Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Life Sci Alliance ; 6(8)2023 08.
Article in English | MEDLINE | ID: mdl-37230801

ABSTRACT

Repeated injury of the lung epithelium is proposed to be the main driver of idiopathic pulmonary fibrosis (IPF). However, available therapies do not specifically target the epithelium and human models of fibrotic epithelial damage with suitability for drug discovery are lacking. We developed a model of the aberrant epithelial reprogramming observed in IPF using alveolar organoids derived from human-induced pluripotent stem cells stimulated with a cocktail of pro-fibrotic and inflammatory cytokines. Deconvolution of RNA-seq data of alveolar organoids indicated that the fibrosis cocktail rapidly increased the proportion of transitional cell types including the KRT5 - /KRT17 + aberrant basaloid phenotype recently identified in the lungs of IPF patients. We found that epithelial reprogramming and extracellular matrix (ECM) production persisted after removal of the fibrosis cocktail. We evaluated the effect of the two clinically approved compounds for IPF, nintedanib and pirfenidone, and found that they reduced the expression of ECM and pro-fibrotic mediators but did not completely reverse epithelial reprogramming. Thus, our system recapitulates key aspects of IPF and is a promising system for drug discovery.


Subject(s)
Idiopathic Pulmonary Fibrosis , Pluripotent Stem Cells , Humans , Alveolar Epithelial Cells/metabolism , Lung/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Fibrosis , Pluripotent Stem Cells/metabolism , Organoids/metabolism
2.
Respir Res ; 24(1): 51, 2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36788603

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease that affects 3 million people worldwide. Senescence and small extracellular vesicles (sEVs) have been implicated in the pathogenesis of IPF, although how sEVs promote disease remains unclear. Here, we profile sEVs from bronchial epithelial cells and determine small RNA (smRNA) content. METHODS: Conditioned media was collected and sEVs were isolated from normal human bronchial epithelial cells (NHBEs) and IPF-diseased human bronchial epithelial cells (DHBEs). RESULTS: Increased sEV release from DHBEs compared to NHBEs (n = 4; p < 0.05) was detected by nanoparticle tracking analysis. NHBEs co-cultured with DHBE-derived sEVs for 72 h expressed higher levels of SA-ß-Gal and γH2AX protein, p16 and p21 RNA and increased secretion of IL6 and IL8 proteins (all n = 6-8; p < 0.05). sEVs were also co-cultured with healthy air-liquid interface (ALI) cultures and similar results were observed, with increases in p21 and p16 gene expression and IL6 and IL8 (basal and apical) secretion (n = 6; p < 0.05). Transepithelial electrical resistance (TEER) measurements, a reflection of epithelial barrier integrity, were decreased upon the addition of DHBE-derived sEVs (n = 6; p < 0.05). smRNA-sequencing identified nineteen significantly differentially expressed miRNA in DHBE-derived sEVs compared to NHBE-derived sEVs, with candidate miRNAs validated by qPCR (all n = 5; p < 0.05). Four of these miRNAs were upregulated in NHBEs co-cultured with DHBE-derived sEVs and three in healthy ALI cultures co-cultured with DHBE-derived sEVs (n = 3-4; p < 0.05). CONCLUSIONS: This data demonstrates that DHBE-derived sEVs transfer senescence to neighbouring healthy cells, promoting the disease state in IPF.


Subject(s)
Extracellular Vesicles , Idiopathic Pulmonary Fibrosis , MicroRNAs , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Extracellular Vesicles/metabolism
3.
J Allergy Clin Immunol ; 150(6): 1415-1426.e9, 2022 12.
Article in English | MEDLINE | ID: mdl-35917932

ABSTRACT

BACKGROUND: Patients with asthma often suffer from frequent respiratory viral infections and reduced virus clearance. Lung resident memory T cells provide rapid protection against viral reinfections. OBJECTIVE: Because the development of resident memory T cells relies on the lung microenvironment, we investigated the impact of allergen sensitization on the development of virus-specific lung resident memory T cells and viral clearance. METHODS: Mice were sensitized with house dust mite extract followed by priming with X47 and a subsequent secondary influenza infection. Antiviral memory T-cell response and protection to viral infection was assessed before and after secondary influenza infection, respectively. Gene set variation analysis was performed on data sets from the U-BIOPRED asthma cohort using an IFN-γ-induced epithelial cell signature and a tissue resident memory T-cell signature. RESULTS: Viral loads were higher in lungs of sensitized compared with nonsensitized mice after secondary infection, indicating reduced virus clearance. X47 priming induced fewer antiviral lung resident memory CD8 T cells and resulted in lower pulmonary IFN-γ levels in the lungs of sensitized as compared with nonsensitized mice. Using data from the U-BIOPRED cohort, we found that patients with enrichment of epithelial IFN-γ-induced genes in nasal brushings and bronchial biopsies were also enriched in resident memory T-cell-associated genes, had more epithelial CD8 T cells, and reported significantly fewer exacerbations. CONCLUSIONS: The allergen-sensitized lung microenvironment interferes with the formation of antiviral resident memory CD8 T cells in lungs and virus clearance. Defective antiviral memory response might contribute to increased susceptibility of patients with asthma to viral exacerbations.


Subject(s)
Influenza, Human , Memory T Cells , Mice , Animals , Humans , Lung , CD8-Positive T-Lymphocytes , Allergens
4.
JCI Insight ; 7(16)2022 08 22.
Article in English | MEDLINE | ID: mdl-35993367

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unmet medical need. It is characterized by formation of scar tissue leading to a progressive and irreversible decline in lung function. IPF is associated with repeated injury, which may alter the composition of the extracellular matrix (ECM). Here, we demonstrate that IPF patient-derived pulmonary ECM drives profibrotic response in normal human lung fibroblasts (NHLF) in a 3D spheroid assay. Next, we reveal distinct alterations in composition of the diseased ECM, identifying potentially novel associations with IPF. Growth differentiation factor 15 (GDF15) was identified among the most significantly upregulated proteins in the IPF lung-derived ECM. In vivo, GDF15 neutralization in a bleomycin-induced lung fibrosis model led to significantly less fibrosis. In vitro, recombinant GDF15 (rGDF15) stimulated α smooth muscle actin (αSMA) expression in NHLF, and this was mediated by the activin receptor-like kinase 5 (ALK5) receptor. Furthermore, in the presence of rGDF15, the migration of NHLF in collagen gel was reduced. In addition, we observed a cell type-dependent effect of GDF15 on the expression of cell senescence markers. Our data suggest that GDF15 mediates lung fibrosis through fibroblast activation and differentiation, implicating a potential direct role of this matrix-associated cytokine in promoting aberrant cell responses in disease.


Subject(s)
Extracellular Matrix , Growth Differentiation Factor 15 , Idiopathic Pulmonary Fibrosis , Extracellular Matrix/metabolism , Fibrosis/genetics , Fibrosis/metabolism , Growth Differentiation Factor 15/biosynthesis , Growth Differentiation Factor 15/genetics , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Signal Transduction
5.
Sci Rep ; 11(1): 21584, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732748

ABSTRACT

The unfolded protein response (UPR) is a direct consequence of cellular endoplasmic reticulum (ER) stress and a key disease driving mechanism in IPF. The resolution of the UPR is directed by PPP1R15A (GADD34) and leads to the restoration of normal ribosomal activity. While the role of PPP1R15A has been explored in lung epithelial cells, the role of this UPR resolving factor has yet to be explored in lung mesenchymal cells. The objective of the current study was to determine the expression and role of PPP1R15A in IPF fibroblasts and in a bleomycin-induced lung fibrosis model. A survey of IPF lung tissue revealed that PPP1R15A expression was markedly reduced. Targeting PPP1R15A in primary fibroblasts modulated TGF-ß-induced fibroblast to myofibroblast differentiation and exacerbated pulmonary fibrosis in bleomycin-challenged mice. Interestingly, the loss of PPP1R15A appeared to promote lung fibroblast senescence. Taken together, our findings demonstrate the major role of PPP1R15A in the regulation of lung mesenchymal cells, and regulation of PPP1R15A may represent a novel therapeutic strategy in IPF.


Subject(s)
Cellular Senescence , Fibrosis/metabolism , Protein Phosphatase 1/genetics , Unfolded Protein Response , Aged , Animals , Bleomycin , Cell Differentiation , Cell Proliferation , Endoplasmic Reticulum Stress , Female , Fibroblasts/metabolism , Genotype , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Indoles/pharmacology , Lung/metabolism , Male , Mesoderm/cytology , Mice , Middle Aged , Morpholines/pharmacology , Protein Phosphatase 1/physiology , Sequence Analysis, RNA , Transforming Growth Factor beta/metabolism
6.
Nature ; 597(7877): 527-532, 2021 09.
Article in English | MEDLINE | ID: mdl-34375979

ABSTRACT

Genome-wide association studies have uncovered thousands of common variants associated with human disease, but the contribution of rare variants to common disease remains relatively unexplored. The UK Biobank contains detailed phenotypic data linked to medical records for approximately 500,000 participants, offering an unprecedented opportunity to evaluate the effect of rare variation on a broad collection of traits1,2. Here we study the relationships between rare protein-coding variants and 17,361 binary and 1,419 quantitative phenotypes using exome sequencing data from 269,171 UK Biobank participants of European ancestry. Gene-based collapsing analyses revealed 1,703 statistically significant gene-phenotype associations for binary traits, with a median odds ratio of 12.4. Furthermore, 83% of these associations were undetectable via single-variant association tests, emphasizing the power of gene-based collapsing analysis in the setting of high allelic heterogeneity. Gene-phenotype associations were also significantly enriched for loss-of-function-mediated traits and approved drug targets. Finally, we performed ancestry-specific and pan-ancestry collapsing analyses using exome sequencing data from 11,933 UK Biobank participants of African, East Asian or South Asian ancestry. Our results highlight a significant contribution of rare variants to common disease. Summary statistics are publicly available through an interactive portal ( http://azphewas.com/ ).


Subject(s)
Biological Specimen Banks , Databases, Genetic , Disease/genetics , Exome/genetics , Genetic Variation/genetics , Adult , Aged , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Phenotype , Proteins/chemistry , Proteins/genetics , United Kingdom , Exome Sequencing
7.
Commun Biol ; 4(1): 392, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758299

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal disorder characterised by progressive, destructive lung scarring. Despite substantial progress, the genetic determinants of this disease remain incompletely defined. Using whole genome and whole exome sequencing data from 752 individuals with sporadic IPF and 119,055 UK Biobank controls, we performed a variant-level exome-wide association study (ExWAS) and gene-level collapsing analyses. Our variant-level analysis revealed a novel association between a rare missense variant in SPDL1 and IPF (NM_017785.5:g.169588475 G > A p.Arg20Gln; p = 2.4 × 10-7, odds ratio = 2.87, 95% confidence interval: 2.03-4.07). This signal was independently replicated in the FinnGen cohort, which contains 1028 cases and 196,986 controls (combined p = 2.2 × 10-20), firmly associating this variant as an IPF risk allele. SPDL1 encodes Spindly, a protein involved in mitotic checkpoint signalling during cell division that has not been previously described in fibrosis. To the best of our knowledge, these results highlight a novel mechanism underlying IPF, providing the potential for new therapeutic discoveries in a disease of great unmet need.


Subject(s)
Cell Cycle Proteins/genetics , Idiopathic Pulmonary Fibrosis/genetics , Mutation, Missense , Aged , Case-Control Studies , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Male , Phenotype , Exome Sequencing
8.
PLoS One ; 15(12): e0243807, 2020.
Article in English | MEDLINE | ID: mdl-33332401

ABSTRACT

Induced pluripotent stem cells (iPSC) offer the possibility to generate diverse disease-relevant cell types, from any genetic background with the use of cellular reprogramming and directed differentiation. This provides a powerful platform for disease modeling, drug screening and cell therapeutics. The critical question is how the differentiated iPSC-derived cells translate to their primary counterparts. Our refinement of a published differentiation protocol produces a CD14+ monocytic lineage at a higher yield, in a smaller format and at a lower cost. These iPSC-derived monocytes can be further differentiated into macrophages or dendritic cells (DC), both with similar morphological and functional profiles as compared to their primary counterparts. Transcriptomic analysis of iPSC-derived cells at different stages of differentiation as well as comparison to their blood-derived counterparts demonstrates a complete switch of iPSCs to cells expressing a monocyte, macrophage or DC specific gene profile. iPSC-derived macrophages respond to LPS treatment by inducing expression of classic macrophage pro-inflammatory response markers. Interestingly, though iPSC-derived DC show similarities to monocyte derived DC, they are more similar transcriptionally to a newly described subpopulation of AXL+ DC. Thus, our study provides a detailed and accurate profile of iPSC-derived monocytic lineage cells.


Subject(s)
Dendritic Cells/cytology , Induced Pluripotent Stem Cells/cytology , Macrophages/cytology , Transcription, Genetic , Cell Differentiation , Cell Lineage , Dendritic Cells/metabolism , Humans , Macrophages/metabolism
9.
Sci Rep ; 9(1): 20407, 2019 12 31.
Article in English | MEDLINE | ID: mdl-31892708

ABSTRACT

Bleomycin hydrolase (BLMH) is a well-conserved cysteine protease widely expressed in several mammalian tissues. In skin, which contains high levels of BLMH, this protease is involved in the degradation of citrullinated filaggrin monomers into free amino acids important for skin hydration. Interestingly, the expression and activity of BLMH is reduced in patients with atopic dermatitis (AD) and psoriasis, and BLMH knockout mice acquire tail dermatitis. Apart from its already known function, we have discovered a novel role of BLMH in the regulation of inflammatory chemokines and wound healing. We show that lowered BLMH levels in keratinocytes result in increased release of the pro-inflammatory chemokines CXCL8 and GROα, which are upregulated in skin from AD patients compared to healthy individuals. Conditioned media from keratinocytes expressing low levels of BLMH increased chemotaxis by neutrophils and caused a delayed wound healing in the presence of low-level TNFα. This defective wound healing was improved by blocking the shared receptor of CXCL8 and GROα, namely CXCR2, using a specific receptor antagonist. Collectively, our results present a novel function of BLMH in regulating the secretion of chemokines involved in inflammation and wound healing in human keratinocytes.


Subject(s)
Chemokines/metabolism , Cysteine Endopeptidases/metabolism , Inflammation/metabolism , Keratinocytes/metabolism , Wound Healing/physiology , Cell Line , Chemotaxis/drug effects , Chemotaxis/physiology , Culture Media, Conditioned , Cysteine Endopeptidases/genetics , Filaggrin Proteins , Humans , Inflammation/genetics , Keratinocytes/drug effects , Skin/drug effects , Skin/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Wound Healing/drug effects
10.
Proc Natl Acad Sci U S A ; 114(30): E6250-E6259, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28698364

ABSTRACT

Continuous contraction-relaxation cycles of the heart require strong and stable connections of cardiac myocytes (CMs) with the extracellular matrix (ECM) to preserve sarcolemmal integrity. CM attachment to the ECM is mediated by integrin complexes localized at the muscle adhesion sites termed costameres. The ubiquitously expressed cytoskeletal protein talin (Tln) is a component of muscle costameres that links integrins ultimately to the sarcomere. There are two talin genes, Tln1 and Tln2. Here, we tested the function of these two Tln forms in myocardium where Tln2 is the dominant isoform in postnatal CMs. Surprisingly, global deletion of Tln2 in mice caused no structural or functional changes in heart, presumably because CM Tln1 became up-regulated. Tln2 loss increased integrin activation, although levels of the muscle-specific ß1D-integrin isoform were reduced by 50%. With this result, we produced mice that had simultaneous loss of both CM Tln1 and Tln2 and found that cardiac dysfunction occurred by 4 wk with 100% mortality by 6 mo. ß1D integrin and other costameric proteins were lost from the CMs, and membrane integrity was compromised. Given that integrin protein reduction occurred with Tln loss, rescue of the phenotype was attempted through transgenic integrin overexpression, but this could not restore WT CM integrin levels nor improve heart function. Our results show that CM Tln2 is essential for proper ß1D-integrin expression and that Tln1 can substitute for Tln2 in preserving heart function, but that loss of all Tln forms from the heart-muscle cell leads to myocyte instability and a dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/genetics , Integrin beta1/metabolism , Myocytes, Cardiac/metabolism , Talin/genetics , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Mice , Myocardium/pathology , Myocytes, Cardiac/physiology , Talin/metabolism , Talin/physiology
12.
PLoS One ; 12(3): e0173115, 2017.
Article in English | MEDLINE | ID: mdl-28248992

ABSTRACT

BACKGROUND: BET proteins (BRD2, BRD3, BRDT and BRD4) belong to the family of bromodomain containing proteins, which form a class of transcriptional co-regulators. BET proteins bind to acetylated lysine residues in the histones of nucleosomal chromatin and function either as co-activators or co-repressors of gene expression. An imbalance between HAT and HDAC activities resulting in hyperacetylation of histones has been identified in COPD. We hypothesized that pan-BET inhibitor (JQ1) treatment of BET protein interactions with hyperacetylated sites in the chromatin will regulate excessive activation of pro-inflammatory genes in key inflammatory drivers of alveolar macrophages (AM) in COPD. METHODS AND FINDINGS: Transcriptome analysis of AM from COPD patients indicated up-regulation of macrophage M1 type genes upon LPS stimulation. Pan-BET inhibitor JQ1 treatment attenuated expression of multiple genes, including pro-inflammatory cytokines and regulators of innate and adaptive immune cells. We demonstrated for the first time that JQ1 differentially modulated LPS-induced cytokine release from AM or peripheral blood mononuclear cells (PBMC) of COPD patients compared to PBMC of healthy controls. Using the BET regulated gene signature, we identified a subset of COPD patients, which we propose to benefit from BET inhibition. CONCLUSIONS: This work demonstrates that the effects of pan-BET inhibition through JQ1 treatment of inflammatory cells differs between COPD patients and healthy controls, and the expression of BET protein regulated genes is altered in COPD. These findings provide evidence of histone hyperacetylation as a mechanism driving chronic inflammatory changes in COPD.


Subject(s)
Chromatin Assembly and Disassembly , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Transcription Factors/metabolism , Azepines/pharmacology , Case-Control Studies , Cell Cycle Proteins , Cells, Cultured , Chromatin/drug effects , Chromatin/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Monocytes/drug effects , Monocytes/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Pulmonary Disease, Chronic Obstructive/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , Triazoles/pharmacology
13.
J Clin Invest ; 124(3): 1098-113, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24531545

ABSTRACT

Podocytes are specialized actin-rich epithelial cells that line the kidney glomerular filtration barrier. The interface between the podocyte and the glomerular basement membrane requires integrins, and defects in either α3 or ß1 integrin, or the α3ß1 ligand laminin result in nephrotic syndrome in murine models. The large cytoskeletal protein talin1 is not only pivotal for integrin activation, but also directly links integrins to the actin cytoskeleton. Here, we found that mice lacking talin1 specifically in podocytes display severe proteinuria, foot process effacement, and kidney failure. Loss of talin1 in podocytes caused only a modest reduction in ß1 integrin activation, podocyte cell adhesion, and cell spreading; however, the actin cytoskeleton of podocytes was profoundly altered by the loss of talin1. Evaluation of murine models of glomerular injury and patients with nephrotic syndrome revealed that calpain-induced talin1 cleavage in podocytes might promote pathogenesis of nephrotic syndrome. Furthermore, pharmacologic inhibition of calpain activity following glomerular injury substantially reduced talin1 cleavage, albuminuria, and foot process effacement. Collectively, these findings indicate that podocyte talin1 is critical for maintaining the integrity of the glomerular filtration barrier and provide insight into the pathogenesis of nephrotic syndrome.


Subject(s)
Glomerular Filtration Barrier/pathology , Nephrotic Syndrome/metabolism , Podocytes/metabolism , Talin/physiology , Actin Cytoskeleton/metabolism , Animals , Calpain/metabolism , Cell Adhesion , Cells, Cultured , Focal Adhesions/metabolism , Humans , Integrin beta1/metabolism , Mice , Mice, Knockout , Nephrotic Syndrome/pathology , Proteinuria/genetics , Proteinuria/metabolism , Proteolysis , Renal Insufficiency/genetics , Renal Insufficiency/metabolism
14.
J Biol Chem ; 288(6): 4252-64, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23266827

ABSTRACT

Integrins are adhesive, signaling, and mechanotransduction proteins. Talin (Tln) activates integrins and links it to the actin cytoskeleton. Vertebrates contain two talin genes, tln1 and tln2. How Tln1 and Tln2 function in cardiac myocytes (CMs) is unknown. Tln1 and Tln2 expression were evaluated in the normal embryonic and adult mouse heart as well as in control and failing human adult myocardium. Tln1 function was then tested in the basal and mechanically stressed myocardium after cardiomyocyte-specific excision of the Tln1 gene. During embryogenesis, both Tln forms are highly expressed in CMs, but in the mature heart Tln2 becomes the main Tln isoform, localizing to the costameres. Tln1 expression is minimal in the adult CM. With pharmacological and mechanical stress causing hypertrophy, Tln1 is up-regulated in CMs and is specifically detected at costameres, suggesting its importance in the compensatory response to CM stress. In human failing heart, CM Tln1 also increases compared with control samples from normal functioning myocardium. To directly test Tln1 function in CMs, we generated CM-specific Tln1 knock-out mice (Tln1cKO). Tln1cKO mice showed normal basal cardiac structure and function but when subjected to pressure overload showed blunted hypertrophy, less fibrosis, and improved cardiac function versus controls. Acute responses of ERK1/2, p38, Akt, and glycogen synthase kinase 3 after mechanical stress were strongly blunted in Tln1cKO mice. Given these results, we conclude that Tln1 and Tln2 have distinct functions in the myocardium. Our data show that reduction of CM Tln1 expression can lead to improved cardiac remodeling following pressure overload.


Subject(s)
Cardiomegaly/metabolism , Myocardium/metabolism , Talin/biosynthesis , Adult , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Humans , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological/genetics , Talin/genetics , Up-Regulation/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Mol Cell Biol ; 33(4): 830-44, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230271

ABSTRACT

To determine talin1's role in osteoclasts, we mated TLN1(fl/fl) mice with those expressing cathepsin K-Cre (CtsK-TLN1) to delete the gene in mature osteoclasts or with lysozyme M-Cre (LysM-TLN1) mice to delete TLN1 in all osteoclast lineage cells. Absence of TLN1 impairs macrophage colony-stimulating factor (M-CSF)-stimulated inside-out integrin activation and cytoskeleton organization in mature osteoclasts. Talin1-deficient precursors normally express osteoclast differentiation markers when exposed to M-CSF and receptor activator of nuclear factor κB (RANK) ligand but attach to substrate and migrate poorly, arresting their development into mature resorptive cells. In keeping with inhibited resorption, CtsK-TLN1 mice exhibit an ∼5-fold increase in bone mass. Osteoclast-specific deletion of Rap1 (CtsK-Rap1), which promotes talin/ß integrin recognition, yields similar osteopetrotic mice. The fact that the osteopetrosis of CtsK-TLN1 and CtsK-Rap1 mice is substantially more severe than that of those lacking αvß3 is likely due to added failed activation of ß1 integrins. In keeping with osteoclast dysfunction, mice in whom talin is deleted late in the course of osteoclastogenesis are substantially protected from ovariectomy-induced osteoporosis and the periarticular osteolysis attending inflammatory arthritis. Thus, talin1 and Rap1 are critical for resorptive function, and their selective inhibition in mature osteoclasts retards pathological bone loss.


Subject(s)
Osteoclasts/cytology , Osteoclasts/pathology , Talin/metabolism , rap GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Female , Gene Deletion , Integrin alphaVbeta3/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Osteoclasts/metabolism , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Talin/genetics , rap GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/genetics
16.
Biochem Biophys Res Commun ; 426(2): 190-5, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22925892

ABSTRACT

Mice homozygous for several Tln2 gene targeted alleles are viable and fertile. Here we show that although the expression of talin2 protein is drastically reduced in muscle from these mice, other tissues continue to express talin2 albeit at reduced levels. We therefore generated a Tln2 allele lacking the entire coding sequence (Tln2(cd)). Tln2(cd/cd) mice were viable and fertile, and the genotypes of Tln2(cd/+) intercrosses were at the expected Mendelian ratio. Tln2(cd/cd) mice showed no major difference in body mass or the weight of the major organs compared to wild-type, although they displayed a mildly dystrophic phenotype. Moreover, Tln2(cd/cd) mouse embryo fibroblasts showed no obvious defects in cell adhesion, migration or proliferation. However, the number of Tln2(cd/cd) pups surviving to adulthood was variable suggesting that such mice have an underlying defect.


Subject(s)
Embryonic Development/genetics , Fertility , Talin/physiology , Animals , Body Weight , Cell Adhesion , Cell Movement , Cell Proliferation , Female , Fibroblasts/physiology , Gene Deletion , Male , Mice , Mice, Knockout , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Talin/genetics
17.
Blood ; 119(18): 4275-82, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22431571

ABSTRACT

In inflammation, neutrophils and other leukocytes roll along the microvascular endothelium before arresting and transmigrating into inflamed tissues. Arrest requires conformational activation of the integrin lymphocyte function-associated antigen-1 (LFA-1). Mutations of the FERMT3 gene encoding kindlin-3 underlie the human immune deficiency known as leukocyte adhesion deficiency-III. Both kindlin-3 and talin-1, another FERM domain-containing cytoskeletal protein, are required for integrin activation, but their individual roles in the induction of specific integrin conformers are unclear. Here, we induce differential LFA-1 activation in neutrophils through engagement of the selectin ligand P-selectin glycoprotein ligand-1 or the chemokine receptor CXCR2. We find that talin-1 is required for inducing LFA-1 extension, which corresponds to intermediate affinity and induces neutrophil slow rolling, whereas both talin-1 and kindlin-3 are required for induction of the high-affinity conformation of LFA-1 with an open headpiece, which results in neutrophil arrest. In vivo, both slow rolling and arrest are defective in talin-1-deficient neutrophils, whereas only arrest is defective in kindlin-3-deficient neutrophils. We conclude that talin-1 and kindlin-3 serve distinct functions in LFA-1 activation.


Subject(s)
Chemotaxis, Leukocyte/physiology , Cytoskeletal Proteins/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Neutrophils/metabolism , Talin/physiology , Animals , Bone Marrow Transplantation , Cell Adhesion , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Green Fluorescent Proteins/analysis , HL-60 Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , K562 Cells , Lymphocyte Function-Associated Antigen-1/chemistry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/metabolism , Protein Binding , Protein Conformation , RNA Interference , RNA, Small Interfering/pharmacology , Radiation Chimera , Specific Pathogen-Free Organisms , Talin/antagonists & inhibitors , Talin/genetics
18.
Eur J Cell Biol ; 91(3): 180-91, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22306379

ABSTRACT

Talins are adaptor proteins that connect the integrin family of cell adhesion receptors to cytoskeletal actin. Vertebrates express two closely related talins encoded by separate genes, and while it is well established that talin1 plays a key role in cell adhesion and spreading, little is known about the role of talin2. To facilitate such studies, we report the characterisation of 4 new isoform-specific talin mouse monoclonal antibodies that work in Western blotting, immuno-precipitation, immuno-fluorescence and immuno-histochemistry. Using these antibodies, we show that talin1 and talin2 do not form heterodimers, and that they are differentially localised within the cell. Talin1 was concentrated in peripheral focal adhesions while talin2 was observed in both focal and fibrillar adhesions, and knock-down of talin2 compromised fibronectin fibrillogenesis. Although differentiated human macrophages express both isoforms, only talin1 showed discrete staining and was localised to the ring structure of podosomes. However, siRNA-mediated knock-down of macrophage talin2 led to a significant reduction in podosomal matrix degradation. We have also used the antibodies to localise each isoform in tissue sections using both cryostat and paraffin-embedded material. In skeletal muscle talin2 was localised to both myotendinous junctions and costameres while talin1 was restricted to the former structure. In contrast, both isoforms co-localised in kidney with staining of the glomerulus, and the tubular epithelial and interstitial cells of the cortex and medulla. We anticipate that these antibodies will form a valuable resource for future studies on the function of the two major talin isoforms.


Subject(s)
Antibodies, Monoclonal , Fibronectins/metabolism , Macrophages/ultrastructure , Protein Isoforms/analysis , Talin/metabolism , Animals , Antibody Specificity , Focal Adhesions/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Protein Isoforms/metabolism , RNA, Small Interfering , Rats
19.
J Immunol ; 187(12): 6256-67, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22075696

ABSTRACT

T cell-APC contact initiates T cell activation and is maintained by the integrin LFA-1. Talin1, an LFA-1 regulator, localizes to the immune synapse (IS) with unknown roles in T cell activation. In this study, we show that talin1-deficient T cells have defects in contact-dependent T cell stopping and proliferation. Although talin1-deficient T cells did not form stable interactions with APCs, transient contacts were sufficient to induce signaling. In contrast to prior models, LFA-1 polarized to T cell-APC contacts in talin1-deficient T cells, but vinculin and F-actin polarization at the IS was impaired. These results indicate that T cell proliferation requires sustained, talin1-mediated T cell-APC interactions and that talin1 is necessary for F-actin polarization and the stability of the IS.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Immunological Synapses/immunology , Lymphocyte Activation/immunology , Talin/physiology , Actins/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Communication/genetics , Cell Polarity/genetics , Cell Polarity/immunology , Cell Proliferation , Cells, Cultured , Immunological Synapses/genetics , Lymphocyte Activation/genetics , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , Talin/deficiency , Talin/genetics , Vinculin/metabolism
20.
Mol Cell Biol ; 31(16): 3366-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21670148

ABSTRACT

Talin is a cytoskeletal protein that binds to integrin ß cytoplasmic tails and regulates integrin activation. Talin1 ablation in mice disrupts gastrulation and causes embryonic lethality. However, the role of talin in mammalian epithelial morphogenesis is poorly understood. Here we demonstrate that embryoid bodies (EBs) differentiated from talin1-null embryonic stem cells are defective in integrin adhesion complex assembly, epiblast elongation, and lineage differentiation. These defects are accompanied by a significant reduction in integrin ß1 protein levels due to accelerated degradation through an MG-132-sensitive proteasomal pathway. Overexpression of integrin ß1 or MG-132 treatment in mutant EBs largely rescues the phenotype. In addition, epiblast cells isolated from talin1-null EBs exhibit impaired cell spreading and focal adhesion formation. Transfection of the mutant cells with green fluorescent protein (GFP)-tagged wild-type but not mutant talin1 that is defective in integrin binding normalizes integrin ß1 protein levels and restores focal adhesion formation. Significantly, cell adhesion and spreading are also improved by overexpression of integrin ß1. All together, these results suggest that talin1 binding to integrin promotes epiblast adhesion and morphogenesis in part by preventing integrin ß1 degradation.


Subject(s)
Integrins/metabolism , Morphogenesis , Talin/physiology , Animals , Cell Adhesion , Cells, Cultured , Embryoid Bodies , Embryonic Stem Cells , Epithelium/embryology , Focal Adhesions , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Protein Stability
SELECTION OF CITATIONS
SEARCH DETAIL
...