Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Curr Biol ; 32(14): 3033-3047.e9, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35688156

ABSTRACT

Physiological and pathological cardiac stress induced by exercise and hypertension, respectively, increase the hemodynamic load for the heart and trigger specific hypertrophic signals in cardiomyocytes leading to adaptive or maladaptive cardiac hypertrophy responses involving a mechanosensitive remodeling of the contractile cytoskeleton. Integrins sense load and have been implicated in cardiac hypertrophy, but how they discriminate between the two types of cardiac stress and translate mechanical loads into specific cytoskeletal signaling pathways is not clear. Here, we report that the focal adhesion protein ß-parvin is highly expressed in cardiomyocytes and facilitates the formation of cell protrusions, the serial assembly of newly synthesized sarcomeres, and the hypertrophic growth of neonatal rat ventricular cardiomyocytes (NRVCs) in vitro. In addition, physiological mechanical loading of NRVCs by either the application of cyclic, uni-axial stretch, or culture on physiologically stiff substrates promotes NRVC elongation in a ß-parvin-dependent manner, which is achieved by binding of ß-parvin to α/ß-PIX, which in turn activates Rac1. Importantly, loss-of-function studies in mice also revealed that ß-parvin is essential for the exercise-induced cardiac hypertrophy response in vivo. Our results identify ß-parvin as a novel mechano-responsive signaling hub in hypertrophic cardiomyocytes that drives cell elongation in response to physiological mechanical loads.


Subject(s)
Focal Adhesions , Myocytes, Cardiac , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Integrins/metabolism , Mice , Myocytes, Cardiac/metabolism , Rats , Sarcomeres/pathology
2.
J Cell Sci ; 135(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35532004

ABSTRACT

The vitronectin receptor integrin αVß5 can reside in two distinct adhesion structures - focal adhesions (FAs) and flat clathrin lattices (FCLs). Here, we investigate the mechanism that regulates the subcellular distribution of ß5 in keratinocytes and show that ß5 has approximately 7- and 5-fold higher affinity for the clathrin adaptors ARH (also known as LDLRAP1) and Numb, respectively, than for the talin 1 (TLN1); all proteins that bind to the membrane-proximal NPxY motif of the ß5 cytoplasmic domain. Using mass spectrometry, we identified ß5 interactors, including the Rho GEFs p115Rho-GEF and GEF-H1 (also known as ARHGEF1 and ARHGEF2, respectively), and the serine protein kinase MARK2, depletion of which diminishes the clustering of ß5 in FCLs. Replacement of two serine residues (S759 and S762) in the ß5 cytoplasmic domain with phospho-mimetic glutamate residues causes a shift in the localization of ß5 from FAs into FCLs without affecting the interactions with MARK2, p115Rho-GEF or GEF-H1. Instead, we demonstrate that changes in the actomyosin-based cellular contractility by ectopic expression of activated Rho or disruption of microtubules regulates ß5 localization. Finally, we present evidence that ß5 in either FAs or FCLs functions to promote adhesion to vitronectin, cell spreading, and proliferation.


Subject(s)
Clathrin , Receptors, Vitronectin , Cell Adhesion/physiology , Cell Proliferation , Clathrin/metabolism , Focal Adhesions/metabolism , Receptors, Vitronectin/metabolism , Serine/metabolism
3.
Front Cell Dev Biol ; 10: 836797, 2022.
Article in English | MEDLINE | ID: mdl-35309920

ABSTRACT

Integrins and discoidin domain receptors (DDRs) 1 and 2 promote cell adhesion and migration on both fibrillar and non fibrillar collagens. Collagen I contains DDR and integrin selective binding motifs; however, the relative contribution of these two receptors in regulating cell migration is unclear. DDR1 has five isoforms (DDR1a-e), with most cells expressing the DDR1a and DDR1b isoforms. We show that human embryonic kidney 293 cells expressing DDR1b migrate more than DDR1a expressing cells on DDR selective substrata as well as on collagen I in vitro. In addition, DDR1b expressing cells show increased lung colonization after tail vein injection in nude mice. DDR1a and DDR1b differ from each other by an extra 37 amino acids in the DDR1b cytoplasmic domain. Interestingly, these 37 amino acids contain an NPxY motif which is a central control module within the cytoplasmic domain of ß integrins and acts by binding scaffold proteins, including talin. Using purified recombinant DDR1 cytoplasmic tail proteins, we show that DDR1b directly binds talin with higher affinity than DDR1a. In cells, DDR1b, but not DDR1a, colocalizes with talin and integrin ß1 to focal adhesions and enhances integrin ß1-mediated cell migration. Moreover, we show that DDR1b promotes cell migration by enhancing Rac1 activation. Mechanistically DDR1b interacts with the GTPase-activating protein (GAP) Breakpoint cluster region protein (BCR) thus reducing its GAP activity and enhancing Rac activation. Our study identifies DDR1b as a major driver of cell migration and talin and BCR as key players in the interplay between integrins and DDR1b in regulating cell migration.

4.
Blood ; 139(23): 3402-3417, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35303071

ABSTRACT

Neutrophils are key players during host defense and sterile inflammation. Neutrophil dysfunction is a characteristic feature of the acquired immunodeficiency during kidney disease. We speculated that the impaired renal clearance of the intrinsic purine metabolite soluble uric acid (sUA) may account for neutrophil dysfunction. Indeed, hyperuricemia (HU, serum UA of 9-12 mg/dL) related or unrelated to kidney dysfunction significantly diminished neutrophil adhesion and extravasation in mice with crystal- and coronavirus-related sterile inflammation using intravital microscopy and an air pouch model. This impaired neutrophil recruitment was partially reversible by depleting UA with rasburicase. We validated these findings in vitro using either neutrophils or serum from patients with kidney dysfunction-related HU with or without UA depletion, which partially normalized the defective migration of neutrophils. Mechanistically, sUA impaired ß2 integrin activity and internalization/recycling by regulating intracellular pH and cytoskeletal dynamics, physiological processes that are known to alter the migratory and phagocytic capability of neutrophils. This effect was fully reversible by blocking intracellular uptake of sUA via urate transporters. In contrast, sUA had no effect on neutrophil extracellular trap formation in neutrophils from healthy subjects or patients with kidney dysfunction. Our results identify an unexpected immunoregulatory role of the intrinsic purine metabolite sUA, which contrasts the well-known immunostimulatory effects of crystalline UA. Specifically targeting UA may help to overcome certain forms of immunodeficiency, for example in kidney dysfunction, but may enhance sterile forms of inflammation.


Subject(s)
CD18 Antigens , Uric Acid , Animals , CD18 Antigens/metabolism , Humans , Immunity, Innate , Inflammation , Mice , Neutrophil Infiltration , Neutrophils , Uric Acid/pharmacology , Uric Acid/urine
5.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-34996844

ABSTRACT

Integrins require an activation step before ligand binding and signaling that is mediated by talin and kindlin binding to the ß integrin cytosolic domain (ß-tail). Conflicting reports exist about the contribution of phosphorylation of a conserved threonine motif in the ß1-tail (ß1-pT788/pT789) to integrin activation. We show that widely used and commercially available antibodies against ß1-pT788/pT789 integrin do not detect specific ß1-pT788/pT789 integrin signals in immunoblots of several human and mouse cell lysates but bind bi-phosphorylated threonine residues in numerous proteins, which were identified by mass spectrometry experiments. Furthermore, we found that fibroblasts and epithelial cells expressing the phospho-mimicking ß1-TT788/789DD integrin failed to activate ß1 integrins and displayed reduced integrin ligand binding, adhesion initiation and cell spreading. These cellular defects are specifically caused by the inability of kindlin to bind ß1-tail polypeptides carrying a phosphorylated threonine motif or phospho-mimicking TT788/789DD substitutions. Our findings indicate that the double-threonine motif in ß1-class integrins is not a major phosphorylation site but if phosphorylated would curb integrin function.


Subject(s)
Integrin beta1 , Threonine , Amino Acid Motifs/physiology , Animals , Cells, Cultured , Fibroblasts/metabolism , Humans , Integrin beta1/chemistry , Integrin beta1/metabolism , Mice , Phosphorylation , Threonine/chemistry , Threonine/metabolism
6.
mBio ; 12(5): e0106221, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34663090

ABSTRACT

While Staphylococcus aureus has classically been considered an extracellular pathogen, these bacteria are also capable of being taken up by host cells, including nonprofessional phagocytes such as endothelial cells, epithelial cells, or osteoblasts. The intracellular S. aureus lifestyle contributes to infection development. The predominant recognition and internalization pathway appears to be the binding of the bacteria via a fibronectin bridge to the α5ß1-integrin on the host cell membrane, followed by phagocytosis. Although osteoblasts showed high expression of α5ß1-integrin and fibronectin, and bacteria adhered to osteoblasts to a high proportion, here we demonstrate by internalization assays and immunofluorescence microscopy that S. aureus was less engulfed in osteoblasts than in epithelial cells. The addition of exogenous fibronectin during the infection of cells with S. aureus resulted in an increased uptake by epithelial cells but not by osteoblasts. This contrasts with the previous conception of the uptake mechanism, where high expression of integrin and fibronectin would promote the bacterial uptake into host cells. Extracellular fibronectin surrounding osteoblasts, but not epithelial cells, is organized in a fibrillary network. The inhibition of fibril formation, the short interfering RNA-mediated reduction of fibronectin expression, and the disruption of the fibronectin-fibril meshwork all resulted in a significant increase in S. aureus uptake by osteoblasts. Thus, the network of fibronectin fibrils appears to strongly reduce the uptake of S. aureus into a given host cell, indicating that the supramolecular structure of fibronectin determines the capacity of particular host cells to internalize the pathogen. IMPORTANCE Traditionally, Staphylococcus aureus has been considered an extracellular pathogen. However, among other factors, the frequent failure of antimicrobial therapy and the ability of the pathogen to cause recurrent disease have established the concept of eukaryotic invasion of the pathogen, thereby evading the host's immune system. In the current model of host cell invasion, bacteria initially bind to α5ß1 integrin on the host cell side via a fibronectin bridge, which eventually leads to phagocytosis of S. aureus by host cells. However, in this study, we demonstrate that not the crude amount but the supramolecular structure of fibronectin molecules deposited on the eukaryotic cell surface plays an essential role in bacterial uptake by host cells. Our findings explain the large differences of S. aureus uptake efficacy in different host cell types as well as in vivo differences between courses of bacterial infections and the localization of bacteria in different clinical settings.


Subject(s)
Endothelial Cells/microbiology , Fibronectins/metabolism , Host Microbial Interactions , Osteoblasts/microbiology , Staphylococcus aureus/physiology , A549 Cells , Adhesins, Bacterial/metabolism , Cells, Cultured , Fibronectins/genetics , Humans , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Phagocytosis , Staphylococcus aureus/pathogenicity
7.
Nat Commun ; 12(1): 4086, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215727

ABSTRACT

Sarcomeres, the basic contractile units of striated muscle, produce the forces driving muscular contraction through cross-bridge interactions between actin-containing thin filaments and myosin II-based thick filaments. Until now, direct visualization of the molecular architecture underlying sarcomere contractility has remained elusive. Here, we use in situ cryo-electron tomography to unveil sarcomere contraction in frozen-hydrated neonatal rat cardiomyocytes. We show that the hexagonal lattice of the thick filaments is already established at the neonatal stage, with an excess of thin filaments outside the trigonal positions. Structural assessment of actin polarity by subtomogram averaging reveals that thin filaments in the fully activated state form overlapping arrays of opposite polarity in the center of the sarcomere. Our approach provides direct evidence for thin filament sliding during muscle contraction and may serve as a basis for structural understanding of thin filament activation and actomyosin interactions inside unperturbed cellular environments.


Subject(s)
Actins/metabolism , Muscle Contraction/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Actin Cytoskeleton , Animals , Male , Mice, Inbred C57BL , Muscle, Skeletal/chemistry , Muscle, Striated , Myocytes, Cardiac/ultrastructure , Myofibrils , Rats , Rats, Wistar , Sarcomeres/ultrastructure
8.
Nat Commun ; 12(1): 3104, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035280

ABSTRACT

Focal adhesions (FAs) initiate chemical and mechanical signals involved in cell polarity, migration, proliferation and differentiation. Super-resolution microscopy revealed that FAs are organized at the nanoscale into functional layers from the lower plasma membrane to the upper actin cytoskeleton. Yet, how FAs proteins are guided into specific nano-layers to promote interaction with given targets is unknown. Using single protein tracking, super-resolution microscopy and functional assays, we link the molecular behavior and 3D nanoscale localization of kindlin with its function in integrin activation inside FAs. We show that immobilization of integrins in FAs depends on interaction with kindlin. Unlike talin, kindlin displays free diffusion along the plasma membrane outside and inside FAs. We demonstrate that the kindlin Pleckstrin Homology domain promotes membrane diffusion and localization to the membrane-proximal integrin nano-layer, necessary for kindlin enrichment and function in FAs. Using kindlin-deficient cells, we show that kindlin membrane localization and diffusion are crucial for integrin activation, cell spreading and FAs formation. Thus, kindlin uses a different route than talin to reach and activate integrins, providing a possible molecular basis for their complementarity during integrin activation.


Subject(s)
Cell Membrane/metabolism , Focal Adhesions/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Binding Sites , Cell Adhesion , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Integrins/genetics , Membrane Proteins/genetics , Mice, Knockout , Motion , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Neoplasm Proteins/genetics , Protein Binding
9.
Nat Commun ; 12(1): 919, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568673

ABSTRACT

Single-molecule localization microscopy (SMLM) enabling the investigation of individual proteins on molecular scales has revolutionized how biological processes are analysed in cells. However, a major limitation of imaging techniques reaching single-protein resolution is the incomplete and often unknown labeling and detection efficiency of the utilized molecular probes. As a result, fundamental processes such as complex formation of distinct molecular species cannot be reliably quantified. Here, we establish a super-resolution microscopy framework, called quantitative single-molecule colocalization analysis (qSMCL), which permits the identification of absolute molecular quantities and thus the investigation of molecular-scale processes inside cells. The method combines multiplexed single-protein resolution imaging, automated cluster detection, in silico data simulation procedures, and widely applicable experimental controls to determine absolute fractions and spatial coordinates of interacting species on a true molecular level, even in highly crowded subcellular structures. The first application of this framework allowed the identification of a long-sought ternary adhesion complex-consisting of talin, kindlin and active ß1-integrin-that specifically forms in cell-matrix adhesion sites. Together, the experiments demonstrate that qSMCL allows an absolute quantification of multiplexed SMLM data and thus should be useful for investigating molecular mechanisms underlying numerous processes in cells.


Subject(s)
Cytoskeletal Proteins/chemistry , Integrin beta1/chemistry , Muscle Proteins/chemistry , Single Molecule Imaging/methods , Talin/chemistry , Animals , Cell Adhesion , Cell Line , Humans , Mice , Single Molecule Imaging/instrumentation
10.
J Cell Sci ; 133(18)2020 09 23.
Article in English | MEDLINE | ID: mdl-32843574

ABSTRACT

Integrin function depends on the continuous internalization of integrins and their subsequent endosomal recycling to the plasma membrane to drive adhesion dynamics, cell migration and invasion. Here we assign a pivotal role for Rabgap1 (GAPCenA) in the recycling of endocytosed active ß1 integrins to the plasma membrane. The phosphotyrosine-binding (PTB) domain of Rabgap1 binds to the membrane-proximal NPxY motif in the cytoplasmic domain of ß1 integrin subunits on endosomes. Silencing Rabgap1 in mouse fibroblasts leads to the intracellular accumulation of active ß1 integrins, alters focal adhesion formation, and decreases cell migration and cancer cell invasion. Functionally, Rabgap1 facilitates active ß1 integrin recycling to the plasma membrane through attenuation of Rab11 activity. Taken together, our results identify Rabgap1 as an important factor for conformation-specific integrin trafficking and define the role of Rabgap1 in ß1-integrin-mediated cell migration in mouse fibroblasts and breast cancer cells.


Subject(s)
Endosomes , Integrin beta1 , Animals , Cell Adhesion , Cell Membrane , Cell Movement , GTPase-Activating Proteins , Integrin beta1/genetics , Integrins , Mice , Microtubule-Associated Proteins
11.
JBMR Plus ; 3(6): e10130, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31346562

ABSTRACT

Profilin 1 (Pfn1), a regulator of actin polymerization, controls cell movement in a context-dependent manner. Pfn1 supports the locomotion of most adherent cells by assisting actin-filament elongation, as has been shown in skeletal progenitor cells in our previous study. However, because Pfn1 has also been known to inhibit migration of certain cells, including T cells, by suppressing branched-end elongation of actin filaments, we hypothesized that its roles in osteoclasts may be different from that of osteoblasts. By investigating the osteoclasts in culture, we first verified that Pfn1-knockdown (KD) enhances bone resorption in preosteoclastic RAW264.7 cells, despite having a comparable number and size of osteoclasts. Pfn1-KD in bone marrow cells showed similar results. Mechanistically, Pfn1-KD osteoclasts appeared more mobile than in controls. In vivo, the osteoclast-specific conditional Pfn1-deficient mice (Pfn1-cKO) by CathepsinK-Cre driver demonstrated postnatal skeletal phenotype, including dwarfism, craniofacial deformities, and long-bone metaphyseal osteolytic expansion, by 8 weeks of age. Metaphyseal and diaphyseal femurs were drastically expanded with suppressed trabecular bone mass as indicated by µCT analysis. Histologically, TRAP-positive osteoclasts were increased at endosteal metaphysis to diaphysis of Pfn1-cKO mice. The enhanced movement of Pfn1-cKO osteoclasts in culture was associated with a slight increase in cell size and podosome belt length, as well as an increase in bone-resorbing activity. Our study, for the first time, demonstrated that Pfn1 has critical roles in inhibiting osteoclast motility and bone resorption, thereby contributing to essential roles in postnatal skeletal homeostasis. Our study also provides novel insight into understanding skeletal deformities in human disorders.

12.
J Cell Sci ; 131(22)2018 11 21.
Article in English | MEDLINE | ID: mdl-30333137

ABSTRACT

Integrins are α/ß heterodimers that interconvert between inactive and active states. In the active state the α/ß cytoplasmic domains recruit integrin-activating proteins and separate the transmembrane and cytoplasmic (TMcyto) domains (unclasped TMcyto). Conversely, in the inactive state the α/ß TMcyto domains bind integrin-inactivating proteins, resulting in the association of the TMcyto domains (clasped TMcyto). Here, we report the isolation of integrin cytoplasmic tail interactors using either lipid bicelle-incorporated integrin TMcyto domains (α5, αM, αIIb, ß1, ß2 and ß3 integrin TMcyto) or a clasped, lipid bicelle-incorporated αMß2 TMcyto. Among the proteins found to preferentially bind clasped rather than the isolated αM and ß2 subunits was L-plastin (LCP1, also known as plastin-2), which binds to and maintains the inactive state of αMß2 integrin in vivo and thereby regulates leukocyte adhesion to integrin ligands under flow. Our findings offer a global view on cytoplasmic proteins interacting with different integrins and provide evidence for the existence of conformation-specific integrin interactors.


Subject(s)
Cell Adhesion/physiology , Leukocytes/cytology , Leukocytes/metabolism , Macrophage-1 Antigen/metabolism , Microfilament Proteins/metabolism , Animals , Cell Membrane/metabolism , Cytoplasm/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Protein Binding , Protein Conformation , RAW 264.7 Cells
13.
PLoS One ; 13(7): e0200558, 2018.
Article in English | MEDLINE | ID: mdl-30001432

ABSTRACT

Focal adhesion kinase (FAK) is an intensely studied non-receptor tyrosine kinase with roles in cancer and other common human diseases. Despite the large interest in FAK, the in vivo contribution of FAK auto-phosphorylation site tyrosine (Y) 397 to FAK function is incompletely understood. To study FAK Y397 in vivo we analyzed mice with 'non-phosphorylatable' Y-to-phenylalanine (F) and 'phospho-mimicking' Y-to-glutamate (E) mutations in the germline. We found that FAK Y397F mice die early during embryogenesis with abnormal angiogenesis like FAK kinase-dead mice. When Y397 is mutated to a glutamate mice survive beyond mid-gestation like mice where Y397 is lost by deletion of FAK exon 15. In culture, defects in proliferation, invasion and gene expression were more severe with the FAK Y397F than with the FAK Y397E mutation despite the inability of FAK Y397E to bind SRC. Conditional expression of FAK Y397F or Y397E in unchallenged avascular epidermis, however, resulted in no appreciable phenotype. We conclude that FAK Y397 is required for the highly dynamic tissue remodeling during development but dispensable for normal homeostasis of avascular epidermis. In contrast to the Y397F mutation, FAK Y397E retains sufficient biological activity to allow for development beyond mid-gestation.


Subject(s)
Epidermis/enzymology , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Enzymologic/physiology , Homeostasis/physiology , Skin Physiological Phenomena , Amino Acid Substitution , Animals , Base Sequence , Focal Adhesion Kinase 1/genetics , Mice , Mice, Knockout , Mutation, Missense , Phosphorylation/physiology , Sequence Deletion , Tyrosine/genetics , Tyrosine/metabolism
14.
J Cell Physiol ; 233(1): 259-268, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28233307

ABSTRACT

Osteocytes are the most abundant cells in bone and regulate bone metabolism in coordination with osteoblasts and osteoclasts. However, the molecules that control osteocytes are still incompletely understood. Profilin1 is an actin-binding protein that is involved in actin polymerization. Osteocytes possess characteristic dendritic process formed based on actin cytoskeleton. Here, we examined the expression of profilin1 and its function in osteocytes. Profilin1 mRNA was expressed in osteocytic MLO-Y4 cells and its levels were gradually increased along with the time in culture. With regard to functional aspect, knockdown of profilin1 by siRNA enhanced BMP-induced increase in alkaline phosphatase expression levels in MLO-Y4 cells. Profilin1 knockdown suppressed the levels of dendritic processes and migration of MLO-Y4 cells. Since aging causes an increase in ROS in the body, we further examined the effects of hydrogen peroxide on the expression of profilin1. Hydrogen peroxide treatment increased the levels of profilin1 mRNA in MLO-Y4 cells in contrast to the decline in alkaline phosphatase. Profilin1 was expressed not only in MLO-Y4cells but also in the primary cultures of osteocytes. Importantly, profilin1 mRNA levels in primary cultures of osteocytes were higher than those in primary cultures of osteoblasts. To examine in vivo role of profilin1 in osteocytes, profilin1 was conditionally knocked out by using DMP1-cre and profilin1 floxed mice. This conditional deletion of profilin1 specifically in osteocytes resulted in reduction in the levels of bone volume and bone mineral density. These data indicate that profilin1 is expressed in osteocytes and regulates cell shape, migration and bone mass.


Subject(s)
Cell Movement , Cell Shape , Femur/metabolism , Osteocytes/metabolism , Profilins/metabolism , Alkaline Phosphatase/metabolism , Animals , Bone Density , Bone Remodeling , Cell Line , Cell Movement/drug effects , Cell Shape/drug effects , Femur/diagnostic imaging , Femur/drug effects , Gene Expression Regulation , Genotype , Hydrogen Peroxide/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Osteocytes/drug effects , Phenotype , Primary Cell Culture , Profilins/deficiency , Profilins/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Transfection , X-Ray Microtomography
15.
Cell Mol Life Sci ; 75(9): 1671-1685, 2018 05.
Article in English | MEDLINE | ID: mdl-29116364

ABSTRACT

Low density lipoprotein receptor-related protein (LRP) 1 modulates cell adhesion and motility under normal and pathological conditions. Previous studies documented that LRP1 binds several integrin receptors and mediates their trafficking to the cell surface and endocytosis. However, the mechanism by which LRP1 may regulate integrin activation remains unknown. Here we report that LRP1 promotes the activation and subsequent degradation of ß1 integrin and thus supports cell adhesion, spreading, migration and integrin signaling on fibronectin. LRP1 interacts with surface ß1 integrin, binds the integrin activator kindlin2 and stimulates ß1 integrin-kindlin2 complex formation. Specifically, serine 76 in the LRP1 cytoplasmic tail is crucial for the interaction with kindlin2, ß1 integrin activation and cell adhesion. Interestingly, a loss of LRP1 induces the accumulation of several integrin receptors on the cell surface. Following internalization, intracellular trafficking of integrins is driven by LRP1 in a protein kinase C- and class II myosin-dependent manner. Ultimately, LRP1 dictates the fate of endocytosed ß1 integrin by directing it down the pathway of lysosomal and proteasomal degradation. We propose that LRP1 mediates cell adhesion by orchestrating a multi-protein pathway to activate, traffic and degrade integrins. Thus, LRP1 may serve as a focal point in the integrin quality control system to ensure a firm connection to the extracellular matrix.


Subject(s)
Integrin beta1/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Animals , Cell Membrane/metabolism , Cells, Cultured , Embryo, Mammalian , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Mice , Mice, Knockout , Protein Transport/genetics , Proteolysis , Receptors, LDL/genetics , Receptors, LDL/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
16.
Cell ; 171(6): 1368-1382.e23, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29195076

ABSTRACT

Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.


Subject(s)
Bacterial Infections/immunology , Blood Platelets/immunology , Animals , Bacteria/classification , Blood Platelets/cytology , Blood Vessels/injuries , Blood Vessels/pathology , Calcium/metabolism , Cell Movement , Cell Polarity , Humans , Inflammation/immunology , Integrins/metabolism , Mice , Myosins/metabolism , Neutrophils/cytology
17.
J Cell Biol ; 216(11): 3785-3798, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28912124

ABSTRACT

Cell spreading requires the coupling of actin-driven membrane protrusion and integrin-mediated adhesion to the extracellular matrix. The integrin-activating adaptor protein kindlin-2 plays a central role for cell adhesion and membrane protrusion by directly binding and recruiting paxillin to nascent adhesions. Here, we report that kindlin-2 has a dual role during initial cell spreading: it binds paxillin via the pleckstrin homology and F0 domains to activate Rac1, and it directly associates with the Arp2/3 complex to induce Rac1-mediated membrane protrusions. Consistently, abrogation of kindlin-2 binding to Arp2/3 impairs lamellipodia formation and cell spreading. Our findings identify kindlin-2 as a key protein that couples cell adhesion by activating integrins and the induction of membrane protrusions by activating Rac1 and supplying Rac1 with the Arp2/3 complex.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Cell Adhesion , Cell Shape , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , Muscle Proteins/metabolism , Paxillin/metabolism , Pseudopodia/metabolism , Actin-Related Protein 2-3 Complex/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Genotype , Mice, Knockout , Muscle Proteins/deficiency , Muscle Proteins/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Paxillin/genetics , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Talin/deficiency , Talin/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
18.
Cell Cycle ; 16(24): 2366-2373, 2017.
Article in English | MEDLINE | ID: mdl-28699810

ABSTRACT

Profilin-1 (Pfn1) is an important actin-regulatory protein that is downregulated in human breast cancer and when forcibly elevated, it suppresses the tumor-initiating ability of triple-negative breast cancer cells. In this study, we demonstrate that Pfn1 overexpression reduces the stem-like phenotype (a key biologic feature associated with higher tumor-initiating potential) of MDA-MB-231 (MDA-231) triple-negative breast cancer cells. Interestingly, the stem-like trait of MDA-231 cells is also attenuated upon depletion of Pfn1. A comparison of cancer stem cell gene (CSC) gene expression signatures between depleted and elevated conditions of Pfn1 further suggest that Pfn1 may be somehow involved in regulating the expression of a few CSC-related genes including MUC1, STAT3, FZD7, and ITGB1. Consistent with the reduced stem-like phenotype associated with loss-of-function of Pfn1, xenograft studies showed lower tumor-initiating frequency of Pfn1-depleted MDA-231 cells compared to their control counterparts. In MMTV:PyMT mouse model, homozygous but not heterozygous deletion of Pfn1 gene leads to severe genetic mosaicism and positive selection of Pfn1-proficient tumor cells further supporting the contention that a complete lack of Pfn1 is likely not conducive for efficient tumor initiation capability of breast cancer cells. In summary, these findings suggest that the maintenance of optimal stemness and tumor-initiating ability of breast cancer cells requires a balanced expression of Pfn1.


Subject(s)
Profilins/metabolism , Animals , Carcinogens , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-1/metabolism , Neoplastic Stem Cells/metabolism , Profilins/antagonists & inhibitors , Profilins/genetics , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
20.
Blood ; 130(7): 847-858, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28615221

ABSTRACT

Trafficking of polymorphonuclear neutrophils (PMNs) during inflammation critically depends on the ß2 integrins lymphocyte function-associated antigen 1 (LFA-1) (CD11a/CD18) and macrophage-1 antigen (CD11b/CD18). Here, we identify coronin 1A (Coro1A) as a novel regulator of ß2 integrins that interacts with the cytoplasmic tail of CD18 and is crucial for induction of PMN adhesion and postadhesion events, including adhesion strengthening, spreading, and migration under flow conditions. Transition of PMN rolling to firm adhesion critically depends on Coro1A by regulating the accumulation of high-affinity LFA-1 in focal zones of adherent cells. Defective integrin affinity regulation in the genetic absence of Coro1A impairs leukocyte adhesion and extravasation in inflamed cremaster muscle venules in comparison with control animals. In a Helicobacter pylori mouse infection model, PMN infiltration into the gastric mucosa is dramatically reduced in Coro1A-/- mice, resulting in an attenuated gastric inflammation. Thus, Coro1A represents an important novel player in integrin biology, with key functions in PMN trafficking during innate immunity.


Subject(s)
4-Butyrolactone/analogs & derivatives , CD18 Antigens/metabolism , Cell Movement , Immunity, Innate , Neutrophils/cytology , Neutrophils/metabolism , 4-Butyrolactone/metabolism , Actins/metabolism , Animals , Calcium Signaling , Cell Adhesion , Gastritis/immunology , Gastritis/microbiology , Gastritis/pathology , Helicobacter pylori/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophage-1 Antigen/metabolism , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/metabolism , Rheology
SELECTION OF CITATIONS
SEARCH DETAIL
...