ; 33(2): ar14, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34851720

ABSTRACT

Distant organ metastasis is linked to poor prognosis during cancer progression. The expression level of the focal adhesion adapter protein paxillin varies among different human cancers, but its role in tumor progression is unclear. Herein we utilize a newly generated PyMT mammary tumor mouse model with conditional paxillin ablation in breast tumor epithelial cells, combined with in vitro three-dimensional (3D) tumor organoids invasion analysis and 2D calcium switch assays, to assess the roles for paxillin in breast tumor cell invasion. Paxillin had little effect on primary tumor initiation and growth but is critical for the formation of distant lung metastasis. In paxillin-depleted 3D tumor organoids, collective cell invasion was substantially perturbed. The 2D cell culture revealed paxillin-dependent stabilization of adherens junctions (AJ). Mechanistically, paxillin is required for AJ assembly through facilitating E-cadherin endocytosis and recycling and HDAC6-mediated microtubule acetylation. Furthermore, Rho GTPase activity analysis and rescue experiments with a RhoA activator or Rac1 inhibitor suggest paxillin is potentially regulating the E-cadherin-dependent junction integrity and contractility through control of the balance of RhoA and Rac1 activities. Together, these data highlight new roles for paxillin in the regulation of cell-cell adhesion and collective tumor cell migration to promote the formation of distance organ metastases.


Subject(s)
Adherens Junctions/physiology , Neoplasm Invasiveness/genetics , Paxillin/metabolism , Adherens Junctions/genetics , Animals , Breast/metabolism , Breast Neoplasms/metabolism , Cadherins/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Epithelial Cells/metabolism , Female , Focal Adhesions/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Paxillin/genetics , rho GTP-Binding Proteins/metabolism
6.
Mol Biol Cell ; 32(18): 1724-1736, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34081532

ABSTRACT

The vascular system is precisely regulated to adjust blood flow to organismal demand, thereby guaranteeing adequate perfusion under varying physiological conditions. Mechanical forces, such as cyclic circumferential stretch, are among the critical stimuli that dynamically adjust vessel distribution and diameter, but the precise mechanisms of adaptation to changing forces are unclear. We find that endothelial monolayers respond to cyclic stretch by transient remodeling of the vascular endothelial cadherin-based adherens junctions and the associated actomyosin cytoskeleton. Time-resolved proteomic profiling reveals that this remodeling is driven by calcium influx through the mechanosensitive Piezo1 channel, triggering Rho activation to increase actomyosin contraction. As the mechanical stimulus persists, calcium signaling is attenuated through transient down-regulation of Piezo1 protein. At the same time, filamins are phosphorylated to increase monolayer stiffness, allowing mechanoadaptation to restore junctional integrity despite continuing exposure to stretch. Collectively, this study identifies a biphasic response to cyclic stretch, consisting of an initial calcium-driven junctional mechanoresponse, followed by mechanoadaptation facilitated by monolayer stiffening.


Subject(s)
Actin Cytoskeleton/metabolism , Actomyosin , Antigens, CD/metabolism , Cadherins/metabolism , Calcium Signaling , Mechanotransduction, Cellular , Actomyosin/metabolism , Adherens Junctions/physiology , Antigens, CD/genetics , Biomechanical Phenomena , Cadherins/genetics , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Calcium Signaling/drug effects , Cytochalasin D/pharmacology , Filamins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Ion Channels/genetics , Ion Channels/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Protein Interaction Maps , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
7.
J Cell Biol ; 220(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33939796

ABSTRACT

The neural tube forms when neural stem cells arrange into a pseudostratified, single-cell-layered epithelium, with a marked apico-basal polarity, and in which adherens junctions (AJs) concentrate in the subapical domain. We previously reported that sustained ß-catenin expression promotes the formation of enlarged apical complexes (ACs), enhancing apico-basal polarity, although the mechanism through which this occurs remained unclear. Here, we show that ß-catenin interacts with phosphorylated pro-N-cadherin early in its transit through the Golgi apparatus, promoting propeptide excision and the final maturation of N-cadherin. We describe a new ß-catenin-dependent interaction of N-cadherin with Drebrin-like (Dbnl), an actin-binding protein that is involved in anterograde Golgi trafficking of proteins. Notably, Dbnl knockdown led to pro-N-cadherin accumulation and limited AJ formation. In brief, we demonstrate that Dbnl and Drebrin-like ß-catenin assist in the maturation of pro-N-cadherin, which is critical for AJ formation and for the recruitment AC components like aPKC and, consequently, for the maintenance of apico-basal polarity.


Subject(s)
Adherens Junctions/physiology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Polarity , Microfilament Proteins/metabolism , Neural Stem Cells/metabolism , beta Catenin/metabolism , Antigens, CD/genetics , Cadherins/genetics , Cells, Cultured , HEK293 Cells , Humans , Microfilament Proteins/genetics , Neural Stem Cells/cytology , beta Catenin/genetics , src Homology Domains/genetics
8.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-33999996

ABSTRACT

Movement of epithelial cells in a tissue occurs through neighbor exchange and drives tissue shape changes. It requires intercellular junction remodeling, a process typically powered by the contractile actomyosin cytoskeleton. This has been investigated mainly in homogeneous epithelia, where intercalation takes minutes. However, in some tissues, intercalation involves different cell types and can take hours. Whether slow and fast intercalation share the same mechanisms remains to be examined. To address this issue, we used the fly eye, where the cone cells exchange neighbors over ∼10 h to shape the lens. We uncovered three pathways regulating this slow mode of cell intercalation. First, we found a limited requirement for MyosinII. In this case, mathematical modeling predicts an adhesion-dominant intercalation mechanism. Genetic experiments support this prediction, revealing a role for adhesion through the Nephrin proteins Roughest and Hibris. Second, we found that cone cell intercalation is regulated by the Notch pathway. Third, we show that endocytosis is required for membrane removal and Notch activation. Taken together, our work indicates that adhesion, endocytosis and Notch can direct slow cell intercalation during tissue morphogenesis.


Subject(s)
Cell Adhesion/physiology , Drosophila Proteins/metabolism , Drosophila/embryology , Endocytosis/physiology , Receptors, Notch/metabolism , Retina/embryology , Retinal Cone Photoreceptor Cells/metabolism , Actomyosin/metabolism , Adherens Junctions/physiology , Animals , Body Patterning/physiology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Communication , Drosophila Proteins/genetics , Epithelial Cells/cytology , Eye Proteins/metabolism , Focal Adhesions/physiology , Membrane Proteins/metabolism , Myosin Type II/metabolism , Receptors, Notch/genetics , Signal Transduction/physiology
10.
J Cell Biol ; 220(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33914026

ABSTRACT

Rac1 GTPase is hyperactivated in tumors and contributes to malignancy. Rac1 disruption of junctions requires its effector PAK1, but the precise mechanisms are unknown. Here, we show that E-cadherin is internalized via micropinocytosis in a PAK1-dependent manner without catenin dissociation and degradation. In addition to internalization, PAK1 regulates E-cadherin transport by fine-tuning Rab small GTPase function. PAK1 phosphorylates a core Rab regulator, RabGDIß, but not RabGDIα. Phosphorylated RabGDIß preferentially associates with Rab5 and Rab11, which is predicted to promote Rab retrieval from membranes. Consistent with this hypothesis, Rab11 is activated by Rac1, and inhibition of Rab11 function partially rescues E-cadherin destabilization. Thus, Rac1 activation reduces surface cadherin levels as a net result of higher bulk flow of membrane uptake that counteracts Rab11-dependent E-cadherin delivery to junctions (recycling and/or exocytosis). This unique small GTPase crosstalk has an impact on Rac1 and PAK1 regulation of membrane remodeling during epithelial dedifferentiation, adhesion, and motility.


Subject(s)
Adherens Junctions/physiology , Exocytosis , Keratinocytes/physiology , p21-Activated Kinases/metabolism , rab GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Actins/metabolism , Adherens Junctions/chemistry , Cells, Cultured , Humans , Keratinocytes/cytology , Signal Transduction , p21-Activated Kinases/genetics , rab GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/genetics
11.
J Cell Biol ; 220(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33688935

ABSTRACT

Epithelia are continuously self-renewed, but how epithelial integrity is maintained during the morphological changes that cells undergo in mitosis is not well understood. Here, we show that as epithelial cells round up when they enter mitosis, they exert tensile forces on neighboring cells. We find that mitotic cell-cell junctions withstand these tensile forces through the mechanosensitive recruitment of the actin-binding protein vinculin to cadherin-based adhesions. Surprisingly, vinculin that is recruited to mitotic junctions originates selectively from the neighbors of mitotic cells, resulting in an asymmetric composition of cadherin junctions. Inhibition of junctional vinculin recruitment in neighbors of mitotic cells results in junctional breakage and weakened epithelial barrier. Conversely, the absence of vinculin from the cadherin complex in mitotic cells is necessary to successfully undergo mitotic rounding. Our data thus identify an asymmetric mechanoresponse at cadherin adhesions during mitosis, which is essential to maintain epithelial integrity while at the same time enable the shape changes of mitotic cells.


Subject(s)
Adherens Junctions/physiology , Epithelial Cells/physiology , Epithelium/physiology , Intercellular Junctions/physiology , Mitosis/physiology , Actin Cytoskeleton/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cell Line , Dogs , Epithelial Cells/metabolism , Epithelium/metabolism , Intercellular Junctions/metabolism , Madin Darby Canine Kidney Cells , Microfilament Proteins/metabolism
12.
Dev Cell ; 56(6): 761-780.e7, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33725480

ABSTRACT

Vinculin, a mechanotransducer associated with both adherens junctions (AJs) and focal adhesions (FAs), plays a central role in force transmission through cell-cell and cell-substratum contacts. We generated the conditional knockout (cKO) of vinculin in murine skin that results in the loss of bulge stem cell (BuSC) quiescence and promotes continual cycling of the hair follicles. Surprisingly, we find that the AJs in vinculin cKO cells are mechanically weak and impaired in force generation despite increased junctional expression of E-cadherin and α-catenin. Mechanistically, we demonstrate that vinculin functions by keeping α-catenin in a stretched/open conformation, which in turn regulates the retention of YAP1, another potent mechanotransducer and regulator of cell proliferation, at the AJs. Altogether, our data provide mechanistic insights into the hitherto-unexplored regulatory link between the mechanical stability of cell junctions and contact-inhibition-mediated maintenance of BuSC quiescence.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/physiology , Hair Follicle/physiology , Mechanotransduction, Cellular , Stem Cells/physiology , Vinculin/physiology , alpha Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion , Female , Hair Follicle/cytology , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Stem Cells/cytology , YAP-Signaling Proteins , alpha Catenin/genetics
13.
Dev Cell ; 56(2): 202-212, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33453154

ABSTRACT

Cell-cell junctions, in particular adherens junctions, are major determinants of tissue mechanics during morphogenesis and homeostasis. In attempts to link junctional mechanics to tissue mechanics, many have utilized explicitly or implicitly equilibrium approaches based on adhesion energy, surface energy, and contractility to determine the mechanical equilibrium at junctions. However, it is increasingly clear that they have significant limitations, such as that it remains challenging to link the dynamics of the molecular components to the resulting physical properties of the junction, to its remodeling ability, and to its adhesion strength. In this perspective, we discuss recent attempts to consider the aspect of energy dissipation at junctions to draw contact points with soft matter physics where energy loss plays a critical role in adhesion theories. We set the grounds for a theoretical framework of the junction mechanics that bridges the dynamics at the molecular scale to the mechanics at the tissue scale.


Subject(s)
Adherens Junctions/physiology , Biophysics , Cell Adhesion , Homeostasis , Mechanotransduction, Cellular , Morphogenesis , Animals , Humans
14.
Mol Biol Cell ; 32(4): 348-361, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33378218

ABSTRACT

Sustained cell migration is essential for wound healing and cancer metastasis. The epidermal growth factor receptor (EGFR) signaling cascade is known to drive cell migration and proliferation. While the signal transduction downstream of EGFR has been extensively investigated, our knowledge of the initiation and maintenance of EGFR signaling during cell migration remains limited. The metalloprotease TACE (tumor necrosis factor alpha converting enzyme) is responsible for producing active EGFR family ligands in the via ligand shedding. Sustained TACE activity may perpetuate EGFR signaling and reduce a cell's reliance on exogenous growth factors. Using a cultured keratinocyte model system, we show that depletion of α-catenin perturbs adherens junctions, enhances cell proliferation and motility, and decreases dependence on exogenous growth factors. We show that the underlying mechanism for these observed phenotypical changes depends on enhanced autocrine/paracrine release of the EGFR ligand transforming growth factor alpha in a TACE-dependent manner. We demonstrate that proliferating keratinocyte epithelial cell clusters display waves of oscillatory extracellular signal-regulated kinase (ERK) activity, which can be eliminated by TACE knockout, suggesting that these waves of oscillatory ERK activity depend on autocrine/paracrine signals produced by TACE. These results provide new insights into the regulatory role of adherens junctions in initiating and maintaining autocrine/paracrine signaling with relevance to wound healing and cellular transformation.


Subject(s)
ADAM17 Protein/metabolism , Adherens Junctions/metabolism , alpha Catenin/metabolism , ADAM17 Protein/physiology , Adherens Junctions/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , HaCaT Cells , Humans , Metalloproteases/metabolism , Paracrine Communication/physiology , Phosphorylation , Signal Transduction , Transforming Growth Factor alpha/metabolism , alpha Catenin/physiology
15.
Am J Pathol ; 191(3): 438-453, 2021 03.
Article in English | MEDLINE | ID: mdl-33345995

ABSTRACT

Hepatic stellate cells (HSCs) are resident mesenchymal cells in the space of Disse interposed between liver sinusoidal endothelial cells and hepatocytes. Thorn-like microprojections, or spines, project out from the cell surface of HSCs, crossing the space of Disse, to establish adherens junctions with neighboring hepatocytes. Although HSC activation is initiated largely from stimulation by adjacent cells, isolated HSCs also activate spontaneously in primary culture on plastic. Therefore, other unknown HSC-initiating factors apart from paracrine stimuli may promote activation. The dissociation of adherens junctions between HSCs and hepatocytes as an activating signal for HSCs was explored, establishing epithelial cadherin (E-cadherin) as an adhesion molecule linking hepatocytes and HSCs. In vivo, following carbon tetrachloride-induced liver injury, HSCs lost their spines and dissociated from adherens junctions in the early stages of injury, and were subsequently activated along with an increase in YAP/TAZ expression. After abrogation of liver injury, HSCs reconstructed their spines and adherens junctions. In vitro, reconstitution of E-cadherin-containing adherens junctions by forced E-cadherin expression quiesced HSCs and suppressed TAZ expression. Additionally, increase of TAZ expression leading to the activation of HSCs by autocrine stimulation of transforming growth factor-ß, was revealed as a mechanism of spontaneous activation. Thus, we have uncovered a critical event required for HSC activation through enhanced TAZ-mediated mechanotransduction after the loss of adherens junctions between HSCs and hepatocytes.


Subject(s)
Adherens Junctions/physiology , Cadherins/metabolism , Endothelial Cells/physiology , Hepatic Stellate Cells/physiology , Hepatocytes/physiology , Mechanotransduction, Cellular , Animals , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Hepatic Stellate Cells/cytology , Hepatocytes/cytology , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Signal Transduction
16.
Mol Biol Cell ; 32(3): 226-236, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33326251

ABSTRACT

Although the actomyosin cytoskeleton has been implicated in clathrin-mediated endocytosis, a clear requirement for actomyosin in clathrin-independent endocytosis (CIE) has not been demonstrated. We discovered that the Rho-associated kinase ROCK2 is required for CIE of MHCI and CD59 through promotion of myosin II activity. Myosin IIA promoted internalization of MHCI and myosin IIB drove CD59 uptake in both HeLa and polarized Caco2 intestinal epithelial cells. In Caco2 cells, myosin IIA localized to the basal cortex and apical brush border and mediated MHCI internalization from the basolateral domain, while myosin IIB localized at the basal cortex and apical cell-cell junctions and promoted CD59 uptake from the apical membrane. Atomic force microscopy demonstrated that myosin IIB mediated apical epithelial tension in Caco2 cells. Thus, specific cargoes are internalized by ROCK2-mediated activation of myosin II isoforms to mediate spatial regulation of CIE, possibly by modulation of local cortical tension.


Subject(s)
Endocytosis/physiology , Myosin Type II/metabolism , rho-Associated Kinases/metabolism , Actin Cytoskeleton/metabolism , Actomyosin/metabolism , Adherens Junctions/physiology , CD59 Antigens/metabolism , Caco-2 Cells , Cadherins/metabolism , Clathrin/metabolism , Cytoskeletal Proteins/physiology , Cytoskeleton/metabolism , Epithelial Cells/cytology , HeLa Cells , Histocompatibility Antigens Class I/metabolism , Humans , Myosin Type II/physiology , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Protein Isoforms/metabolism , rho-Associated Kinases/physiology
17.
PLoS Biol ; 18(11): e3000946, 2020 11.
Article in English | MEDLINE | ID: mdl-33253145

ABSTRACT

Inflammation of the central nervous system (CNS) induces endothelial blood-brain barrier (BBB) opening as well as the formation of a tight junction barrier between reactive astrocytes at the Glia Limitans. We hypothesized that the CNS parenchyma may acquire protection from the reactive astrocytic Glia Limitans not only during neuroinflammation but also when BBB integrity is compromised in the resting state. Previous studies found that astrocyte-derived Sonic hedgehog (SHH) stabilizes the BBB during CNS inflammatory disease, while endothelial-derived desert hedgehog (DHH) is expressed at the BBB under resting conditions. Here, we investigated the effects of endothelial Dhh on the integrity of the BBB and Glia Limitans. We first characterized DHH expression within endothelial cells at the BBB, then demonstrated that DHH is down-regulated during experimental autoimmune encephalomyelitis (EAE). Using a mouse model in which endothelial Dhh is inducibly deleted, we found that endothelial Dhh both opens the BBB via the modulation of forkhead box O1 (FoxO1) transcriptional activity and induces a tight junctional barrier at the Glia Limitans. We confirmed the relevance of this glial barrier system in human multiple sclerosis active lesions. These results provide evidence for the novel concept of "chronic neuroinflammatory tolerance" in which BBB opening in the resting state is sufficient to stimulate a protective barrier at the Glia Limitans that limits the severity of subsequent neuroinflammatory disease. In summary, genetic disruption of the BBB generates endothelial signals that drive the formation under resting conditions of a secondary barrier at the Glia Limitans with protective effects against subsequent CNS inflammation. The concept of a reciprocally regulated CNS double barrier system has implications for treatment strategies in both the acute and chronic phases of multiple sclerosis pathophysiology.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Brain Barrier/physiopathology , Adherens Junctions/pathology , Adherens Junctions/physiology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , Astrocytes/pathology , Astrocytes/physiology , Cadherins/genetics , Cadherins/physiology , Capillary Permeability/genetics , Capillary Permeability/physiology , Claudin-5/genetics , Claudin-5/physiology , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Hedgehog Proteins/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Tight Junctions/pathology , Tight Junctions/physiology
18.
Mol Biol Cell ; 31(23): 2557-2569, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32903148

ABSTRACT

Epithelia can eliminate apoptotic cells by apical extrusion. This is a complex morphogenetic event where expulsion of the apoptotic cell is accompanied by rearrangement of its immediate neighbors to form a rosette. A key mechanism for extrusion is constriction of an actomyosin network that neighbor cells form at their interface with the apoptotic cell. Here we report a complementary process of cytoskeletal relaxation that occurs when cortical contractility is down-regulated at the junctions between those neighbor cells themselves. This reflects a mechanosensitive Src family kinase (SFK) signaling pathway that is activated in neighbor cells when the apoptotic cell relaxes shortly after injury. Inhibiting SFK signaling blocks both the expulsion of apoptotic cells and the rosette formation among their neighbor cells. This reveals the complex pattern of spatially distinct contraction and relaxation that must be established in the neighboring epithelium for apoptotic cells to be extruded.


Subject(s)
Adherens Junctions/metabolism , Apoptosis/physiology , src-Family Kinases/metabolism , Actin Cytoskeleton/metabolism , Actomyosin/metabolism , Adherens Junctions/physiology , Epithelial Cells/metabolism , Epithelium/metabolism , Epithelium/physiology , Humans , MCF-7 Cells , Morphogenesis , Signal Transduction , src-Family Kinases/physiology
19.
Sci Rep ; 10(1): 14124, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32839490

ABSTRACT

The present study shows chronic adjustments in the myotendinous junction (MTJ) in response to different ladder-based resistance training (LRT) protocols. Thirty adult male Wistar rats were divided into groups: sedentary (S), calisthenics (LRT without additional load [C]), and resistance-trained (LRT with extra weight [R]). We demonstrated longer lengths of sarcoplasmatic invaginations in the trained groups; however, evaginations were seen mainly in group R. We showed a greater thickness of sarcoplasmatic invaginations in groups C and R, in addition to greater evaginations in R. We also observed thinner basal lamina in trained groups. The support collagen layer (SCL) adjacent to the MTJ and the diameters of the transverse fibrils were larger in R. We also discovered a niche of telocytes in the MTJ with electron micrographs of the plantar muscle and with immunostaining with CD34+ in the gastrocnemius muscle near the blood vessels and pericytes. We concluded that the continuous adjustments in the MTJ ultrastructure were the result of tissue plasticity induced by LRT, which is causally related to muscle hypertrophy and, consequently, to the remodeling of the contact interface. Also, we reveal the existence of a collagen layer adjacent to MTJ and discover a new micro anatomic location of telocytes.


Subject(s)
Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Physical Conditioning, Animal/physiology , Resistance Training/methods , Sarcoplasmic Reticulum/physiology , Telocytes/physiology , Adaptation, Physiological/physiology , Adherens Junctions/physiology , Animals , Basement Membrane/physiology , Cell Adhesion , Cell Movement/physiology , Cell-Matrix Junctions/physiology , Collagen/metabolism , Male , Rats , Rats, Wistar , Sedentary Behavior
20.
Biomed Pharmacother ; 125: 109942, 2020 May.
Article in English | MEDLINE | ID: mdl-32044715

ABSTRACT

The transmembrane protein Crumbs (Crb), a key regulator of apical polarity, has a known involvement in establishment of the apical zonula adherens in epithelia, although the precise mechanism remains elusive. The zonula adherens are required to maintain the integrity and orderly arrangement of epithelia. Loss of the zonula adherens leads to morphogenetic defects in the tissues derived from epithelium. In this study, we revealed that the intracellular tail of Crb2a promoted the apical distribution of adherens junctions (AJs) in zebrafish retinal and lens epithelia, but caused assembly into unstable punctum adherens-like adhesion plaques. The extracellular region of Crb2a guided the transformation of AJs from the punctum adherens into stable zonula adherens. Accordingly, a truncated form of Crb2a lacking the extracellular region (Crb2aΔEX) could only partially rescue the retinal patterning defects in crb2a null mutant zebrafish (crb2am289). By contrast, constitutive over-expression of Crb2aΔEX disrupted the integrity of the outer limiting membrane in photoreceptors, which is derived from the zonula adherens of the retinal neuroepithelium. This study demonstrated that both the extracellular region and the intracellular tail of Crb2a are required to guide the formation of the apical zonula adherens.


Subject(s)
Adherens Junctions/physiology , Membrane Proteins/metabolism , Morphogenesis/physiology , Zebrafish Proteins/metabolism , Animals , Epithelium/metabolism , Extracellular Space/physiology , Intracellular Space/physiology , Lens, Crystalline/metabolism , Membrane Proteins/genetics , Mutation , Retina/metabolism , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...