Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Genes Dev ; 34(15-16): 1051-1064, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32675324

ABSTRACT

YAP1 is a transcriptional coactivator and the principal effector of the Hippo signaling pathway, which is causally implicated in human cancer. Several YAP1 gene fusions have been identified in various human cancers and identifying the essential components of this family of gene fusions has significant therapeutic value. Here, we show that the YAP1 gene fusions YAP1-MAMLD1, YAP1-FAM118B, YAP1-TFE3, and YAP1-SS18 are oncogenic in mice. Using reporter assays, RNA-seq, ChIP-seq, and loss-of-function mutations, we can show that all of these YAP1 fusion proteins exert TEAD-dependent YAP activity, while some also exert activity of the C'-terminal fusion partner. The YAP activity of the different YAP1 fusions is resistant to negative Hippo pathway regulation due to constitutive nuclear localization and resistance to degradation of the YAP1 fusion proteins. Genetic disruption of the TEAD-binding domain of these oncogenic YAP1 fusions is sufficient to inhibit tumor formation in vivo, while pharmacological inhibition of the YAP1-TEAD interaction inhibits the growth of YAP1 fusion-expressing cell lines in vitro. These results highlight TEAD-dependent YAP activity found in these gene fusions as critical for oncogenesis and implicate these YAP functions as potential therapeutic targets in YAP1 fusion-positive tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/genetics , Oncogene Proteins, Fusion/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Gene Expression Regulation , Humans , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Nuclear Localization Signals , Nucleotide Motifs , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/chemistry , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
2.
Genes Dev ; 30(7): 798-811, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27013234

ABSTRACT

Cell-cell adhesion protein αE-catenin inhibits skin squamous cell carcinoma (SCC) development; however, the mechanisms responsible for this function are not completely understood. We report here that αE-catenin inhibits ß4 integrin-mediated activation of SRC tyrosine kinase.SRCis the first discovered oncogene, but the protein substrate critical for SRC-mediated transformation has not been identified. We found that YAP1, the pivotal effector of the Hippo signaling pathway, is a direct SRC phosphorylation target, and YAP1 phosphorylation at three sites in its transcription activation domain is necessary for SRC-YAP1-mediated transformation. We uncovered a marked increase in this YAP1 phosphorylation in human and mouse SCC tumors with low/negative expression of αE-catenin. We demonstrate that the tumor suppressor function of αE-catenin involves negative regulation of the ß4 integrin-SRC signaling pathway and that SRC-mediated phosphorylation and activation of YAP1 are an alternative to the canonical Hippo signaling pathway that directly connect oncogenic tyrosine kinase signaling with YAP1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/physiopathology , Oncogene Protein pp60(v-src)/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , alpha Catenin/metabolism , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Keratinocytes/cytology , Keratinocytes/pathology , Mice , Phosphorylation , Protein Transport , YAP-Signaling Proteins
3.
Genes Dev ; 30(24): 2696-2709, 2016 12 15.
Article in English | MEDLINE | ID: mdl-28087714

ABSTRACT

Disruption of apical-basal polarity is implicated in developmental disorders and cancer; however, the mechanisms connecting cell polarity proteins with intracellular signaling pathways are largely unknown. We determined previously that membrane-associated guanylate kinase (MAGUK) protein discs large homolog 5 (DLG5) functions in cell polarity and regulates cellular proliferation and differentiation via undefined mechanisms. We report here that DLG5 functions as an evolutionarily conserved scaffold and negative regulator of Hippo signaling, which controls organ size through the modulation of cell proliferation and differentiation. Affinity purification/mass spectrometry revealed a critical role of DLG5 in the formation of protein assemblies containing core Hippo kinases mammalian ste20 homologs 1/2 (MST1/2) and Par-1 polarity proteins microtubule affinity-regulating kinases 1/2/3 (MARK1/2/3). Consistent with this finding, Hippo signaling is markedly hyperactive in mammalian Dlg5-/- tissues and cells in vivo and ex vivo and in Drosophila upon dlg5 knockdown. Conditional deletion of Mst1/2 fully rescued the phenotypes of brain-specific Dlg5 knockout mice. Dlg5 also interacts genetically with Hippo effectors Yap1/Taz Mechanistically, we show that DLG5 inhibits the association between MST1/2 and large tumor suppressor homologs 1/2 (LATS1/2), uses its scaffolding function to link MST1/2 with MARK3, and inhibits MST1/2 kinase activity. These data reveal a direct connection between cell polarity proteins and Hippo, which is essential for proper development of multicellular organisms.


Subject(s)
Cell Polarity/genetics , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Drosophila/embryology , Drosophila/enzymology , Drosophila/genetics , Gene Deletion , Gene Knockdown Techniques , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Protein Binding , Protein Domains , Protein Serine-Threonine Kinases/genetics , Proteomics , RNA Interference , Tumor Suppressor Proteins/genetics
4.
EMBO Rep ; 22(11): e52532, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34515392

ABSTRACT

Transforming growth factor-beta (TGFß) is a multifunctional cytokine with a well-established role in mammary gland development and both oncogenic and tumor-suppressive functions. The extracellular matrix (ECM) indirectly regulates TGFß activity by acting as a storage compartment of latent-TGFß, but how TGFß is released from the ECM via proteolytic mechanisms remains largely unknown. In this study, we demonstrate that hepsin, a type II transmembrane protease overexpressed in 70% of breast tumors, promotes canonical TGFß signaling through the release of latent-TGFß from the ECM storage compartment. Mammary glands in hepsin CRISPR knockout mice showed reduced TGFß signaling and increased epithelial branching, accompanied by increased levels of fibronectin and latent-TGFß1, while overexpression of hepsin in mammary tumors increased TGFß signaling. Cell-free and cell-based experiments showed that hepsin is capable of direct proteolytic cleavage of fibronectin but not latent-TGFß and, importantly, that the ability of hepsin to activate TGFß signaling is dependent on fibronectin. Altogether, this study demonstrates a role for hepsin as a regulator of the TGFß pathway in the mammary gland via a novel mechanism involving proteolytic downmodulation of fibronectin.


Subject(s)
Fibronectins , Transforming Growth Factor beta , Animals , Fibronectins/metabolism , Mice , Proteolysis , Serine Endopeptidases/genetics , Transforming Growth Factor beta/metabolism
5.
Dev Biol ; 475: 205-221, 2021 07.
Article in English | MEDLINE | ID: mdl-33428889

ABSTRACT

YAP1 is a transcriptional co-activator whose activity is controlled by the Hippo signaling pathway. In addition to important functions in normal tissue homeostasis and regeneration, YAP1 has also prominent functions in cancer initiation, aggressiveness, metastasis, and therapy resistance. In this review we are discussing the molecular functions of YAP1 and its roles in cancer, with a focus on the different mechanisms of de-regulation of YAP1 activity in human cancers, including inactivation of upstream Hippo pathway tumor suppressors, regulation by intersecting pathways, miRNAs, and viral oncogenes. We are also discussing new findings on the function and biology of the recently identified family of YAP1 gene fusions, that constitute a new type of activating mutation of YAP1 and that are the likely oncogenic drivers in several subtypes of human cancers. Lastly, we also discuss different strategies of therapeutic inhibition of YAP1 functions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Oncogene Proteins, Fusion/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Oncogenes/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , YAP-Signaling Proteins
6.
Development ; 139(9): 1599-610, 2012 May.
Article in English | MEDLINE | ID: mdl-22492354

ABSTRACT

To gain insights into the cellular mechanisms of neurogenesis, we analyzed retinal neuroepithelia deficient for Llgl1, a protein implicated in apicobasal cell polarity, asymmetric cell division, cell shape and cell cycle exit. We found that vertebrate retinal neuroepithelia deficient for Llgl1 retained overt apicobasal polarity, but had expanded apical domains. Llgl1 retinal progenitors also had increased Notch activity and reduced rates of neurogenesis. Blocking Notch function by depleting Rbpj restored normal neurogenesis. Experimental expansion of the apical domain, through inhibition of Shroom3, also increased Notch activity and reduced neurogenesis. Significantly, in wild-type retina, neurogenic retinal progenitors had smaller apical domains compared with proliferative neuroepithelia. As nuclear position during interkinetic nuclear migration (IKNM) has been previously linked with cell cycle exit, we analyzed this phenomenon in cells depleted of Llgl1. We found that although IKNM was normal, the relationship between nuclear position and neurogenesis was shifted away from the apical surface, consistent with increased pro-proliferative and/or anti-neurogenic signals associated with the apical domain. These data, in conjunction with other findings, suggest that, in retinal neuroepithelia, the size of the apical domain modulates the strength of polarized signals that influence neurogenesis.


Subject(s)
Cell Cycle Proteins/deficiency , Neuroepithelial Cells/metabolism , Neurogenesis/physiology , Receptors, Notch/metabolism , Retina/cytology , Zebrafish Proteins/deficiency , Zebrafish/physiology , Animals , Bromodeoxyuridine , Cell Cycle Proteins/metabolism , Microfilament Proteins/metabolism , Oligonucleotides/genetics , Time-Lapse Imaging , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Dev Biol ; 377(2): 375-84, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23466739

ABSTRACT

Cell polarity plays an important role in tissue morphogenesis; however, the mechanisms of polarity and their role in mammalian development are still poorly understood. We show here that membrane-associated guanylate kinase protein Dlg5 is required for proper branching morphogenesis and progenitor differentiation in mammalian lung. We found that during lung development Dlg5 functions as an apical-basal polarity protein, which is necessary for the apical maintenance of atypical protein kinase C (aPKC). These results identify Dlg5 as a regulator of apical polarity complexes and uncover the critical function of Dlg5 in branching morphogenesis and differentiation of lung progenitor cells.


Subject(s)
Cell Differentiation/physiology , Cell Polarity/genetics , Guanylate Kinases/metabolism , Lung/embryology , Membrane Proteins/metabolism , Morphogenesis/physiology , Protein Kinase C/metabolism , Stem Cells/physiology , Animals , Blotting, Western , DNA Primers/genetics , Fluorescent Antibody Technique , Lung/cytology , Lung/metabolism , Mice , Mice, Knockout , Microarray Analysis
8.
Curr Opin Cell Biol ; 18(5): 499-506, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16859905

ABSTRACT

Cadherin-catenin adhesion is pivotal for the development of multicellular organisms. Features such as a large repertoire of homotypically interacting cadherins, rapid assembly and disassembly, and a connection to a force-generating actin cytoskeleton make cadherin-mediated junctions ideal structures for the execution of complex changes in cell and tissue morphology during development. Recent findings highlight the role of cadherin-catenin proteins as critical regulators of major developmental pathways. We re-evaluate the significance of cadherin-catenin adhesion structures and propose that in addition to intercellular adhesion, they may be used as biosensors of the external cellular environment that help adjust the behavior of individual cells to ensure survival of the entire organism.


Subject(s)
Cadherins/metabolism , Catenins/metabolism , Embryo, Mammalian , Embryo, Nonmammalian , Morphogenesis , Animals , Body Patterning , Central Nervous System/embryology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Epidermis/anatomy & histology , Epidermis/embryology , Neural Crest/anatomy & histology , Neural Crest/physiology , Signal Transduction/physiology
9.
Subcell Biochem ; 60: 379-414, 2012.
Article in English | MEDLINE | ID: mdl-22674080

ABSTRACT

Cadherins and catenins are the central cell-cell adhesion molecules in adherens junctions (AJs). This chapter reviews the knowledge concerning the role of cadherins and catenins in epithelial cancer and examines the published literature demonstrating the changes in the expression and function of these proteins in human cancer and the association of these changes with patient outcomes. The chapter also covers the mechanistic studies aiming at uncovering the significance of changes in cadherin and catenin expression in cancer and potential molecular mechanisms responsible for the causal role of AJs in cancer initiation and progression.


Subject(s)
Adherens Junctions/physiology , Cell Adhesion Molecules/metabolism , Neoplasms/pathology , Signal Transduction , Animals , Humans , Neoplasms/metabolism
10.
bioRxiv ; 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36945368

ABSTRACT

Loss of cell polarity and tissue disorganization occurs in majority of epithelial cancers. Studies in simple model organisms identified molecular mechanisms responsible for the establishment and maintenance of cellular polarity, which play a pivotal role in establishing proper tissue architecture. The exact role of these cell polarity pathways in mammalian cancer is not completely understood. Here we analyzed the mammalian orthologs of drosophila apical-basal polarity gene lethal giant larvae ( lgl ), which regulates asymmetric stem cell division and functions as a tumor suppressor in flies. There are two mammalian orthologs of lgl ( Llgl1 and Llgl2 ). To determine the role of the entire lgl signaling pathway in mammals we generated mice with ablation of both Llgl1 and Llgl2 in skin epidermis using K14-Cre ( Llgl1/2 -/- cKO mice). Surprisingly, we found that ablation of Llgl1/2 genes does not impact epidermal polarity in adult mice. However, old Llgl1/2 cKO mice present with focal skin lesions which are missing epidermal layer and ripe with inflammation. To determine the role of lgl signaling pathway in cancer we generated Trp53 -/- /Llgl1/2 -/- cKO and Trp53 -/+ /Llgl1/2 -/- cKO mice. Loss of Llgl1/2 promoted squamous cell carcinoma (SCC) development in Trp53 -/- cKO and caused SCC in Trp53 -/+ cKO mice, while no cancer was observed in Trp53 -/+ cKO controls. Mechanistically, we show that ablation of Llgl1/2 causes activation of aPKC and upregulation of NF-kB signaling pathway, which may be necessary for SCC in Trp53 -/+ /Llgl1/2 -/- cKO mice. We conclude that Lgl signaling pathway functions as a tumor suppressor in mammalian skin epidermis.

11.
Nat Commun ; 14(1): 7435, 2023 11 16.
Article in English | MEDLINE | ID: mdl-37973913

ABSTRACT

SND1 and MTDH are known to promote cancer and therapy resistance, but their mechanisms and interactions with other oncogenes remain unclear. Here, we show that oncoprotein ERG interacts with SND1/MTDH complex through SND1's Tudor domain. ERG, an ETS-domain transcription factor, is overexpressed in many prostate cancers. Knocking down SND1 in human prostate epithelial cells, especially those overexpressing ERG, negatively impacts cell proliferation. Transcriptional analysis shows substantial overlap in genes regulated by ERG and SND1. Mechanistically, we show that ERG promotes nuclear localization of SND1/MTDH. Forced nuclear localization of SND1 prominently increases its growth promoting function irrespective of ERG expression. In mice, prostate-specific Snd1 deletion reduces cancer growth and tumor burden in a prostate cancer model (PB-Cre/Ptenflox/flox/ERG mice), Moreover, we find a significant overlap between prostate transcriptional signatures of ERG and SND1. These findings highlight SND1's crucial role in prostate tumorigenesis, suggesting SND1 as a potential therapeutic target in prostate cancer.


Subject(s)
Prostatic Neoplasms , Animals , Humans , Male , Mice , Cell Transformation, Neoplastic/genetics , Endonucleases/genetics , Endonucleases/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Tudor Domain
12.
Leukemia ; 37(10): 2027-2035, 2023 10.
Article in English | MEDLINE | ID: mdl-37587260

ABSTRACT

Scribble complex proteins can influence cell fate decisions and self-renewal capacity of hematopoietic cells. While specific cellular functions of Scribble complex members are conserved in mammalian hematopoiesis, they appear to be highly context dependent. Using CRISPR/Cas9-based genetic screening, we have identified Scribble complex-related liabilities in AML including LLGL1. Despite its reported suppressive function in HSC self-renewal, inactivation of LLGL1 in AML confirms its relevant role for proliferative capacity and development of AML. Its function was conserved in human and murine models of AML and across various genetic backgrounds. Inactivation of LLGL1 results in loss of stemness-associated gene-expression including HoxA-genes and induces a GMP-like phenotype in the leukemia stem cell compartment. Re-expression of HoxA9 facilitates functional and phenotypic rescue. Collectively, these data establish LLGL1 as a specific dependency and putative target in AML and emphasizes its cell-type specific functions.


Subject(s)
Cytoskeletal Proteins , Hematopoietic Stem Cells , Leukemia, Myeloid, Acute , Animals , Humans , Mice , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Cytoskeletal Proteins/genetics
13.
Dev Cell ; 13(3): 338-50, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17765678

ABSTRACT

Epithelial tubes represent fundamental building blocks of metazoan organisms; however, the mechanisms responsible for their formation and maintenance are not well understood. Here, we show that the evolutionarily conserved coiled-coil MAGUK protein Dlg5 is required for epithelial tube maintenance in mammalian brain and kidneys. We demonstrate that Dlg5(-/-) mice develop fully penetrant hydrocephalus and kidney cysts caused by a deficiency in membrane delivery of cadherin-catenin adhesion complexes and loss of cell polarity. Dlg5 travels with cadherin-containing vesicles and binds to syntaxin 4, a t-SNARE protein that regulates fusion of transport vesicles with the lateral membrane domain. We propose that Dlg5 functions in plasma membrane delivery of cadherins by linking cadherin-containing transport vesicles with the t-SNARE targeting complex. These findings show that Dlg5 is causally involved in hydrocephalus and renal cysts and reveal that targeted membrane delivery of cadherin-catenin adhesion complexes is critical for cell polarity and epithelial tube maintenance.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Hydrocephalus/etiology , Polycystic Kidney Diseases/etiology , Adherens Junctions/metabolism , Animals , Animals, Newborn , Cadherins/genetics , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Cell Polarity , Cells, Cultured , Dogs , Electroporation , Epithelial Cells/metabolism , Fibroblasts/metabolism , Guanylate Kinases/genetics , Humans , Hydrocephalus/pathology , In Situ Hybridization , Kidney/cytology , Kinetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Biological , Polycystic Kidney Diseases/pathology , Qa-SNARE Proteins/metabolism , Transfection , beta Catenin/metabolism
14.
Cancer Cell ; 6(2): 185-95, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15324701

ABSTRACT

The majority of cancer-related deaths are associated with metastasis; however, little is known about the mechanisms of this process. Hepsin is a cell surface serine protease that is markedly upregulated in human prostate cancer; however, the functional significance of this upregulation is unknown. We report here that hepsin overexpression in prostate epithelium in vivo causes disorganization of the basement membrane. Overexpression of hepsin in a mouse model of nonmetastasizing prostate cancer has no impact on cell proliferation, but causes disorganization of the basement membrane and promotes primary prostate cancer progression and metastasis to liver, lung, and bone. We provide in vivo evidence that upregulation of a cell surface serine protease in a primary tumor promotes cancer progression and metastasis.


Subject(s)
Neoplasm Metastasis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Serine Endopeptidases/metabolism , Animals , Apoptosis , Basement Membrane/metabolism , Basement Membrane/pathology , Basement Membrane/ultrastructure , Cell Differentiation , Cell Division , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Prostate/anatomy & histology , Prostate/metabolism , Prostate/pathology , Serine Endopeptidases/genetics
15.
Dev Cell ; 10(2): 153-4, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16459292

ABSTRACT

Cell divisions are necessary, but also very disruptive for morphogenesis. Dividing cells lose many intercellular contacts and polarized features. This breaks the magnificent topology of the developing embryo and, if left unrepaired, can lead to severe tissue disorganization. A recent study demonstrated that cells use the planar cell polarity pathway to reestablish polarity and reintegrate daughter cells into developing tissue.


Subject(s)
Cell Division/physiology , Cell Polarity/physiology , Animals , Cell Adhesion , Cell Division/genetics , Cell Polarity/genetics , Morphogenesis , Mutation , Nervous System/embryology , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics
16.
Proc Natl Acad Sci U S A ; 105(6): 2105-10, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18245377

ABSTRACT

A significant proportion of human prostate cancers carry a chromosomal rearrangement resulting in the overexpression of the ETS transcription factor, ERG; however, the functional significance of this event is poorly understood. We report here that up-regulation of ERG transcript is sufficient for the initiation of prostate neoplasia. In agreement with measurements of ERG transcripts, we found that ERG protein is expressed in neoplastic human prostate epithelium. Overexpression of ERG in prostate cell lines increased cell invasion. Moreover, targeted expression of this transcript in vivo in luminal prostate epithelial cells of transgenic mice results in initiation of prostate neoplasia observed as the development of focal precancerous prostatic intraepithelial neoplasia (PIN). Similar to human cancers, luminal epithelial cells in these PIN lesions displace diminishing in numbers basal epithelial cells and establish direct contact with the stromal cell compartment. Loss of basal cells is considered to be one of the critical hallmarks of human prostate cancer; however, the mechanisms responsible for this event were unknown. We propose that up-regulation of ERG in human prostate cancer activates cell invasion programs that subsequently displace basal cells by neoplastic epithelium. Our data demonstrate that ERG plays an important causal role in the transformation of prostate epithelium and should be considered as a target for prevention or early therapeutic intervention.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/physiology , Prostate/pathology , Prostatic Intraepithelial Neoplasia/pathology , Trans-Activators/physiology , Animals , Cell Line , DNA-Binding Proteins/genetics , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Male , Mice , Prostate/enzymology , Prostatic Intraepithelial Neoplasia/enzymology , RNA, Messenger/genetics , Serine Endopeptidases/metabolism , Trans-Activators/genetics , Transcriptional Regulator ERG
17.
Dev Cell ; 56(6): 761-780.e7, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33725480

ABSTRACT

Vinculin, a mechanotransducer associated with both adherens junctions (AJs) and focal adhesions (FAs), plays a central role in force transmission through cell-cell and cell-substratum contacts. We generated the conditional knockout (cKO) of vinculin in murine skin that results in the loss of bulge stem cell (BuSC) quiescence and promotes continual cycling of the hair follicles. Surprisingly, we find that the AJs in vinculin cKO cells are mechanically weak and impaired in force generation despite increased junctional expression of E-cadherin and α-catenin. Mechanistically, we demonstrate that vinculin functions by keeping α-catenin in a stretched/open conformation, which in turn regulates the retention of YAP1, another potent mechanotransducer and regulator of cell proliferation, at the AJs. Altogether, our data provide mechanistic insights into the hitherto-unexplored regulatory link between the mechanical stability of cell junctions and contact-inhibition-mediated maintenance of BuSC quiescence.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/physiology , Hair Follicle/physiology , Mechanotransduction, Cellular , Stem Cells/physiology , Vinculin/physiology , alpha Catenin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion , Female , Hair Follicle/cytology , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Stem Cells/cytology , YAP-Signaling Proteins , alpha Catenin/genetics
18.
Prostate ; 70(6): 591-600, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19938013

ABSTRACT

BACKGROUND: Hepsin is a cell surface protease that is over-expressed in more than 90% of human prostate cancer cases. The previously developed Probasin-hepsin/Large Probasin-T antigen (PB-hepsin/LPB-Tag) bigenic mouse model of prostate cancer demonstrates that hepsin promotes primary tumors that are a mixture of adenocarcinoma and neuroendocrine (NE) lesions, and metastases that are NE in nature. However, since the majority of human prostate tumors are adenocarcinomas, the contribution of hepsin in the progression of adenocarcinoma requires further investigation. METHODS: We crossed the PB-hepsin mice with PB-Hi-myc transgenic mouse model of prostate adenocarcinoma and characterized the tumor progression in the resulting PB-hepsin/PB-Hi-myc bigenic mice. RESULTS: We report that PB-hepsin/PB-Hi-myc bigenic mice develop invasive adenocarcinoma at 4.5 months. Further, histological analysis of the 12- to 17-month-old mice revealed that the PB-hepsin/PB-Hi-myc model develops a higher grade adenocarcinoma compared with age-matched tumors expressing only PB-Hi-myc. Consistent with targeting hepsin to the prostate, the PB-hepsin/PB-Hi-myc tumors showed higher hepsin expression as compared to the age-matched myc tumors. Furthermore, endogenous expression of hepsin increased in the PB-Hi-myc mice as the tumors progressed. CONCLUSIONS: Although we did not detect any metastases from the prostates in either the PB-hepsin/PB-Hi-myc or the PB-Hi-myc mice, our data suggests that hepsin and myc cooperate during the progression to high-grade prostatic adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Disease Progression , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Serine Endopeptidases/metabolism , Adenocarcinoma/pathology , Androgen-Binding Protein/genetics , Androgen-Binding Protein/metabolism , Animals , Disease Models, Animal , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Serine Endopeptidases/genetics , Time Factors
19.
Urol Oncol ; 38(9): 736.e1-736.e10, 2020 09.
Article in English | MEDLINE | ID: mdl-32674955

ABSTRACT

OBJECTIVE: ERG rearrangements are frequent and early events in prostate cancer. The functional role of rearranged ERG, however, is still incompletely understood. ERG rearrangements are maintained during prostate cancer progression suggesting that they may confer a selective advantage. The molecular basis of this notion is the subject of this study. METHODS: A variety of immunological methods were used to characterize the effects of rearranged ERG on p53. Consequences of an overexpression of N-terminally deleted ERG on p53 function were interrogated by measuring apoptosis and cellular senescence in the presence or absence of exogenous DNA damage. Effects of N-terminally deleted ERG on the transactivation function of p53 were analyzed by qRT-PCR. RESULTS: We show that overexpression of ERG leads to an increased basal level of DNA damage and a stabilization of p53 that involves a sequestration of its E3 ubiquitin ligase, MDM2, into nucleoli. A higher p53 expression was also observed in vivo in an ERG-overexpressing prostatic intraepithelial neoplasia mouse model. The correlation between ERG and p53 expression was corroborated in 163 patients with prostate cancer. ERG overexpression was found to inhibit both apoptosis and cellular senescence induced by exogenous DNA damage. Mechanistically, this protective effect of ERG involved an abrogation of the DNA damage-induced expression of p53 target genes. CONCLUSIONS: By protecting tumor cells from the antiproliferative consequences of genotoxic stress, ERG may allow the survival and proliferation of genomically unstable tumor cells. Targeting ERG may therefore represent a promising strategy to suppress such adverse features during prostate cancer progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Rearrangement , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Aged , Animals , Humans , Male , Mice , Middle Aged , Transcriptional Regulator ERG/genetics , Tumor Cells, Cultured
20.
Dev Cell ; 3(3): 367-81, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12361600

ABSTRACT

To enable stratification and barrier function, the epidermis must permit self-renewal while maintaining adhesive connections. By generating K14-GFP-actin mice to monitor actin dynamics in cultured primary keratinocytes, we uncovered a role for the actin cytoskeleton in establishing cellular organization. During epidermal sheet formation, a polarized network of nascent intercellular junctions and radial actin cables assemble in the apical plane of the monolayer. These actin fibers anchor to a central actin-myosin network, creating a tension-based plane of cytoskeleton across the apical surface of the sheet. Movement of the sheet surface relative to its base expands the zone of intercellular overlap, catalyzing new sites for nascent intercellular junctions. This polarized cytoskeleton is dependent upon alpha-catenin, Rho, and Rock, and its regulation may be important for wound healing and/or stratification, where coordinated tissue movements are involved.


Subject(s)
Actins/physiology , Cytoskeleton/ultrastructure , Epidermis/ultrastructure , Protein Serine-Threonine Kinases/physiology , rho GTP-Binding Proteins/physiology , Actin Cytoskeleton/ultrastructure , Adherens Junctions/physiology , Adherens Junctions/ultrastructure , Amides/pharmacology , Animals , Animals, Newborn , Cadherins/metabolism , Calcium/pharmacology , Cell Polarity , Cells, Cultured , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/physiology , Desmosomes/ultrastructure , Enzyme Inhibitors/pharmacology , Epidermal Cells , Epidermis/physiology , Epithelial Cells/ultrastructure , Green Fluorescent Proteins , Intracellular Signaling Peptides and Proteins , Keratinocytes/physiology , Keratinocytes/ultrastructure , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Models, Biological , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Recombinant Fusion Proteins/metabolism , Transgenes/physiology , alpha Catenin , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL