Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33903236

ABSTRACT

Molecular events that drive the development of precancerous lesions in the bronchial epithelium, which are precursors of lung squamous cell carcinoma (LUSC), are poorly understood. We demonstrate that disruption of epithelial cellular polarity, via the conditional deletion of the apical determinant Crumbs3 (Crb3), initiates and sustains precancerous airway pathology. The loss of Crb3 in adult luminal airway epithelium promotes the uncontrolled activation of the transcriptional regulators YAP and TAZ, which stimulate intrinsic signals that promote epithelial cell plasticity and paracrine signals that induce basal-like cell growth. We show that aberrant polarity and YAP/TAZ-regulated gene expression associates with human bronchial precancer pathology and disease progression. Analyses of YAP/TAZ-regulated genes further identified the ERBB receptor ligand Neuregulin-1 (NRG1) as a key transcriptional target and therapeutic targeting of ERBB receptors as a means of preventing and treating precancerous cell growth. Our observations offer important molecular insight into the etiology of LUSC and provides directions for potential interception strategies of lung cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Membrane Glycoproteins/genetics , Neuregulin-1/genetics , Precancerous Conditions/genetics , YAP-Signaling Proteins/genetics , Carcinoma, Squamous Cell/pathology , Cell Polarity/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Epithelium/pathology , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Signal Transduction/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics
2.
J Invest Dermatol ; 138(1): 78-88, 2018 01.
Article in English | MEDLINE | ID: mdl-28870693

ABSTRACT

Systemic sclerosis (scleroderma, SSc) is a devastating fibrotic disease with few treatment options. Fumaric acid esters, including dimethyl fumarate (DMF, Tecfidera; Biogen, Cambridge, MA), have shown therapeutic effects in several disease models, prompting us to determine whether DMF is effective as a treatment for SSc dermal fibrosis. We found that DMF blocks the profibrotic effects of transforming growth factor-ß (TGFß) in SSc skin fibroblasts. Mechanistically, we found that DMF treatment reduced nuclear localization of transcriptional coactivator with PDZ binding motif (TAZ) and Yes-associated protein (YAP) proteins via inhibition of the phosphatidylinositol 3 kinase/protein kinase B (Akt) pathway. In addition, DMF abrogated TGFß/Akt1 mediated inhibitory phosphorylation of glycogen kinase 3ß (GSK3ß) and a subsequent ß-transducin repeat-containing proteins (ßTRCP) mediated proteasomal degradation of TAZ, as well as a corresponding decrease of TAZ/YAP transcriptional targets. Depletion of TAZ/YAP recapitulated the antifibrotic effects of DMF. We also confirmed the increase of TAZ/YAP in skin biopsies from patients with diffuse SSc. We further showed that DMF significantly diminished nuclear TAZ/YAP localization in fibroblasts cultured on a stiff surface. Importantly, DMF prevented bleomycin-induced skin fibrosis in mice. Together, our work demonstrates a mechanism of the antifibrotic effect of DMF via inhibition of Akt1/GSK3ß/TAZ/YAP signaling and confirms a critical role of TAZ/YAP in mediating the profibrotic responses in dermal fibroblasts. This study supports the use of DMF as a treatment for SSc dermal fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dimethyl Fumarate/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Scleroderma, Systemic/drug therapy , Signal Transduction/drug effects , Adult , Animals , Biopsy , Bleomycin/toxicity , Cell Cycle Proteins , Cell Nucleus/metabolism , Cells, Cultured , Dimethyl Fumarate/therapeutic use , Disease Models, Animal , Female , Fibroblasts , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phosphatidylinositol 3-Kinase/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Scleroderma, Systemic/pathology , Skin/drug effects , Skin/pathology , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transforming Growth Factor beta/metabolism , Treatment Outcome , YAP-Signaling Proteins
3.
J Clin Invest ; 127(10): 3866-3876, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28920925

ABSTRACT

P-element-induced wimpy testes (Piwi) proteins are known for suppressing retrotransposon activation in the mammalian germline. However, whether Piwi protein or Piwi-dependent functions occur in the mammalian soma is unclear. Contrary to germline-restricted expression, we observed that Piwi-like Miwi2 mRNA is indeed expressed in epithelial cells of the lung in adult mice and that it is induced during pneumonia. Further investigation revealed that MIWI2 protein localized to the cytoplasm of a discrete population of multiciliated airway epithelial cells. Isolation and next-generation sequencing of MIWI2-positive multiciliated cells revealed that they are phenotypically distinct from neighboring MIWI2-negative multiciliated cells. Mice lacking MIWI2 exhibited an altered balance of airway epithelial cells, demonstrating fewer multiciliated cells and an increase in club cells. During pneumococcal pneumonia, Miwi2-deficient mice exhibited increased expression of inflammatory mediators and increased immune cell recruitment, leading to enhanced bacterial clearance. Taken together, our data delineate MIWI2-dependent functions outside of the germline and demonstrate the presence of distinct subsets of airway multiciliated cells that can be discriminated by MIWI2 expression. By demonstrating roles for MIWI2 in airway cell identity and pulmonary innate immunity, these studies elucidate unanticipated physiological functions for Piwi proteins in somatic tissues.


Subject(s)
Argonaute Proteins/immunology , Epithelial Cells/immunology , Gene Expression Regulation , Immunity, Innate , Lung/immunology , Respiratory Mucosa/immunology , Animals , Argonaute Proteins/genetics , Female , Male , Mice , Mice, Knockout , RNA-Binding Proteins
4.
Elife ; 62017 05 11.
Article in English | MEDLINE | ID: mdl-28492365

ABSTRACT

Salivary glands, such as submandibular glands (SMGs), are composed of branched epithelial ductal networks that terminate in acini that together produce, transport and secrete saliva. Here, we show that the transcriptional regulator Yap, a key effector of the Hippo pathway, is required for the proper patterning and morphogenesis of SMG epithelium. Epithelial deletion of Yap in developing SMGs results in the loss of ductal structures, arising from reduced expression of the EGF family member Epiregulin, which we show is required for the expansion of Krt5/Krt14-positive ductal progenitors. We further show that epithelial deletion of the Lats1 and Lats2 genes, which encode kinases that restrict nuclear Yap localization, results in morphogenesis defects accompanied by an expansion of Krt5/Krt14-positive cells. Collectively, our data indicate that Yap-induced Epiregulin signaling promotes the identity of SMG ductal progenitors and that removal of nuclear Yap by Lats1/2-mediated signaling is critical for proper ductal maturation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Epiregulin/metabolism , Epithelium/embryology , Morphogenesis , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Submandibular Gland/embryology , Tumor Suppressor Proteins/metabolism , Animals , Body Patterning , Cell Cycle Proteins , Gene Deletion , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Stem Cells/physiology , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
5.
Dev Cell ; 34(3): 283-96, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26235047

ABSTRACT

Epithelial cells undergo dynamic polarity changes as organs pattern, but the relationship between epithelial polarity and cell fate is poorly understood. Using the developing lung as a model, we found that distinct alterations in apical-basal polarity dictate airway epithelial differentiation. We demonstrate that Crb3, a Crumbs isoform that determines epithelial apical domain identity, is required for airway differentiation by controlling the localization of the transcriptional regulator Yap. We show that Crb3 promotes the interaction between Yap and the Hippo pathway kinases Lats1/2 at apical cell junctions to induce Yap phosphorylation and cytoplasmic retention, which drive cell differentiation. Loss of Crb3 in developing mouse airways or isolated adult airway progenitors results in unrestricted nuclear Yap activity and consequent cell differentiation defects. Our findings demonstrate that polarity-dependent cues control airway cell differentiation, offering important molecular insights into organ patterning.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation/genetics , Epithelial Cells/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Lung/embryology , Membrane Glycoproteins , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA Interference , RNA, Small Interfering , Respiratory Mucosa/cytology , Stem Cells/cytology , Trans-Activators/metabolism , YAP-Signaling Proteins
6.
Development ; 142(2): 258-67, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25564622

ABSTRACT

Basal cells are multipotent airway progenitors that generate distinct epithelial cell phenotypes crucial for homeostasis and repair of the conducting airways. Little is known about how these progenitor cells expand and transition to differentiation to form the pseudostratified airway epithelium in the developing and adult lung. Here, we show by genetic and pharmacological approaches that endogenous activation of Notch3 signaling selectively controls the pool of undifferentiated progenitors of upper airways available for differentiation. This mechanism depends on the availability of Jag1 and Jag2, and is key to generating a population of parabasal cells that later activates Notch1 and Notch2 for secretory-multiciliated cell fate selection. Disruption of this mechanism resulted in aberrant expansion of basal cells and altered pseudostratification. Analysis of human lungs showing similar abnormalities and decreased NOTCH3 expression in subjects with chronic obstructive pulmonary disease suggests an involvement of NOTCH3-dependent events in the pathogenesis of this condition.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Epithelial Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Lung/embryology , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Respiratory Mucosa/embryology , Signal Transduction/physiology , Animals , Blotting, Western , Cell Culture Techniques , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Hybridization , Jagged-1 Protein , Mice , Microscopy, Confocal , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Respiratory Mucosa/cytology , Serrate-Jagged Proteins , Species Specificity
7.
Dev Cell ; 30(2): 137-50, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-25043473

ABSTRACT

How epithelial progenitor cells integrate local signals to balance expansion with differentiation during organogenesis is still little understood. Here, we provide evidence that the Hippo pathway effector Yap is a key regulator of this process in the developing lung. We show that when epithelial tubules are forming and branching, a nucleocytoplasmic shift in Yap localization marks the boundary between the airway and the distal lung compartments. At this transition zone, Yap specifies a transcriptional program that controls Sox2 expression and ultimately generates the airway epithelium. Without Yap, epithelial progenitors are unable to properly respond to local TGF-ß-induced cues and control levels and distribution of Sox2 to form airways. Yap levels and subcellular localization also markedly influence Sox2 expression and differentiation of adult airway progenitors. Our data reveal a role for the Hippo-Yap pathway in integrating growth-factor-induced cues in the developing and adult lung potentially key for homeostasis and regeneration repair.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Body Patterning , Cell Differentiation , Embryonic Stem Cells/cytology , Epithelial Cells/cytology , Lung/cytology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Embryonic Stem Cells/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Hippo Signaling Pathway , Lung/embryology , Lung/metabolism , Mice , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , YAP-Signaling Proteins
8.
J Biol Chem ; 289(19): 13461-74, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24648515

ABSTRACT

Uncontrolled transforming growth factor-ß (TGFß) signaling promotes aggressive metastatic properties in late-stage breast cancers. However, how TGFß-mediated cues are directed to induce tumorigenic events is poorly understood, particularly given that TGFß has clear tumor suppressing activity in other contexts. Here, we demonstrate that the transcriptional regulators TAZ and YAP (TAZ/YAP), key effectors of the Hippo pathway, are necessary to promote and maintain TGFß-induced tumorigenic phenotypes in breast cancer cells. Interactions between TAZ/YAP, TGFß-activated SMAD2/3, and TEAD transcription factors reveal convergent roles for these factors in the nucleus. Genome-wide expression analyses indicate that TAZ/YAP, TEADs, and TGFß-induced signals coordinate a specific pro-tumorigenic transcriptional program. Importantly, genes cooperatively regulated by TAZ/YAP, TEAD, and TGFß, such as the novel targets NEGR1 and UCA1, are necessary for maintaining tumorigenic activity in metastatic breast cancer cells. Nuclear TAZ/YAP also cooperate with TGFß signaling to promote phenotypic and transcriptional changes in nontumorigenic cells to overcome TGFß-repressive effects. Our work thus identifies cross-talk between nuclear TAZ/YAP and TGFß signaling in breast cancer cells, revealing novel insight into late-stage disease-driving mechanisms.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Acyltransferases , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules, Neuronal , Cell Cycle Proteins , Cell Line, Tumor , Female , GPI-Linked Proteins , Genome-Wide Association Study , Humans , Neoplasm Metastasis , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...