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










Publication year range
1.
J Neurosci ; 41(17): 3808-3821, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33727334

ABSTRACT

To assemble the functional circuits of the nervous system, the neuronal axonal growth cones must be precisely guided to their proper targets, which can be achieved through cell-surface guidance receptor activation by ligand binding in the periphery. We investigated the function of paxillin, a focal adhesion protein, as an essential growth cone guidance intermediary in the context of spinal lateral motor column (LMC) motor axon trajectory selection in the limb mesenchyme. Using in situ mRNA detection, we first show paxillin expression in LMC neurons of chick and mouse embryos at the time of spinal motor axon extension into the limb. Paxillin loss-of-function and gain-of-function using in ovo electroporation in chick LMC neurons, of either sex, perturbed LMC axon trajectory selection, demonstrating an essential role of paxillin in motor axon guidance. In addition, a neuron-specific paxillin deletion in mice led to LMC axon trajectory selection errors. We also show that knocking down paxillin attenuates the growth preference of LMC neurites against ephrins in vitro, and erythropoietin-producing human hepatocellular (Eph)-mediated retargeting of LMC axons in vivo, suggesting paxillin involvement in Eph-mediated LMC motor axon guidance. Finally, both paxillin knockdown and ectopic expression of a nonphosphorylable paxillin mutant attenuated the retargeting of LMC axons caused by Src overexpression, implicating paxillin as a Src target in Eph signal relay in this context. In summary, our findings demonstrate that paxillin is required for motor axon guidance and suggest its essential role in the ephrin-Eph signaling pathway resulting in motor axon trajectory selection.SIGNIFICANCE STATEMENT During the development of neural circuits, precise connections need to be established among neurons or between neurons and their muscle targets. A protein family found in neurons, Eph, is essential at different stages of neural circuit formation, including nerve outgrowth and pathfinding, and is proposed to mediate the onset and progression of several neurodegenerative diseases, such as Alzheimer's disease. To investigate how Ephs relay their signals to mediate nerve growth, we investigated the function of a molecule called paxillin and found it important for the development of spinal nerve growth toward their muscle targets, suggesting its role as an effector of Eph signals. Our work could thus provide new information on how neuromuscular connectivity is properly established during embryonic development.


Subject(s)
Axons/physiology , Paxillin/physiology , Spinal Cord/growth & development , Animals , Axon Guidance/physiology , Chick Embryo , Electroporation , Ephrins/physiology , Female , Gene Knockdown Techniques , Genes, src/genetics , Humans , Male , Mice , MicroRNAs/genetics , Motor Neurons/physiology , Mutation/genetics , Neurites/physiology , Spinal Cord/cytology
2.
FASEB J ; 35(3): e21369, 2021 03.
Article in English | MEDLINE | ID: mdl-33554392

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) translocates intracellularly and promotes cell migration, but how subcellular TM4SF5 traffic is regulated to guide cellular migration is unknown. We investigated the influences of the extracellular environment and intracellular signaling on the TM4SF5 traffic with regard to migration directionality. Cell adhesion to fibronectin (FN) but not poly-l-lysine enhanced the traffic velocity and straightness of the TM4SF5WT (but not palmitoylation-deficient mutant TM4SF5Pal- ) toward the leading edges, depending on tubulin acetylation. Acetylated-microtubules in SLAC2B-positive cells reached mostly the juxtanuclear regions, but reached-out toward the leading edges upon SLAC2B suppression. TM4SF5 expression caused SLAC2B not to be localized at the leading edges. TM4SF5 colocalization with HDAC6 depended on paxillin expression. The trimeric complex consisting of TM4SF5, HDAC6, and SLAC2B might, thus, be enriched at the perinuclear cytosols toward the leading edges. More TM4SF5WT translocation to the leading edges was possible when acetylated-microtubules reached the frontal edges following HDAC6 inhibition by paxillin presumably at new cell-FN adhesions, leading to persistent cell migration. Collectively, this study revealed that cell-FN adhesion and microtubule acetylation could control intracellular traffic of TM4SF5 vesicles to the leading edges via coordinated actions of paxillin, SLAC2B, and HDAC6, leading to TM4SF5-dependent cell migration.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Membrane/metabolism , Extracellular Matrix/physiology , Membrane Proteins/metabolism , Microtubules/metabolism , Acetylation , Cell Adhesion , Cell Movement , Fibronectins/physiology , Hep G2 Cells , Histone Deacetylase 6/physiology , Humans , Paxillin/physiology , Protein Transport
3.
Int Rev Cell Mol Biol ; 355: 1-52, 2020.
Article in English | MEDLINE | ID: mdl-32859368

ABSTRACT

The paxillin family of proteins, including paxillin, Hic-5, and leupaxin, are focal adhesion adaptor/scaffolding proteins which localize to cell-matrix adhesions and are important in cell adhesion and migration of both normal and cancer cells. Historically, the role of these proteins in regulating the actin cytoskeleton through focal adhesion-mediated signaling has been well documented. However, studies in recent years have revealed additional functions in modulating the microtubule and intermediate filament cytoskeletons to affect diverse processes including cell polarization, vesicle trafficking and mechanosignaling. Expression of paxillin family proteins in stromal cells is also important in regulating tumor cell migration and invasion through non-cell autonomous effects on the extracellular matrix. Both paxillin and Hic-5 can also influence gene expression through a variety of mechanisms, while their own expression is frequently dysregulated in various cancers. Accordingly, these proteins may serve as valuable targets for novel diagnostic and treatment approaches in cancer.


Subject(s)
Focal Adhesions/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Neoplasm Invasiveness , Neoplasms/metabolism , Paxillin/metabolism , Humans , Intracellular Signaling Peptides and Proteins/physiology , LIM Domain Proteins/physiology , Neoplasms/pathology , Paxillin/physiology , Signal Transduction
4.
J Cell Biol ; 219(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31816055

ABSTRACT

Talin, vinculin, and paxillin are core components of the dynamic link between integrins and actomyosin. Here, we study the mechanisms that mediate their activation and association using a mitochondrial-targeting assay, structure-based mutants, and advanced microscopy. As expected, full-length vinculin and talin are autoinhibited and do not interact with each other. However, contrary to previous models that propose a critical role for forces driving talin-vinculin association, our data show that force-independent relief of autoinhibition is sufficient to mediate their tight interaction. We also found that paxillin can bind to both talin and vinculin when either is inactive. Further experiments demonstrated that adhesions containing paxillin and vinculin can form without talin following integrin activation. However, these are largely deficient in exerting traction forces to the matrix. Our observations lead to a model whereby paxillin contributes to talin and vinculin recruitment into nascent adhesions. Activation of the talin-vinculin axis subsequently leads to the engagement with the traction force machinery and focal adhesion maturation.


Subject(s)
Fibroblasts/metabolism , Focal Adhesions/physiology , Paxillin/physiology , Stress, Mechanical , Talin/antagonists & inhibitors , Vinculin/physiology , Actin Cytoskeleton , Animals , Cells, Cultured , Fibroblasts/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Talin/metabolism
5.
J Cell Sci ; 130(22): 3801-3808, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28972134

ABSTRACT

Cell motility is required for diverse processes during immunity and inflammation. Classically, leukocyte motility is defined as an amoeboid type of migration, however some leukocytes, like macrophages, also employ a more mesenchymal mode of migration. Here, we sought to characterize the mechanisms that regulate neutrophil and macrophage migration in vivo by using real-time imaging of leukocyte motility within interstitial tissues in zebrafish larvae. Neutrophils displayed a rounded morphology and rapid protease-independent motility, lacked defined paxillin puncta, and had persistent rearward polarization of stable F-actin and the microtubule network. By contrast, macrophages displayed an elongated morphology with reduced speed and increased directional persistence and formed paxillin-containing puncta but had a less-defined polarization of the microtubule and actin networks. We also observed differential effects of protease inhibition, microtubule disruption and ROCK inhibition on the efficiency of neutrophil and macrophage motility. Taken together, our findings suggest that larval zebrafish neutrophils and macrophage display distinct modes of migration within interstitial tissues in vivo.


Subject(s)
Macrophages/physiology , Neutrophils/physiology , Animals , Cell Movement , Cell Polarity , Cell Shape , Cell Tracking , Larva/cytology , Microscopy, Fluorescence , Microscopy, Video , Microtubules/metabolism , Paxillin/physiology , Peptide Hydrolases/physiology , Zebrafish , Zebrafish Proteins/physiology
6.
Postepy Hig Med Dosw (Online) ; 70(0): 1087-1094, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27708212

ABSTRACT

Morphology of senescent cells is constantly changing at the molecular level, which in turn leads to disruption of their function. It is connected with reduced ability to synthesize extracellular matrix (ECM) and leads to the dysfunction of integrin adhesion molecules and adhesion clusters. In skin, these factors cause a loss of communication between the extracellular matrix and fibroblasts. This contributes to the appearance of signs of aging. The aim of this study is to draw attention to the very important molecule such as paxillin, which is an adaptor protein with mass of 68 kDa. This family of proteins includes Hic-5, PaxB and leupaxin. Paxillin binds to actin-binding proteins such as vinculin, actopaxin, and kinases (e.g. Integrin-linked kinase (ILK)). Moreover, it plays an important role in the integrity of the matrix, because it transduces transmembrane signaling between integrins and growth factors. Paxillin is a scaffold protein, activating the arrangement and organization of the cytoskeleton. Signaling through paxillin affects the long-term changes in gene expression, cell proliferation, and organization of the ECM. Correct functioning of the ECM is important for the wound healing processes and regeneration of tissues or tissue repair. Decrease or lack of paxillin expression results in changes in the structure and integrity of the ECM, which are manifested by aging of cells and organs. Restoration of the cellular matrix connections would be a significant element in the processes related to the anti-aging activities.


Subject(s)
Aging , Paxillin/metabolism , Signal Transduction , Skin/metabolism , Animals , Cytoskeletal Proteins/metabolism , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Paxillin/physiology , Protein Binding , Skin Physiological Phenomena
7.
J Physiol ; 594(17): 4879-900, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27038336

ABSTRACT

KEY POINTS: In airway smooth muscle, tension development caused by a contractile stimulus requires phosphorylation of the 20 kDa myosin light chain (MLC), which activates crossbridge cycling and the polymerization of a pool of submembraneous actin. The p21-activated kinases (Paks) can regulate the contractility of smooth muscle and non-muscle cells, and there is evidence that this occurs through the regulation of MLC phosphorylation. We show that Pak has no effect on MLC phosphorylation during the contraction of airway smooth muscle, and that it regulates contraction by mediating actin polymerization. We find that Pak phosphorylates the adhesion junction protein, paxillin, on Ser273, which promotes the formation of a signalling complex that activates the small GTPase, cdc42, and the actin polymerization catalyst, neuronal Wiskott-Aldrich syndrome protein (N-WASP). These studies demonstrate a novel role for Pak in regulating the contractility of smooth muscle by regulating actin polymerization. ABSTRACT: The p21-activated kinases (Pak) can regulate contractility in smooth muscle and other cell and tissue types, but the mechanisms by which Paks regulate cell contractility are unclear. In airway smooth muscle, stimulus-induced contraction requires phosphorylation of the 20 kDa light chain of myosin, which activates crossbridge cycling, as well as the polymerization of a small pool of actin. The role of Pak in airway smooth muscle contraction was evaluated by inhibiting acetylcholine (ACh)-induced Pak activation through the expression of a kinase inactive mutant, Pak1 K299R, or by treating tissues with the Pak inhibitor, IPA3. Pak inhibition suppressed actin polymerization and contraction in response to ACh, but it did not affect myosin light chain phosphorylation. Pak activation induced paxillin phosphorylation on Ser273; the paxillin mutant, paxillin S273A, inhibited paxillin Ser273 phosphorylation and inhibited actin polymerization and contraction. Immunoprecipitation analysis of tissue extracts and proximity ligation assays in dissociated cells showed that Pak activation and paxillin Ser273 phosphorylation triggered the formation of an adhesion junction signalling complex with paxillin that included G-protein-coupled receptor kinase-interacting protein (GIT1) and the cdc42 guanine exchange factor, ßPIX (Pak interactive exchange factor). Assembly of the Pak-GIT1-ßPIX-paxillin complex was necessary for cdc42 and neuronal Wiskott-Aldrich syndrome protein (N-WASP) activation, actin polymerization and contraction in response to ACh. RhoA activation was also required for the recruitment of Pak to adhesion junctions, Pak activation, paxillin Ser273 phosphorylation and paxillin complex assembly. These studies demonstrate a novel role for Pak in the regulation of N-WASP activation, actin dynamics and cell contractility.


Subject(s)
Actins/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Paxillin/physiology , Trachea/physiology , p21-Activated Kinases/physiology , Animals , Dogs , Female , Male , Myosin Light Chains/metabolism , Phosphorylation , Polymerization , Wiskott-Aldrich Syndrome Protein, Neuronal/physiology , rhoA GTP-Binding Protein/physiology
8.
Oncotarget ; 6(11): 8698-708, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25826088

ABSTRACT

Stabilization of Bcl-2 protein by paxillin (PXN)-mediated ERK activation was recently reported to cause an unfavorable response to 5-Fluorouracil-based chemotherapy. Here, we present evidence from cell and animal models to demonstrate that stabilization of Bcl-2 protein by phosphorylation at Serine 87 (pBcl-2-S87) via PXN-mediated ERK activation is responsible for cancer cell invasiveness and occurs via upregulation of MMP2 expression. Immunostainings of 190 tumors resected from colorectal cancer patients indicated that PXN expression was positively correlated with Bcl-2, pBcl-2-S87, and MMP2 expression. A positive correlation of pBcl-2-S87 with Bcl-2 and MMP2 was also observed in this study population. Patients with high PXN, Bcl-2, pBcl-2-S87, and MMP2 had poor overall survival (OS) and shorter relapse free survival (RFS). In conclusion, PXN promotes Bcl-2 phosphorylation at Serine 87 via PXN-mediated ERK activation, and its stabilization associated with increased tumor formation efficacy in mice and poor patient outcome in colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/pathology , Neoplasm Proteins/physiology , Paxillin/physiology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Benzimidazoles/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Dasatinib/therapeutic use , Enzyme Activation , Enzyme Induction , Extracellular Signal-Regulated MAP Kinases/physiology , Heterografts , Humans , Kaplan-Meier Estimate , Leupeptins/therapeutic use , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphorylation , Proportional Hazards Models , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Sulfonamides/therapeutic use
9.
J Cell Biol ; 206(3): 395-413, 2014 Aug 04.
Article in English | MEDLINE | ID: mdl-25070956

ABSTRACT

Polarized cell migration is essential for normal organism development and is also a critical component of cancer cell invasion and disease progression. Directional cell motility requires the coordination of dynamic cell-extracellular matrix interactions as well as repositioning of the Golgi apparatus, both of which can be controlled by the microtubule (MT) cytoskeleton. In this paper, we have identified a new and conserved role for the focal adhesion scaffold protein paxillin in regulating the posttranslational modification of the MT cytoskeleton through an inhibitory interaction with the α-tubulin deacetylase HDAC6. We also determined that through HDAC6-dependent regulation of the MT cytoskeleton, paxillin regulates both Golgi organelle integrity and polarized cell invasion and migration in both three-dimensional and two-dimensional matrix microenvironments. Importantly, these data reveal a fundamental role for paxillin in coordinating MT acetylation-dependent cell polarization and migration in both normal and transformed cells.


Subject(s)
Cell Movement , Extracellular Matrix/metabolism , Golgi Apparatus/ultrastructure , Histone Deacetylases/metabolism , Microtubules/metabolism , Paxillin/physiology , Protein Processing, Post-Translational , Acetylation , Animals , Cell Line, Tumor , Cell Polarity , Golgi Apparatus/metabolism , Histone Deacetylase 6 , Humans , Mice , NIH 3T3 Cells
10.
J Cell Sci ; 127(Pt 8): 1672-83, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24522185

ABSTRACT

Although a number of growth factors and receptors are known to control tumor angiogenesis, relatively little is known about the mechanism by which these factors influence the directional endothelial cell migration required for cancer microvessel formation. Recently, it has been shown that the focal adhesion protein paxillin is required for directional migration of fibroblasts in vitro. Here, we show that paxillin knockdown enhances endothelial cell migration in vitro and stimulates angiogenesis during normal development and in response to tumor angiogenic factors in vivo. Paxillin produces these effects by decreasing expression of neuropilin 2 (NRP2). Moreover, soluble factors secreted by tumors that stimulate vascular ingrowth, including vascular endothelial growth factor (VEGF), also decrease endothelial cell expression of paxillin and NRP2, and overexpression of NRP2 reverses these effects. These results suggest that the VEGF-paxillin-NRP2 pathway could represent a new therapeutic target for cancer and other angiogenesis-related diseases.


Subject(s)
Cell Movement , Human Umbilical Vein Endothelial Cells/physiology , Neovascularization, Pathologic/metabolism , Neuropilin-2/genetics , Paxillin/physiology , Animals , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Gene Expression , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Mice, Inbred C57BL , Neoplasm Transplantation , Neuropilin-2/metabolism , Retinal Vessels/physiopathology , Vascular Endothelial Growth Factor A/physiology
11.
Mol Cell ; 49(4): 668-79, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23317503

ABSTRACT

The HIV Nef protein recruits the polycomb protein Eed and mimics an integrin receptor signal for reasons that are not entirely clear. Here we demonstrate that Nef and Eed complex with the integrin effector paxillin to recruit and activate TNFα converting enzyme (TACE alias ADAM 17) and its close relative ADAM10. The activated proteases cleaved proTNFα and were shuttled into extracellular vesicles (EVs). Peripheral blood mononuclear cells that ingested these EVs released TNFα. Analyzing the mechanism, we found that Pak2, an established host cell effector of Nef, phosphorylated paxillin on Ser272/274 to induce TACE-paxillin association and shuttling into EVs via lipid rafts. Conversely, Pak1 phosphorylated paxillin on Ser258, which inhibited TACE association and lipid raft transfer. Interestingly, melanoma cells used an identical mechanism to shuttle predominantly ADAM10 into EVs. We conclude that HIV-1 and cancer cells exploit a paxillin/integrin-controlled mechanism to release TACE/ADAM10-containing vesicles, ensuring better proliferation/growth conditions in their microenvironment.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , Paxillin/physiology , nef Gene Products, Human Immunodeficiency Virus/physiology , p21-Activated Kinases/physiology , ADAM Proteins/blood , ADAM10 Protein , ADAM17 Protein , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Amyloid Precursor Protein Secretases/blood , Case-Control Studies , Enzyme Activation , HEK293 Cells , HIV Infections/blood , HIV Infections/enzymology , Heterogeneous-Nuclear Ribonucleoprotein K , Humans , Melanoma/blood , Melanoma/enzymology , Membrane Microdomains/enzymology , Membrane Proteins/blood , Mutagenesis, Site-Directed , Paxillin/genetics , Paxillin/metabolism , Phosphorylation , Polycomb Repressive Complex 2/metabolism , Protein Binding , Protein Kinase C-delta/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Protein Transport , Ribonucleoproteins/metabolism , Secretory Vesicles/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , p21-Activated Kinases/metabolism
12.
Carcinogenesis ; 33(9): 1769-81, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22678119

ABSTRACT

Endothelial tubular morphogenesis relies on an exquisite interplay of microtubule dynamics and actin remodeling to propel directed cell migration. Recently, the dynamicity and integrity of microtubules have been implicated in the trafficking and efficient translation of the mRNA for HIF-1α (hypoxia-inducible factor), the master regulator of tumor angiogenesis. Thus, microtubule-disrupting agents that perturb the HIF-1α axis and neovascularization cascade are attractive anticancer drug candidates. Here we show that EM011 (9-bromonoscapine), a microtubule-modulating agent, inhibits a spectrum of angiogenic events by interfering with endothelial cell invasion, migration and proliferation. Employing green-fluorescent transgenic zebrafish, we found that EM011 not only inhibited vasculogenesis but also disrupted preexisting vasculature. Mechanistically, EM011 caused proteasome-dependent, VHL-independent HIF-1α degradation and repressed expression of HIF-1α downstream targets, namely VEGF and survivin. Furthermore, EM011 inhibited membrane ruffling and impeded formation of filopodia, lamellipodia and stress fibers, which are critical for cell migration. These events were associated with a drug-mediated decrease in activation of Rho GTPases- RhoA, Cdc42 and Rac1, and correlated with a loss in the geometric precision of centrosome reorientation in the direction of movement. This is the first report to describe a previously unrecognized, antiangiogenic property of a noscapinoid, EM011, and provides evidence for novel anticancer strategies recruited by microtubule-modulating drugs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Dioxoles/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Isoquinolines/pharmacology , Animals , Cell Movement/drug effects , Cell Polarity/drug effects , Cells, Cultured , Centrosome/drug effects , Endothelial Cells/drug effects , Humans , Male , Mice , Microtubules/drug effects , Paxillin/physiology , Transcriptional Activation , rho GTP-Binding Proteins/metabolism
14.
Biochem Biophys Res Commun ; 418(4): 708-13, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22293201

ABSTRACT

Paxillin, a structural and signaling scaffold molecule in focal adhesions (FAs), is considered to be important in intracellular signaling transduction and the cell shape changes in response to cyclic stretching. However, the detailed role of paxillin in stretch-induced morphological changes of endothelial cells (ECs) has not fully determined until date. In this study, in order to understand the role of paxillin in the orientation of ECs exposed to cyclic stretching, we examined the time course of changes in the shape and distribution of FA proteins of paxillin knockdown ECs. Non-treated ECs subjected to 20% cyclic stretching at 0.5Hz oriented perpendicularly to the direction of stretching after 10min of exposure. On the other hand, the orientation of paxillin knockdown ECs was abolished at 10min, but it was observed after 60min of cyclic stretching exposure. Immunofluorescent microscopy revealed that accumulation and redistribution of FA proteins, including focal adhesion kinase (FAK) and integrin ß1, were observed at 10min of exposure to cyclic stretching in non-treated ECs. The accumulation of FAK and integrin ß1 was not prominent in paxillin knockdown ECs under static conditions and after 10min of exposure to cyclic stretching. However, we found that accumulation of FA proteins in paxillin knockdown ECs at 30 and 60min was similar to that in non-transfected ECs. Because paxillin is an adaptor protein offering binding sites for FAK and integrin ß1, which are critical molecules for the early signaling events of focal adhesion formation in ECs, these results suggest that paxillin is required for the early phase of EC orientation in response to cyclic stretching by scaffolding for accumulation of FA proteins.


Subject(s)
Cell Division , Focal Adhesions/physiology , Human Umbilical Vein Endothelial Cells/physiology , Paxillin/physiology , Cells, Cultured , Focal Adhesion Kinase 1/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/cytology , Humans , Integrin beta1/metabolism , Paxillin/genetics
15.
Cancer Res ; 71(22): 6965-75, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21948958

ABSTRACT

Tumor-associated macrophages promote tumor growth by stimulating angiogenesis and suppressing antitumor immunity. Thus, therapeutics that inhibit macrophage recruitment to tumors may provide new avenues for cancer therapy. In this study, we showed how chemoattractants stromal cell-derived growth factor 1 alpha (SDF-1α) and interleukin 1 beta (IL-1ß) collaborate with myeloid cell integrin-α4ß1 to promote tumor inflammation and growth. We found that SDF-1α and IL-1ß are highly expressed in the microenvironments of murine lung, pancreatic, and breast tumors; surprisingly, SDF-1α was expressed only by tumor cells, whereas IL-1ß was produced only by tumor-derived granulocytes and macrophages. In vivo, both factors directly recruited proangiogenic macrophages to tissues, whereas antagonists of both factors suppressed tumor inflammation, angiogenesis, and growth. Signals induced by IL-1ß and SDF-1α promoted the interaction of talin and paxillin with the cytoplasmic tails of integrin-α4ß1, thereby stimulating myeloid cell adhesion to endothelium in vitro and in vivo. Inhibition of integrin-α4ß1, SDF-1α, or IL-1ß was sufficient to block tumor inflammation and growth, and the combined blockade of these molecules greatly accentuated these effects. Furthermore, antagonists of integrin-α4ß1 inhibited chemotherapy-induced tumor inflammation and acted synergistically with chemotherapeutic agents to suppress tumor inflammation and growth. These results show that targeting myeloid cell recruitment mechanisms can be an effective approach to suppress tumor progression.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , Inflammation/prevention & control , Integrin alpha4beta1/antagonists & inhibitors , Interleukin-1beta/antagonists & inhibitors , Neoplasms/therapy , Animals , CD11b Antigen/analysis , Cell Movement , Chemokine CXCL12/physiology , Integrin alpha4beta1/physiology , Interleukin-1beta/physiology , Mice , Mice, Inbred C57BL , Myeloid Cells/physiology , Neoplasms/pathology , Paxillin/physiology , Talin/physiology
16.
Hepatogastroenterology ; 58(112): 1951-5, 2011.
Article in English | MEDLINE | ID: mdl-21940355

ABSTRACT

BACKGROUND/AIMS: The aim of this study was to investigate the effects of paxillin on proliferation, migration, invasion, adhesion and apoptosis of HCT-8 human colorectal cancer cells in vitro. METHODOLOGY: siRNA plasmids, overexpression wild-type plasmids and overexpression mutant plasmids were generated and transfected into HCT-8 cells. The expression of paxillin mRNA and protein was analyzed, and cell proliferation and adhesion were measured. Flow cytometry was used for cell sorting and detection of cell apoptosis. The invasive ability of HCT-8 cells was also observed. RESULTS: The proliferation, migration and invasive capacity of the HCT-8 cells transfected with siRNA paxillin plasmids were inhibited. Overexpression of wild-type paxillin plasmids promoted cell proliferation and also enhanced migration, invasive capacity and metastasis of the cancer cells. Overexpression of mutant paxillin plasmids inactivated the function of phosphorylation, inhibited cell migration and invasion capacity, but cell adhesion and proliferation had no significant difference compared with the normal group. CONCLUSIONS: These findings suggest that paxillin may play an essential role in the progression of colorectal cancer and RNAi targeting of paxillin may be a potential therapeutic strategy for the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms/pathology , Paxillin/physiology , Apoptosis , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/etiology , Colorectal Neoplasms/therapy , Disease Progression , Gene Expression Regulation , Humans , Neoplasm Invasiveness , Paxillin/antagonists & inhibitors , Paxillin/genetics , Plasmids , RNA, Small Interfering/genetics
17.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G601-11, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21757638

ABSTRACT

Paxillin is involved in the regulation of Helicobacter pylori-mediated gastric epithelial cell motility. We investigated the signaling pathways regulating H. pylori-induced paxillin phosphorylation and the effect of the H. pylori virulence factors cag pathogenicity island (PAI) and outer inflammatory protein (OipA) on actin stress fiber formation, cell phenotype, and IL-8 production. Gastric cell infection with live H. pylori induced site-specific phosphorylation of paxillin tyrosine (Y) 31 and Y118 in a time- and concentration-dependent manner. Activated paxillin localized in the cytoplasm at the tips of H. pylori-induced actin stress fibers. Isogenic oipA mutants significantly reduced paxillin phosphorylation at Y31 and Y118 and reduced actin stress fiber formation. In contrast, cag PAI mutants only inhibited paxillin Y118 phosphorylation. Silencing of epidermal growth factor receptor (EGFR), focal adhesion kinase (FAK), or protein kinase B (Akt) expression by small-interfering RNAs or inhibiting kinase activity of EGFR, Src, or phosphatidylinositol 3-kinase (PI3K) markedly reduced H. pylori-induced paxillin phosphorylation and morphologic alterations. Reduced FAK expression or lack of Src kinase activity suppressed H. pylori-induced IL-8 production. Compared with infection with the wild type, infection with the cag PAI mutant and oipA mutant reduced IL-8 production by nearly 80 and 50%. OipA-induced IL-8 production was FAK- and Src-dependent, although a FAK/Src-independent pathway for IL-8 production also exists, and the cag PAI may be mainly involved in this pathway. We propose paxillin as a novel cellular target for converging H. pylori-induced EGFR, FAK/Src, and PI3K/Akt signaling to regulate cytoskeletal reorganization and IL-8 production in part, thus contributing to the H. pylori-induced diseases.


Subject(s)
Actins/metabolism , Bacterial Outer Membrane Proteins/physiology , ErbB Receptors/physiology , Focal Adhesion Kinase 1/physiology , Helicobacter Infections/physiopathology , Helicobacter pylori/metabolism , Interleukin-8/biosynthesis , Paxillin/physiology , Phosphatidylinositol 3-Kinases/physiology , Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Cell Line , Epithelial Cells/microbiology , Genomic Islands , Helicobacter Infections/metabolism , Helicobacter pylori/genetics , Humans , Phosphorylation , Signal Transduction/physiology
18.
Mol Biol Cell ; 22(14): 2551-63, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21633109

ABSTRACT

We have identified the gene C28H8.6 (pxl-1) as the Caenorhabditis elegans orthologue of vertebrate paxillin. PXL-1 contains the four C-terminal LIM domains conserved in paxillin across all species and three of the five LD motifs found in the N-terminal half of most paxillins. In body wall muscle, PXL-1 antibodies and a full-length green fluorescent protein translational fusion localize to adhesion sites in the sarcomere, the functional repeat unit in muscle responsible for contraction. PXL-1 also localizes to ring-shaped structures near the sarcolemma in pharyngeal muscle corresponding to podosome-like sites of actin attachment. Our analysis of a loss-of-function allele of pxl-1, ok1483, shows that loss of paxillin leads to early larval arrested animals with paralyzed pharyngeal muscles and eventual lethality, presumably due to an inability to feed. We rescued the mutant phenotype by expressing paxillin solely in the pharynx and found that these animals survived and are essentially wild type in movement and body wall muscle structure. This indicates a differential requirement for paxillin in these two types of muscle. In pharyngeal muscle it is essential for contraction, whereas in body wall muscle it is dispensable for filament assembly, sarcomere stability, and ultimately movement.


Subject(s)
Caenorhabditis elegans/physiology , Muscle Contraction , Paxillin/physiology , Pharyngeal Muscles/physiology , Actin Cytoskeleton/ultrastructure , Amino Acid Motifs , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Carrier Proteins/metabolism , Cell Survival/genetics , Genes, Lethal , Intracellular Signaling Peptides and Proteins , Larva/genetics , Larva/physiology , Molecular Sequence Data , Mutation , Paxillin/genetics , Pharyngeal Muscles/metabolism , Phenotype , Protein Isoforms/genetics , Protein Structure, Tertiary , Sarcomeres/physiology
19.
J Urol ; 186(1): 295-301, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21600601

ABSTRACT

PURPOSE: We investigated the importance of EPLIN, a cytoskeletal associated protein implicated in cancer, in clinical prostate cancer and its role in the PC-3 prostate cancer cell line (ATCC™). MATERIALS AND METHODS: Full-length human EPLIN cDNA was cloned into a pEF6 expression vector and used to transfect the PC-3 human prostate cancer cell line. Cells over expressing EPLIN were termed PC-3(EPLIN EXP) while wild-type and empty pEF6 vector control cells were designated PC-3(WT) and PC-3(pEF6), respectively. The in vitro and in vivo impact of EPLIN on PC-3 cells was examined using a number of model assays. RESULTS: EPLIN over expression in PC-3 cells resulted in a decrease in the growth rate of this cell line (mean ± SD 0.6 ± 0.17 for PC-3(pEF6) cells vs 0.33 ± 0.01 for PC-3(EPLIN EXP) cells, p <0.01). PC-3(EPLIN EXP) cells were significantly less able to adhere to extracellular matrix than control cells (mean 61.0 ± 12.4 vs 102.8 ± 20.7, p = 0.028). Immunofluorescence staining showed an increased staining profile for paxillin in PC-3(EPLIN EXP) cells compared to wild-type cells. CONCLUSIONS: EPLIN over expression in the PC-3 cell line resulted in decreased in vivo and in vitro growth potential together with decreased cell invasiveness and ability to adhere to extracellular matrix, and enhanced paxillin staining. This further highlights the importance of EPLIN in regulating prostate cancer cell growth and aggressiveness, and suggests a possible connection between EPLIN and paxillin.


Subject(s)
Cytoskeletal Proteins/physiology , Prostatic Neoplasms/pathology , Cell Proliferation , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Invasiveness , Paxillin/genetics , Paxillin/physiology , Tumor Cells, Cultured
20.
J Neurosci ; 31(12): 4535-43, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21430154

ABSTRACT

Sensory hair cells of the inner ear are the mechanoelectric transducers of sound and head motion. In mammals, damage to sensory hair cells leads to hearing or balance deficits. Nonmammalian vertebrates such as birds can regenerate hair cells after injury. In a previous study, we characterized transcription factor gene expression during chicken hair cell regeneration. In those studies, a laser microbeam or ototoxic antibiotics were used to damage the sensory epithelia (SE). The current study focused on 27 genes that were upregulated in regenerating SEs compared to untreated SEs in the previous study. Those genes were knocked down by siRNA to determine their requirement for supporting cell proliferation and to measure resulting changes in the larger network of gene expression. We identified 11 genes necessary for proliferation and also identified novel interactive relationships between many of them. Defined components of the WNT, PAX, and AP1 pathways were shown to be required for supporting cell proliferation. These pathways intersect on WNT4, which is also necessary for proliferation. Among the required genes, the CCAAT enhancer binding protein, CEBPG, acts downstream of Jun Kinase and JUND in the AP1 pathway. The WNT coreceptor LRP5 acts downstream of CEBPG, as does the transcription factor BTAF1. Both of these genes are also necessary for supporting cell proliferation. This is the first large-scale screen of its type and suggests an important intersection between the AP1 pathway, the PAX pathway, and WNT signaling in the regulation of supporting cell proliferation during inner ear hair cell regeneration.


Subject(s)
Ear, Inner/physiology , Hair Cells, Auditory, Inner/physiology , Nerve Regeneration/physiology , RNA Interference/physiology , Transcription Factors/genetics , Animals , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/physiology , Cell Proliferation , Chickens , Epithelium/physiology , Gene Knockdown Techniques , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/physiology , Lasers , Microarray Analysis , Paxillin/genetics , Paxillin/physiology , Saccule and Utricle/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/physiology , Transcription Factor TFIID/genetics , Transcription Factor TFIID/physiology , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt Proteins/physiology , Wnt4 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...