Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 25
Filtrer
3.
Am J Pathol ; 194(2): 195-208, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37981221

RÉSUMÉ

miRNAs are small noncoding RNAs that regulate mRNA targets in a cell-specific manner. miR-29 is expressed in murine and human skin, where it may regulate functions in skin repair. Cutaneous wound healing model in miR-29a/b1 gene knockout mice was used to identify miR-29 targets in the wound matrix, where angiogenesis and maturation of provisional granulation tissue was enhanced in response to genetic deletion of miR-29. Consistently, antisense-mediated inhibition of miR-29 promoted angiogenesis in vitro by autocrine and paracrine mechanisms. These processes are likely mediated by miR-29 target mRNAs released upon removal of miR-29 to improve cell-matrix adhesion. One of these, laminin (Lam)-c2 (also known as laminin γ2), was strongly up-regulated during skin repair in the wound matrix of knockout mice. Unexpectedly, Lamc2 was deposited in the basal membrane of endothelial cells in blood vessels forming in the granulation tissue of knockout mice. New blood vessels showed punctate interactions between Lamc2 and integrin α6 (Itga6) along the length of the proto-vessels, suggesting that greater levels of Lamc2 may contribute to the adhesion of endothelial cells, thus assisting angiogenesis within the wound. These findings may be of translational relevance, as LAMC2 was deposited at the leading edge in human wounds, where it formed a basal membrane for endothelial cells and assisted neovascularization. These results suggest a link between LAMC2, improved angiogenesis, and re-epithelialization.


Sujet(s)
Laminine , microARN , Humains , Animaux , Souris , Laminine/génétique , Cellules endothéliales , Transduction du signal/physiologie , microARN/génétique , Peau , Souris knockout
4.
Biophys J ; 122(16): 3219-3237, 2023 08 22.
Article de Anglais | MEDLINE | ID: mdl-37415335

RÉSUMÉ

Collagen is a key structural component of multicellular organisms and is arranged in a highly organized manner. In structural tissues such as tendons, collagen forms bundles of parallel fibers between cells, which appear within a 24-h window between embryonic day 13.5 (E13.5) and E14.5 during mouse embryonic development. Current models assume that the organized structure of collagen requires direct cellular control, whereby cells actively lay down collagen fibrils from cell surfaces. However, such models appear incompatible with the time and length scales of fibril formation. We propose a phase-transition model to account for the rapid development of ordered fibrils in embryonic tendon, reducing reliance on active cellular processes. We develop phase-field crystal simulations of collagen fibrillogenesis in domains derived from electron micrographs of inter-cellular spaces in embryonic tendon and compare results qualitatively and quantitatively to observed patterns of fibril formation. To test the prediction of this phase-transition model that free protomeric collagen should exist in the inter-cellular spaces before the formation of observable fibrils, we use laser-capture microdissection, coupled with mass spectrometry, which demonstrates steadily increasing free collagen in inter-cellular spaces up to E13.5, followed by a rapid reduction of free collagen that coincides with the appearance of less-soluble collagen fibrils. The model and measurements together provide evidence for extracellular self-assembly of collagen fibrils in embryonic mouse tendon, supporting an additional mechanism for rapid collagen fibril formation during embryonic development.


Sujet(s)
Développement embryonnaire , Matrice extracellulaire , Animaux , Souris , Matrice extracellulaire/métabolisme , Collagène/métabolisme , Membrane cellulaire , Tendons/composition chimique , Tendons/métabolisme
5.
Respir Res ; 24(1): 99, 2023 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-37005656

RÉSUMÉ

Honeycombing is a histological pattern consistent with Usual Interstitial Pneumonia (UIP). Honeycombing refers to cystic airways located at sites of dense fibrosis with marked mucus accumulation. Utilizing laser capture microdissection coupled mass spectrometry (LCM-MS), we interrogated the fibrotic honeycomb airway cells and fibrotic uninvolved airway cells (distant from honeycomb airways and morphologically intact) in specimens from 10 patients with UIP. Non-fibrotic airway cell specimens from 6 patients served as controls. Furthermore, we performed LCM-MS on the mucus plugs found in 6 patients with UIP and 6 patients with mucinous adenocarcinoma. The mass spectrometry data were subject to both qualitative and quantitative analysis and validated by immunohistochemistry. Surprisingly, fibrotic uninvolved airway cells share a similar protein profile to honeycomb airway cells, showing deregulation of the slit and roundabout receptor (Slit and Robo) pathway as the strongest category. We find that (BPI) fold-containing family B member 1 (BPIFB1) is the most significantly increased secretome-associated protein in UIP, whereas Mucin-5AC (MUC5AC) is the most significantly increased in mucinous adenocarcinoma. We conclude that fibrotic uninvolved airway cells share pathological features with fibrotic honeycomb airway cells. In addition, fibrotic honeycomb airway cells are enriched in mucin biogenesis proteins with a marked derangement in proteins essential for ciliogenesis. This unbiased spatial proteomic approach generates novel and testable hypotheses to decipher fibrosis progression.


Sujet(s)
Fibrose pulmonaire idiopathique , Protéome , Humains , Protéomique , Fibrose pulmonaire idiopathique/anatomopathologie , Poumon/anatomopathologie
6.
JCI Insight ; 8(4)2023 02 22.
Article de Anglais | MEDLINE | ID: mdl-36656644

RÉSUMÉ

Hypoxia is a sentinel feature of idiopathic pulmonary fibrosis (IPF). The IPF microenvironment contains high lactate levels, and hypoxia enhances cellular lactate production. Lactate, acting through the GPR81 lactate receptor, serves as a signal molecule regulating cellular processes. We previously identified intrinsically fibrogenic mesenchymal progenitor cells (MPCs) that drive fibrosis in the lungs of patients with IPF. However, whether hypoxia enhances IPF MPC fibrogenicity is unclear. We hypothesized that hypoxia increases IPF MPC fibrogenicity via lactate and its cognate receptor GPR81. Here we show that hypoxia promotes IPF MPC self-renewal. The mechanism involves hypoxia-mediated enhancement of LDHA function and lactate production and release. Hypoxia also increases HIF1α levels, and this increase in turn augments the expression of GPR81. Exogenous lactate operating through GPR81 promotes IPF MPC self-renewal. IHC analysis of IPF lung tissue demonstrates IPF MPCs expressing GPR81 and hypoxic markers on the periphery of the fibroblastic focus. We show that hypoxia enhances IPF MPC fibrogenicity in vivo. We demonstrate that knockdown of GPR81 inhibits hypoxia-induced IPF MPC self-renewal in vitro and attenuates hypoxia-induced IPF MPC fibrogenicity in vivo. Our data demonstrate that hypoxia creates a feed-forward loop that augments IPF MPC fibrogenicity via the lactate/GPR81/HIF1α pathway.


Sujet(s)
Fibrose pulmonaire idiopathique , Cellules souches mésenchymateuses , Humains , Acide lactique/métabolisme , Fibrose pulmonaire idiopathique/métabolisme , Transduction du signal , Cellules souches mésenchymateuses/métabolisme , Hypoxie/métabolisme
7.
JCI Insight ; 7(16)2022 08 22.
Article de Anglais | MEDLINE | ID: mdl-35852874

RÉSUMÉ

Usual interstitial pneumonia (UIP) is a histological pattern characteristic of idiopathic pulmonary fibrosis (IPF). The UIP pattern is patchy with histologically normal lung adjacent to dense fibrotic tissue. At this interface, fibroblastic foci (FF) are present and are sites where myofibroblasts and extracellular matrix (ECM) accumulate. Utilizing laser capture microdissection-coupled mass spectrometry, we interrogated the FF, adjacent mature scar, and adjacent alveoli in 6 fibrotic (UIP/IPF) specimens plus 6 nonfibrotic alveolar specimens as controls. The data were subjected to qualitative and quantitative analysis and histologically validated. We found that the fibrotic alveoli protein signature is defined by immune deregulation as the strongest category. The fibrotic mature scar classified as end-stage fibrosis whereas the FF contained an overabundance of a distinctive ECM compared with the nonfibrotic control. Furthermore, FF were positive for both TGFB1 and TGFB3, whereas the aberrant basaloid cell lining of FF was predominantly positive for TGFB2. In conclusion, spatial proteomics demonstrated distinct protein compositions in the histologically defined regions of UIP/IPF tissue. These data revealed that FF are the main site of collagen biosynthesis and that the adjacent alveoli are abnormal. This essential information will inform future mechanistic studies on fibrosis progression.


Sujet(s)
Fibrose pulmonaire idiopathique , Cicatrice/anatomopathologie , Collagène , Matrice extracellulaire/anatomopathologie , Fibrose , Humains , Fibrose pulmonaire idiopathique/anatomopathologie
8.
Article de Anglais | MEDLINE | ID: mdl-35379658

RÉSUMÉ

Mechanical variables such as stiffness, stress, strain, and fluid shear stress are central to tissue functions, thus, must be maintained within the proper range. Mechanics are especially important in the cardiovascular system and lung, the functions of which are essentially mechanical. Mechanical homeostasis is characterized by negative feedback in which deviations from the optimal value or set point activates mechanisms to return the system to the correct range. In chronic diseases, homeostatic mechanisms are generally overcome or replaced with positive feedback loops that promote disease progression. Recent work has shown that microRNAs (miRNAs) are essential to mechanical homeostasis in a number of biological systems and that perturbations to miRNA biogenesis play key roles in cardiovascular and pulmonary diseases. In this review, we integrate current knowledge of miRNAs in mechanical homeostasis and how these mechanisms are altered in disease.


Sujet(s)
microARN , Évolution de la maladie , Homéostasie , Humains , microARN/génétique , Contrainte mécanique
9.
Life Sci Alliance ; 4(8)2021 08.
Article de Anglais | MEDLINE | ID: mdl-34127548

RÉSUMÉ

IL-13 is implicated in effective repair after acute lung injury and the pathogenesis of chronic diseases such as allergic asthma. Both these processes involve matrix remodelling, but understanding the specific contribution of IL-13 has been challenging because IL-13 shares receptors and signalling pathways with IL-4. Here, we used Nippostrongylus brasiliensis infection as a model of acute lung damage comparing responses between WT and IL-13-deficient mice, in which IL-4 signalling is intact. We found that IL-13 played a critical role in limiting tissue injury and haemorrhaging in the lung, and through proteomic and transcriptomic profiling, identified IL-13-dependent changes in matrix and associated regulators. We further showed a requirement for IL-13 in the induction of epithelial-derived type 2 effector molecules such as RELM-α and surfactant protein D. Pathway analyses predicted that IL-13 induced cellular stress responses and regulated lung epithelial cell differentiation by suppression of Foxa2 pathways. Thus, in the context of acute lung damage, IL-13 has tissue-protective functions and regulates epithelial cell responses during type 2 immunity.


Sujet(s)
Lésion pulmonaire aigüe/parasitologie , Interleukine-13/déficit , Nippostrongylus/pathogénicité , Infections à Strongylida/génétique , Lésion pulmonaire aigüe/génétique , Lésion pulmonaire aigüe/métabolisme , Animaux , Modèles animaux de maladie humaine , Femelle , Analyse de profil d'expression de gènes , Protéines et peptides de signalisation intercellulaire/métabolisme , Mâle , Souris , Protéomique , Infections à Strongylida/métabolisme , Régulation positive
10.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L926-L941, 2021 05 01.
Article de Anglais | MEDLINE | ID: mdl-33719561

RÉSUMÉ

Despite modest improvement in patient outcomes from recent advances in pharmacotherapy targeting fibrogenic signaling pathways, idiopathic pulmonary fibrosis (IPF) remains a major unsolved clinical problem. One reason for this is that available antifibrotic agents slow down but do not arrest fibrotic progression. To arrest fibrotic progression, its obligatory drivers need to be identified. We previously discovered that fibrogenic mesenchymal progenitor cells (MPCs) are key drivers of fibrotic progression in IPF, serving as cells of origin for disease-mediating myofibroblasts. IPF MPCs have high levels of nuclear S100A4, which interacts with the proteasome to promote p53 degradation and self-renewal. However, the mechanism underlying S100A4 accumulation in the nucleus of IPF MPCs remains unknown. Here we show that hyaluronan (HA) is present in the fibroblastic focus together with CD44-expressing MPCs and that ligation of CD44 by HA triggers S100A4 nuclear translocation to support IPF MPC self-renewal. The mechanism involves HA-mediated formation of a CD44/S100A4/transportin 1 complex, which promotes S100A4 nuclear import. In a humanized mouse model of pulmonary fibrosis, IPF MPC fibrogenicity was significantly attenuated by 1) knockdown of CD44 or 2) introduction of an S100A4 mutant construct that prevents S100A4 nuclear import. These data indicate that signaling through the HA/CD44/S100A4 axis is an integral component of IPF MPC fibrogenicity.


Sujet(s)
Noyau de la cellule/métabolisme , Antigènes CD44/métabolisme , Acide hyaluronique/métabolisme , Fibrose pulmonaire idiopathique/métabolisme , Cellules souches mésenchymateuses/métabolisme , Protéine S100A4 liant le calcium/métabolisme , Transduction du signal , Animaux , Noyau de la cellule/génétique , Noyau de la cellule/anatomopathologie , Modèles animaux de maladie humaine , Techniques de knock-down de gènes , Humains , Antigènes CD44/génétique , Acide hyaluronique/génétique , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Cellules souches mésenchymateuses/anatomopathologie , Souris , Complexes multiprotéiques/génétique , Complexes multiprotéiques/métabolisme , Protéine S100A4 liant le calcium/génétique , Caryophérines bêta/génétique , Caryophérines bêta/métabolisme
11.
Sci Rep ; 10(1): 11162, 2020 07 07.
Article de Anglais | MEDLINE | ID: mdl-32636398

RÉSUMÉ

In Idiopathic Pulmonary Fibrosis (IPF), there is unrelenting scarring of the lung mediated by pathological mesenchymal progenitor cells (MPCs) that manifest autonomous fibrogenicity in xenograft models. To determine where along their differentiation trajectory IPF MPCs acquire fibrogenic properties, we analyzed the transcriptome of 335 MPCs isolated from the lungs of 3 control and 3 IPF patients at the single-cell level. Using transcriptional entropy as a metric for differentiated state, we found that the least differentiated IPF MPCs displayed the largest differences in their transcriptional profile compared to control MPCs. To validate entropy as a surrogate for differentiated state functionally, we identified increased CD44 as a characteristic of the most entropic IPF MPCs. Using FACS to stratify IPF MPCs based on CD44 expression, we determined that CD44hi IPF MPCs manifested an increased capacity for anchorage-independent colony formation compared to CD44lo IPF MPCs. To validate our analysis morphologically, we used two differentially expressed genes distinguishing IPF MPCs from control (CD44, cell surface; and MARCKS, intracellular). In IPF lung tissue, pathological MPCs resided in the highly cellular perimeter region of the fibroblastic focus. Our data support the concept that IPF fibroblasts acquire a cell-autonomous pathological phenotype early in their differentiation trajectory.


Sujet(s)
Différenciation cellulaire , Fibrose pulmonaire idiopathique/métabolisme , Poumon/métabolisme , Cellules souches mésenchymateuses/métabolisme , Analyse de séquence d'ARN , Études cas-témoins , Différenciation cellulaire/génétique , Technique d'immunofluorescence , Analyse de profil d'expression de gènes , Humains , Antigènes CD44/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Poumon/anatomopathologie , Cellules souches mésenchymateuses/anatomopathologie
12.
Clin Proteomics ; 17: 24, 2020.
Article de Anglais | MEDLINE | ID: mdl-32565759

RÉSUMÉ

BACKGROUND: Haematoxylin and eosin (H&E)-which respectively stain nuclei blue and other cellular and stromal material pink-are routinely used for clinical diagnosis based on the identification of morphological features. A richer characterization can be achieved by laser capture microdissection coupled to mass spectrometry (LCM-MS), giving an unbiased assay of the proteins that make up the tissue. However, the process of fixing and H&E staining of tissues provides challenges with standard sample preparation methods for mass spectrometry, resulting in low protein yield. Here we describe a microproteomics technique to analyse H&E-stained, formalin-fixed paraffin-embedded (FFPE) tissues. METHODS: Herein, we utilize heat extraction, physical disruption, and in column digestion for the analysis of H&E stained FFPE tissues. Micro-dissected morphologically normal human lung alveoli (0.082 mm3) and human lung blood vessels (0.094 mm3) from FFPE-fixed H&E-stained sections from Idiopathic Pulmonary Fibrosis (IPF) specimens (n = 3 IPF specimens) were then subject to a qualitative and then quantitative proteomics approach using BayesENproteomics. In addition, we tested the sensitivity of this method by processing and analysing a range of micro-dissected human lung blood vessel tissue volumes. RESULTS: This approach yields 1252 uniquely expressed proteins (at a protein identification threshold of 3 unique peptides) with 892 differentially expressed proteins between these regions. In accord with prior knowledge, our methodology approach confirms that human lung blood vessels are enriched with smoothelin, CNN1, ITGA7, MYH11, TAGLN, and PTGIS; whereas morphologically normal human lung alveoli are enriched with cytokeratin-7, -8, -18, -19, 14, and -17. In addition, we identify a total of 137 extracellular matrix (ECM) proteins and immunohistologically validate that laminin subunit beta-1 localizes to morphologically normal human lung alveoli and tenascin localizes to human lung blood vessels. Lastly, we show that this micro-proteomics technique can be applied to tissue volumes as low as 0.0125 mm3. CONCLUSION: Herein we show that our multistep sample preparation methodology of LCM-MS can identify distinct, characteristic proteomic compositions of anatomical features within complex fixed and stained tissues.

13.
Respir Res ; 21(1): 132, 2020 May 29.
Article de Anglais | MEDLINE | ID: mdl-32471489

RÉSUMÉ

BACKGROUND: Chronic tissue injury was shown to induce progressive scarring in fibrotic diseases such as idiopathic pulmonary fibrosis (IPF), while an array of repair/regeneration and stress responses come to equilibrium to determine the outcome of injury at the organ level. In the lung, type I alveolar epithelial (ATI) cells constitute the epithelial barrier, while type II alveolar epithelial (ATII) cells play a pivotal role in regenerating the injured distal lungs. It had been demonstrated that eukaryotic cells possess repair machinery that can quickly patch the damaged plasma membrane after injury, and our previous studies discovered the membrane-mending role of Tripartite motif containing 72 (TRIM72) that expresses in a limited number of tissues including the lung. Nevertheless, the role of alveolar epithelial cell (AEC) repair in the pathogenesis of IPF has not been examined yet. METHOD: In this study, we tested the specific roles of TRIM72 in the repair of ATII cells and the development of lung fibrosis. The role of membrane repair was accessed by saponin assay on isolated primary ATII cells and rat ATII cell line. The anti-fibrotic potential of TRIM72 was tested with bleomycin-treated transgenic mice. RESULTS: We showed that TRIM72 was upregulated following various injuries and in human IPF lungs. However, TRIM72 expression in ATII cells of the IPF lungs had aberrant subcellular localization. In vitro studies showed that TRIM72 repairs membrane injury of immortalized and primary ATIIs, leading to inhibition of stress-induced p53 activation and reduction in cell apoptosis. In vivo studies demonstrated that TRIM72 protects the integrity of the alveolar epithelial layer and reduces lung fibrosis. CONCLUSION: Our results suggest that TRIM72 protects injured lungs and ameliorates fibrosis through promoting post-injury repair of AECs.


Sujet(s)
Pneumocytes/métabolisme , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/prévention et contrôle , Poumon/métabolisme , Protéines à motif tripartite/biosynthèse , Pneumocytes/effets des médicaments et des substances chimiques , Animaux , Bléomycine/toxicité , Femelle , Cellules HEK293 , Humains , Fibrose pulmonaire idiopathique/induit chimiquement , Poumon/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de souche-129 , Souris knockout , Protéines recombinantes/biosynthèse
14.
JCI Insight ; 4(1)2019 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-30626754

RÉSUMÉ

The extracellular matrix (ECM) in idiopathic pulmonary fibrosis (IPF) drives fibrosis progression; however, the ECM composition of the fibroblastic focus (the hallmark lesion in IPF) and adjacent regions remains incompletely defined. Herein, we serially sectioned IPF lung specimens constructed into tissue microarrays and immunostained for ECM components reported to be deregulated in IPF. Immunostained sections were imaged, anatomically aligned, and 3D reconstructed. The myofibroblast core of the fibroblastic focus (defined by collagen I, α-smooth muscle actin, and procollagen I immunoreactivity) was associated with collagens III, IV, V, and VI; fibronectin; hyaluronan; and versican immunoreactivity. Hyaluronan immunoreactivity was also present at the fibroblastic focus perimeter and at sites where early lesions appear to be forming. Fibrinogen immunoreactivity was often observed at regions of damaged epithelium lining the airspace and the perimeter of the myofibroblast core but was absent from the myofibroblast core itself. The ECM components of the fibroblastic focus were distributed in a characteristic and reproducible manner in multiple patients. This information can inform the development of high-fidelity model systems to dissect mechanisms by which the IPF ECM drives fibrosis progression.

15.
J Am Soc Nephrol ; 29(10): 2493-2509, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30097458

RÉSUMÉ

BACKGROUND: Mutation of HNF1B, the gene encoding transcription factor HNF-1ß, is one cause of autosomal dominant tubulointerstitial kidney disease, a syndrome characterized by tubular cysts, renal fibrosis, and progressive decline in renal function. HNF-1ß has also been implicated in epithelial-mesenchymal transition (EMT) pathways, and sustained EMT is associated with tissue fibrosis. The mechanism whereby mutated HNF1B leads to tubulointerstitial fibrosis is not known. METHODS: To explore the mechanism of fibrosis, we created HNF-1ß-deficient mIMCD3 renal epithelial cells, used RNA-sequencing analysis to reveal differentially expressed genes in wild-type and HNF-1ß-deficient mIMCD3 cells, and performed cell lineage analysis in HNF-1ß mutant mice. RESULTS: The HNF-1ß-deficient cells exhibited properties characteristic of mesenchymal cells such as fibroblasts, including spindle-shaped morphology, loss of contact inhibition, and increased cell migration. These cells also showed upregulation of fibrosis and EMT pathways, including upregulation of Twist2, Snail1, Snail2, and Zeb2, which are key EMT transcription factors. Mechanistically, HNF-1ß directly represses Twist2, and ablation of Twist2 partially rescued the fibroblastic phenotype of HNF-1ß mutant cells. Kidneys from HNF-1ß mutant mice showed increased expression of Twist2 and its downstream target Snai2. Cell lineage analysis indicated that HNF-1ß mutant epithelial cells do not transdifferentiate into kidney myofibroblasts. Rather, HNF-1ß mutant epithelial cells secrete high levels of TGF-ß ligands that activate downstream Smad transcription factors in renal interstitial cells. CONCLUSIONS: Ablation of HNF-1ß in renal epithelial cells leads to the activation of a Twist2-dependent transcriptional network that induces EMT and aberrant TGF-ß signaling, resulting in renal fibrosis through a cell-nonautonomous mechanism.


Sujet(s)
Goutte/génétique , Goutte/anatomopathologie , Facteur nucléaire hépatocytaire HNF-1 bêta/génétique , Hyperuricémie/génétique , Hyperuricémie/anatomopathologie , Maladies du rein/génétique , Maladies du rein/anatomopathologie , Animaux , Lignée cellulaire , Lignage cellulaire/génétique , Modèles animaux de maladie humaine , Transition épithélio-mésenchymateuse/génétique , Femelle , Fibrose , Gènes dominants , Goutte/métabolisme , Facteur nucléaire hépatocytaire HNF-1 bêta/déficit , Facteur nucléaire hépatocytaire HNF-1 bêta/métabolisme , Humains , Hyperuricémie/métabolisme , Rein/métabolisme , Rein/anatomopathologie , Maladies du rein/métabolisme , Mâle , Souris , Souris transgéniques , Mutation , Protéines de répression/déficit , Protéines de répression/génétique , Protéines de répression/métabolisme , Transduction du signal , Facteur de croissance transformant bêta/métabolisme , Protéine-1 apparentée à Twist/déficit , Protéine-1 apparentée à Twist/génétique , Protéine-1 apparentée à Twist/métabolisme
16.
Am J Respir Crit Care Med ; 198(4): 486-496, 2018 08 15.
Article de Anglais | MEDLINE | ID: mdl-29579397

RÉSUMÉ

RATIONALE: The lung extracellular matrix (ECM) in idiopathic pulmonary fibrosis (IPF) mediates progression of fibrosis by decreasing fibroblast expression of miR-29 (microRNA-29), a master negative regulator of ECM production. The molecular mechanism is undefined. IPF-ECM is stiffer than normal. Stiffness drives fibroblast ECM production in a YAP (yes-associated protein)-dependent manner, and YAP is a known regulator of miR-29. Therefore, we tested the hypothesis that negative regulation of miR-29 by IPF-ECM was mediated by mechanotransduction of stiffness. OBJECTIVES: To determine how IPF-ECM negatively regulates miR-29. METHODS: We decellularized lung ECM using detergents and prepared polyacrylamide hydrogels of defined stiffness by varying acrylamide concentrations. Mechanistic studies were guided by immunohistochemistry of IPF lung and used cell culture, RNA-binding protein assays, and xenograft models. MEASUREMENTS AND MAIN RESULTS: Contrary to our hypothesis, we excluded fibroblast mechanotransduction of ECM stiffness as the primary mechanism deregulating miR-29. Instead, systematic examination of miR-29 biogenesis revealed a microRNA processing defect that impeded processing of miR-29 into its mature bioactive forms. Immunohistochemical analysis of the microRNA processing machinery in IPF lung specimens revealed decreased Dicer1 expression in the procollagen-rich myofibroblastic core of fibroblastic foci compared with the focus perimeter and adjacent alveolar walls. Mechanistically, IPF-ECM increased association of the Dicer1 transcript with RNA binding protein AUF1 (AU-binding factor 1), and Dicer1 knockdown conferred primary human lung fibroblasts with cell-autonomous fibrogenicity in zebrafish and mouse lung xenograft models. CONCLUSIONS: Our data identify suppression of fibroblast Dicer1 expression in the myofibroblast-rich IPF fibroblastic focus core as a central step in the mechanism by which the ECM sustains fibrosis progression in IPF.


Sujet(s)
DEAD-box RNA helicases/génétique , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , microARN/métabolisme , Ribonuclease III/génétique , Animaux , Prolifération cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Matrice extracellulaire/métabolisme , Fibroblastes/métabolisme , Fibrose/génétique , Fibrose/anatomopathologie , Humains , Fibrose pulmonaire idiopathique/métabolisme , Poumon/métabolisme , Poumon/anatomopathologie , Souris , Danio zébré
17.
J Clin Invest ; 128(1): 45-53, 2018 01 02.
Article de Anglais | MEDLINE | ID: mdl-29293088

RÉSUMÉ

The extracellular matrix (ECM) is dynamically tuned to optimize physiological function. Its major properties, including composition and mechanics, profoundly influence cell biology. Cell-ECM interactions operate through an integrated set of sensor and effector circuits that use several classes of receptors and signal transduction pathways. At the single-cell level, the ECM governs differentiation, metabolism, motility, orientation, proliferation, and survival. At the cell population level, the ECM provides higher-order guidance that is essential for physiological function. When pathological changes in the ECM lead to impairment of organ function, we use the term "fibrosis." In this Review, we differentiate fibrosis initiation from progression and focus primarily on progressive lung fibrosis impairing organ function. We present a working model to explain how the altered ECM is not only a consequence but also a driver of fibrosis. Additionally, we advance the concept that fibrosis progression occurs in a fibrogenic niche that is composed of a fibrogenic ECM that nurtures fibrogenic mesenchymal progenitor cells and their fibrogenic progeny.


Sujet(s)
Mouvement cellulaire , Polarité de la cellule , Matrice extracellulaire/métabolisme , Maladies pulmonaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , Transduction du signal , Animaux , Différenciation cellulaire , Matrice extracellulaire/génétique , Matrice extracellulaire/anatomopathologie , Fibrose , Humains , Maladies pulmonaires/génétique , Maladies pulmonaires/anatomopathologie , Cellules souches mésenchymateuses/anatomopathologie
18.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L127-L136, 2018 01 01.
Article de Anglais | MEDLINE | ID: mdl-28860143

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease, but the mechanisms driving progression remain incompletely defined. We previously reported that the IPF lung harbors fibrogenic mesenchymal progenitor cells (MPCs), which serve as a cell of origin for IPF fibroblasts. Proliferating IPF MPCs are located at the periphery of fibroblastic foci in an active cellular front at the interface between the myofibroblast-rich focus core and adjacent normal alveolar structures. Among a large set of genes that distinguish IPF MPCs from their control counterparts, we identified IL-8 as a candidate mediator of IPF MPC fibrogenicity and driver of fibrotic progression. IPF MPCs and their progeny displayed increased steady-state levels of IL-8 and its cognate receptor CXCR1 and secreted more IL-8 than did controls. IL-8 functioned in an autocrine manner promoting IPF MPC self-renewal and the proliferation and motility of IPF MPC progeny. Secreted IL-8 also functioned in a paracrine manner stimulating macrophage migration. Analysis of IPF lung tissue demonstrated codistribution of IPF MPCs with activated macrophages in the active cellular front of the fibroblastic focus. These findings indicate that IPF MPC-derived IL-8 is capable of expanding the mesenchymal cell population and recruiting activated macrophages cells to actively evolving fibrotic lesions.


Sujet(s)
Mouvement cellulaire , Fibrose pulmonaire idiopathique/anatomopathologie , Interleukine-8/métabolisme , Cellules souches mésenchymateuses/anatomopathologie , Prolifération cellulaire , Cellules cultivées , Humains , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/métabolisme , Interleukine-8/génétique , Cellules souches mésenchymateuses/métabolisme , Transduction du signal
19.
J Clin Invest ; 127(7): 2586-2597, 2017 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-28530639

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a progressive disease with a prevalence of 1 million persons worldwide. The fibrosis spreads from affected alveoli into contiguous alveoli and leads to death by asphyxiation. We previously discovered that the IPF lung harbors fibrogenic mesenchymal progenitor cells (MPCs) that serve as a cell of origin for disease-mediating myofibroblasts. In a prior genomewide transcriptional analysis, we found that IPF MPCs displayed increased expression of S100 calcium-binding A4 (S100A4), a protein linked to cancer cell proliferation and invasiveness. Here, we have examined whether S100A4 mediates MPC fibrogenicity. Ex vivo analysis revealed that IPF MPCs had increased levels of nuclear S100A4, which interacts with L-isoaspartyl methyltransferase to promote p53 degradation and MPC self-renewal. In vivo, injection of human IPF MPCs converted a self-limited bleomycin-induced mouse model of lung fibrosis to a model of persistent fibrosis in an S100A4-dependent manner. S100A4 gain of function was sufficient to confer fibrotic properties to non-IPF MPCs. In IPF tissue, fibroblastic foci contained cells expressing Ki67 and the MPC markers SSEA4 and S100A4. The expression colocalized in an interface region between myofibroblasts in the focus core and normal alveolar structures, defining this region as an active fibrotic front. Our findings indicate that IPF MPCs are intrinsically fibrogenic and that S100A4 confers MPCs with fibrogenicity.


Sujet(s)
Fibrose pulmonaire idiopathique/métabolisme , Cellules souches mésenchymateuses/métabolisme , Protéine S100A4 liant le calcium/métabolisme , Animaux , Modèles animaux de maladie humaine , Femelle , Humains , Fibrose pulmonaire idiopathique/induit chimiquement , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Mâle , Cellules souches mésenchymateuses/anatomopathologie , Souris , Souris transgéniques , Protéine S100A4 liant le calcium/génétique
20.
Am J Respir Cell Mol Biol ; 53(3): 391-9, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25612003

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is characterized by the relentless expansion of fibroblasts depositing type I collagen within the alveolar wall and obliterating the alveolar airspace. MicroRNA (miR)-29 is a potent regulator of collagen expression. In IPF, miR-29 levels are low, whereas type I collagen expression is high. However, the mechanism for suppression of miR-29 and increased type I collagen expression in IPF remains unclear. Here we show that when IPF fibroblasts are seeded on polymerized type I collagen, miR-29c levels are suppressed and type I collagen expression is high. In contrast, miR-29c is high and type I collagen expression is low in control fibroblasts. We demonstrate that the mechanism for suppression of miR-29 during IPF fibroblast interaction with polymerized collagen involves inappropriately low protein phosphatase (PP) 2A function, leading to histone deacetylase (HDA) C4 phosphorylation and decreased nuclear translocation of HDAC4. We demonstrate that overexpression of HDAC4 in IPF fibroblasts restored miR-29c levels and decreased type I collagen expression, whereas knocking down HDAC4 in control fibroblasts suppressed miR-29c levels and increased type I collagen expression. Our data indicate that IPF fibroblast interaction with polymerized type I collagen results in an aberrant PP2A/HDAC4 axis, which suppresses miR-29, causing a pathologic increase in type I collagen expression.


Sujet(s)
Collagène de type I/métabolisme , Fibroblastes/enzymologie , Histone deacetylases/métabolisme , Fibrose pulmonaire idiopathique/métabolisme , microARN/métabolisme , Phosphoprotein Phosphatases/métabolisme , Protéines de répression/métabolisme , Noyau de la cellule/enzymologie , Cellules cultivées , Épigenèse génétique , Humains , Phosphorylation , Protein phosphatase 2C , Maturation post-traductionnelle des protéines , Transport des protéines , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE