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










Publication year range
1.
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
2.
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.

3.
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
4.
iScience ; 20: 25-41, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31546104

ABSTRACT

Normal synapse formation is fundamental to brain function. We show here that an apical-basal polarity (A-BP) protein, Lgl1, is present in the postsynaptic density and negatively regulates glutamatergic synapse numbers by antagonizing the atypical protein kinase Cs (aPKCs). A planar cell polarity protein, Vangl2, which inhibits synapse formation, was decreased in synaptosome fractions of cultured cortical neurons from Lgl1 knockout embryos. Conditional knockout of Lgl1 in pyramidal neurons led to reduction of AMPA/NMDA ratio and impaired plasticity. Lgl1 is frequently deleted in Smith-Magenis syndrome (SMS). Lgl1 conditional knockout led to increased locomotion, impaired novel object recognition and social interaction. Lgl1+/- animals also showed increased synapse numbers, defects in open field and social interaction, as well as stereotyped repetitive behavior. Social interaction in Lgl1+/- could be rescued by NMDA antagonists. Our findings reveal a role of apical-basal polarity proteins in glutamatergic synapse development and function and also suggest a potential treatment for SMS patients with Lgl1 deletion.

5.
Neuron ; 94(3): 517-533.e3, 2017 May 03.
Article in English | MEDLINE | ID: mdl-28472654

ABSTRACT

The concerted production of neurons and glia by neural stem cells (NSCs) is essential for neural circuit assembly. In the developing cerebral cortex, radial glia progenitors (RGPs) generate nearly all neocortical neurons and certain glia lineages. RGP proliferation behavior shows a high degree of non-stochasticity, thus a deterministic characteristic of neuron and glia production. However, the cellular and molecular mechanisms controlling RGP behavior and proliferation dynamics in neurogenesis and glia generation remain unknown. By using mosaic analysis with double markers (MADM)-based genetic paradigms enabling the sparse and global knockout with unprecedented single-cell resolution, we identified Lgl1 as a critical regulatory component. We uncover Lgl1-dependent tissue-wide community effects required for embryonic cortical neurogenesis and novel cell-autonomous Lgl1 functions controlling RGP-mediated glia genesis and postnatal NSC behavior. These results suggest that NSC-mediated neuron and glia production is tightly regulated through the concerted interplay of sequential Lgl1-dependent global and cell intrinsic mechanisms.


Subject(s)
Cell Proliferation/genetics , Glycoproteins/genetics , Neocortex/embryology , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neuroglia/metabolism , Neurons/metabolism , Animals , Cell Polarity , Embryo, Mammalian/metabolism , Glycoproteins/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Neocortex/growth & development , Neocortex/pathology , Neural Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology
6.
Dev Cell ; 41(5): 481-495.e5, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28552558

ABSTRACT

Malformations of the cerebral cortex (MCCs) are devastating developmental disorders. We report here that mice with embryonic neural stem-cell-specific deletion of Llgl1 (Nestin-Cre/Llgl1fl/fl), a mammalian ortholog of the Drosophila cell polarity gene lgl, exhibit MCCs resembling severe periventricular heterotopia (PH). Immunohistochemical analyses and live cortical imaging of PH formation revealed that disruption of apical junctional complexes (AJCs) was responsible for PH in Nestin-Cre/Llgl1fl/fl brains. While it is well known that cell polarity proteins govern the formation of AJCs, the exact mechanisms remain unclear. We show that LLGL1 directly binds to and promotes internalization of N-cadherin, and N-cadherin/LLGL1 interaction is inhibited by atypical protein kinase C-mediated phosphorylation of LLGL1, restricting the accumulation of AJCs to the basolateral-apical boundary. Disruption of the N-cadherin-LLGL1 interaction during cortical development in vivo is sufficient for PH. These findings reveal a mechanism responsible for the physical and functional connection between cell polarity and cell-cell adhesion machineries in mammalian cells.


Subject(s)
Brain/abnormalities , Cell Adhesion/physiology , Cell Polarity/physiology , Embryonic Stem Cells/physiology , Homeodomain Proteins/physiology , Neural Stem Cells/physiology , Periventricular Nodular Heterotopia/pathology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Brain/metabolism , Brain/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Proliferation , Cells, Cultured , Cytoskeletal Proteins , Embryonic Stem Cells/cytology , Female , Mice , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/cytology , Periventricular Nodular Heterotopia/metabolism , Phosphorylation
7.
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
8.
F1000Res ; 4(F1000 Faculty Rev): 550, 2015.
Article in English | MEDLINE | ID: mdl-26339481

ABSTRACT

Cadherin-catenin complexes are critical for the assembly of cell-cell adhesion structures known as adherens junctions. In addition to the mechanical linkage of neighboring cells to each other, these cell-cell adhesion protein complexes have recently emerged as important sensors and transmitters of the extracellular cues inside the cell body and into the nucleus. In the past few years, multiple studies have identified a connection between the cadherin-catenin protein complexes and major intracellular signaling pathways. Those studies are the main focus of this review.

9.
Cancer Cell ; 27(6): 797-808, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26058078

ABSTRACT

The significance of ERG in human prostate cancer is unclear because mouse prostate is resistant to ERG-mediated transformation. We determined that ERG activates the transcriptional program regulated by YAP1 of the Hippo signaling pathway and found that prostate-specific activation of either ERG or YAP1 in mice induces similar transcriptional changes and results in age-related prostate tumors. ERG binds to chromatin regions occupied by TEAD/YAP1 and transactivates Hippo target genes. In addition, in human luminal-type prostate cancer cells, ERG binds to the promoter of YAP1 and is necessary for YAP1 expression. These results provide direct genetic evidence of a causal role for ERG in prostate cancer and reveal a connection between ERG and the Hippo signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Oncogene Proteins/genetics , Phosphoproteins/genetics , Prostatic Neoplasms/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Age Factors , Animals , Cell Cycle Proteins , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Transgenic , Oncogene Proteins/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Porphyrins/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Random Allocation , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Transcriptional Regulator ERG , Translocation, Genetic , Up-Regulation , Verteporfin , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
10.
Dev Cell ; 26(6): 553-4, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24091006

ABSTRACT

Transcriptional factor Pitx2 is a key regulator of left-right asymmetry in the developing gut. In this issue of Developmental Cell, Welsh et al. (2013) identify the formin Daam2, an effector of Wnt signaling, as a key cellular target of Pitx2 required for morphogenetic events during asymmetric gut formation.


Subject(s)
Gastrulation , Homeodomain Proteins/metabolism , Intestines/embryology , Microfilament Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Wnt Signaling Pathway , rho GTP-Binding Proteins/metabolism , Animals , Homeobox Protein PITX2
11.
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
12.
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
13.
Sci Signal ; 4(174): ra33, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21610251

ABSTRACT

The Hippo pathway regulates contact inhibition of cell proliferation and, ultimately, organ size in diverse multicellular organisms. Inactivation of the Hippo pathway promotes nuclear localization of the transcriptional coactivator Yap1, a Hippo pathway effector, and can cause cancer. Here, we show that deletion of αE (α epithelial) catenin in the hair follicle stem cell compartment resulted in the development of skin squamous cell carcinoma in mice. Tumor formation was accelerated by simultaneous deletion of αE-catenin and the tumor suppressor-encoding gene p53. A small interfering RNA screen revealed a functional connection between αE-catenin and Yap1. By interacting with Yap1, αE-catenin promoted its cytoplasmic localization, and Yap1 showed constitutive nuclear localization in αE-catenin-null cells. We also found an inverse correlation between αE-catenin abundance and Yap1 activation in human squamous cell carcinoma tumors. These findings identify αE-catenin as a tumor suppressor that inhibits Yap1 activity and sequesters it in the cytoplasm.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Nucleus/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Protein p53/metabolism , alpha Catenin/metabolism , Active Transport, Cell Nucleus/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Nucleus/genetics , Cell Proliferation , HEK293 Cells , Humans , Mice , Mice, Nude , Mice, Transgenic , Phosphoproteins/genetics , Transcription Factors , Tumor Suppressor Protein p53/genetics , YAP-Signaling Proteins , alpha Catenin/genetics
14.
Cell Cycle ; 9(10): 1990-7, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20436275

ABSTRACT

Citron kinase (CitK), a protein essential to neurogenic cell division in the central nervous system, is highly polarized in neural progenitors. The mechanisms that polarize CitK to cellular domains that line the ventricular surface of neuroepithelium are currently not known. Here we report that Discs large 5 (Dlg5), a member of the MAGUK family, is an interactor of CitK required for CitK polarization. The CitK-Dlg5 interaction was first revealed in a protein array screen of proteins containing PDZ domains, and then subsequently confirmed by co-immunoprecipitation. Moreover, in Dlg5 (-/-) mice CitK fails to polarize in mitotic neuronal precursors. In addition, the total number of mitotic progenitors and the ratio of ventricular to abventricular mitotic progenitors in developing neocortex are significantly decreased in Dlg5 (-/-) embryos. Dlg5 is therefore required to maintain the polarization of a protein essential to neurogenic cytokinesis, and plays a role in localizing cell divisions to the surface of the lateral ventricles in embryonic brain.


Subject(s)
Guanylate Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Brain/embryology , Brain/metabolism , Cell Division/physiology , Cells, Cultured , Guanylate Kinases/genetics , Immunohistochemistry , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Lateral Ventricles/embryology , Lateral Ventricles/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitosis/genetics , Mitosis/physiology , Protein Binding , Protein Serine-Threonine Kinases/genetics , Rats
15.
J Cell Sci ; 121(Pt 9): 1357-62, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18397997

ABSTRACT

beta-Catenin is a crucial mediator of the canonical Wnt-signaling pathway. alpha-catenin is a major beta-catenin-binding protein, and overexpressed alpha-catenin can negatively regulate beta-catenin activity. Thus, alpha-catenin may be an important modulator of the Wnt pathway. We show here that endogenous alpha-catenin has little impact on the transcriptional activity of beta-catenin in developing mammalian organisms. We analyzed beta-catenin signaling in mice with conditional deletion of alphaE-catenin (Ctnna1) in the developing central nervous system. This mutation results in brain hyperplasia and we investigated whether activation of beta-catenin signaling may be at least partially responsible for this phenotype. To reveal potential quantitative or spatial changes in beta-catenin signaling, we used mice carrying a beta-catenin-signaling reporter transgene. In addition, we analyzed the expression of known endogenous targets of the beta-catenin pathway and the amount and localization of beta-catenin in mutant progenitor cells. We found that although loss of alphaE-catenin resulted in disruption of intercellular adhesion and hyperplasia in the developing brain, beta-catenin signaling was not altered. We conclude that endogenous alphaE-catenin has no significant impact on beta-catenin transcriptional activities in the developing mammalian brain.


Subject(s)
Brain/embryology , Mammals/embryology , Signal Transduction , alpha Catenin/metabolism , beta Catenin/metabolism , Animals , Brain/pathology , Cell Nucleus/metabolism , Genes, Reporter , Hyperplasia , Protein Binding , Protein Transport , TCF Transcription Factors/metabolism , Transcriptional Activation/genetics , alpha Catenin/deficiency , beta Catenin/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.
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
18.
Science ; 311(5767): 1609-12, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16543460

ABSTRACT

During development, cells monitor and adjust their rates of accumulation to produce organs of predetermined size. We show here that central nervous system-specific deletion of the essential adherens junction gene, alphaE-catenin, causes abnormal activation of the hedgehog pathway, resulting in shortening of the cell cycle, decreased apoptosis, and cortical hyperplasia. We propose that alphaE-catenin connects cell-density-dependent adherens junctions with the developmental hedgehog pathway and that this connection may provide a negative feedback loop controlling the size of developing cerebral cortex.


Subject(s)
Adherens Junctions/physiology , Cerebral Cortex/embryology , Neurons/physiology , Signal Transduction , Trans-Activators/metabolism , alpha Catenin/physiology , Adherens Junctions/ultrastructure , Animals , Apoptosis , Cell Adhesion , Cell Count , Cell Cycle , Cell Differentiation , Cell Polarity , Central Nervous System/embryology , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Hedgehog Proteins , Hyperplasia , Mice , Mitosis , Models, Biological , Mutation , Neurons/cytology , Neurons/ultrastructure , Oligonucleotide Array Sequence Analysis , Stem Cells/cytology , Stem Cells/ultrastructure , Up-Regulation , alpha Catenin/genetics
19.
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
20.
Genes Dev ; 18(5): 559-71, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-15037549

ABSTRACT

Disruption of cell polarity is seen in many cancers; however, it is generally considered a late event in tumor progression. Lethal giant larvae (Lgl) has been implicated in maintenance of cell polarity in Drosophila and cultured mammalian cells. We now show that loss of Lgl1 in mice results in formation of neuroepithelial rosette-like structures, similar to the neuroblastic rosettes in human primitive neuroectodermal tumors. The newborn Lgl1(-/-) pups develop severe hydrocephalus and die neonatally. A large proportion of Lgl1(-/-) neural progenitor cells fail to exit the cell cycle and differentiate, and, instead, continue to proliferate and die by apoptosis. Dividing Lgl1(-/-) cells are unable to asymmetrically localize the Notch inhibitor Numb, and the resulting failure of asymmetric cell divisions may be responsible for the hyperproliferation and the lack of differentiation. These results reveal a critical role for mammalian Lgl1 in regulating of proliferation, differentiation, and tissue organization and demonstrate a potential causative role of disruption of cell polarity in neoplastic transformation of neuroepithelial cells.


Subject(s)
Cell Polarity/physiology , Cell Transformation, Neoplastic/pathology , Neurocutaneous Syndromes/etiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Brain Diseases/embryology , Brain Diseases/etiology , Brain Diseases/pathology , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic/genetics , Embryonic and Fetal Development/genetics , Epithelial Cells/pathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurocutaneous Syndromes/embryology , Neurocutaneous Syndromes/pathology , Neuroectodermal Tumors, Primitive/pathology , Proteins/genetics , Proteins/physiology , Stem Cells/pathology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...