Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
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.
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
3.
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.

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 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
6.
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
7.
Science ; 370(6520): 1036-1037, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33243876
8.
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
9.
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
10.
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.

11.
F1000Res ; 72018.
Article in English | MEDLINE | ID: mdl-30135717

ABSTRACT

Prostate cancer (PCa) is a disease of mutated and misregulated genes. However, primary prostate tumors have relatively few mutations, and only three genes ( ERG, PTEN, and SPOP) are recurrently mutated in more than 10% of primary tumors. On the other hand, metastatic castration-resistant tumors have more mutations, but, with the exception of the androgen receptor gene ( AR), no single gene is altered in more than half of tumors. Structural genomic rearrangements are common, including ERG fusions, copy gains involving the MYC locus, and copy losses containing PTEN. Overall, instead of being associated with a single dominant driver event, prostate tumors display various combinations of modifications in oncogenes and tumor suppressors. This review takes a broad look at the recent advances in PCa research, including understanding the genetic alterations that drive the disease and how specific mutations can sensitize tumors to potential therapies. We begin with an overview of the genomic landscape of primary and metastatic PCa, enabled by recent large-scale sequencing efforts. Advances in three-dimensional cell culture techniques and mouse models for PCa are also discussed, and particular emphasis is placed on the benefits of patient-derived xenograft models. We also review research into understanding how ETS fusions (in particular, TMPRSS2-ERG) and SPOP mutations contribute to tumor initiation. Next, we examine the recent findings on the prevalence of germline DNA repair mutations in about 12% of patients with metastatic disease and their potential benefit from the use of poly(ADP-ribose) polymerase (PARP) inhibitors and immune modulation. Lastly, we discuss the recent increased prevalence of AR-negative tumors (neuroendocrine and double-negative) and the current state of immunotherapy in PCa. AR remains the primary clinical target for PCa therapies; however, it does not act alone, and better understanding of supporting mutations may help guide the development of novel therapeutic strategies.


Subject(s)
DNA Repair , Genomics , Mutation , Prostatic Neoplasms/genetics , Animals , Humans , Male , Molecular Targeted Therapy , Neoplasm Metastasis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology
13.
Anticancer Res ; 37(7): 3385-3396, 2017 07.
Article in English | MEDLINE | ID: mdl-28668826

ABSTRACT

BACKGROUND/AIM: The aim of the current study was to determine the effects of the ERG small-molecule inhibitor YK-4-279 on ERG+ prostate cancer patient-derived xenografts (PDX). MATERIALS AND METHODS: ERG activity was blocked using YK-4-279 in three subcutaneously-implanted ERG+ (LuCaP 23.1, 86.2 and 35) and one ERG- (LuCaP 96) PDX. Treated animals tumor volume (TV), body weight (BW) and serum prostate-specific antigen (PSA) were compared to vehicle-treated control animals. Gene expression, proliferation, apoptosis, microvessel density and ERG expression were also assessed. RESULTS: Administration of YK-4-279 decreased TV (p=0.026), proliferation (p=0.0038) and PSA (p=0.022) in Severe Combined Immunodeficiency (SCID) mice bearing LuCaP 23.1 tumors. LuCaP 86.2, LuCaP 35 and LuCaP 96 showed no significant changes in TV, or PSA. Mineralocorticoid receptor (MR) and MR-direct target genes were up-regulated in treatment-resistant LuCaP 86.2 and LuCaP 35 PDX. CONCLUSION: YK-4-279 decreased ERG+ LuCaP 23.1 tumor growth, but not LuCaP 86.2 and LuCaP 35 ERG+ tumor growth.


Subject(s)
Heterografts/drug effects , Indoles/pharmacology , Prostatic Neoplasms/drug therapy , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Body Weight/drug effects , Body Weight/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Gene Expression/drug effects , Gene Expression/genetics , Heterografts/metabolism , Humans , Male , Mice , Mice, SCID , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Mineralocorticoid/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Transcriptional Regulator ERG/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
14.
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
15.
Sci Rep ; 7(1): 1109, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28439080

ABSTRACT

Mechanistic studies of deregulated ERG in prostate cancer and other cancers continue to enhance its role in cancer biology and its utility as a biomarker and therapeutic target. Here, we show that ERG, through its physical interaction with androgen receptor, induces AR aggregation and endoplasmic reticulum stress in the prostate glands of ERG transgenic mice. Histomorphological alterations and the expression of ER stress sensors Atf6, Ire1α, Perk, their downstream effectors Grp78/BiP and eIF2α in ERG transgenic mouse prostate glands indicate the presence of chronic ER stress. Transient activation of apoptotic cell death during early age correlated well with the differential regulation of ER stress sensors, in particular Perk. Epithelial cells derived from ERG transgenic mouse prostates have increased prostasphere formation with resistance to radiation induced cell death. Continued activation of cell survival factors, Atf6 and Ire1α during chronic ER stress due to presence of ERG in prostate epithelium induces survival pathways and provides a selection pressure in the continuum of ERG dependent neoplastic process. These novel insights will enhance the understanding of the mechanistic functions of ERG in prostate tumor biology and towards development of early targeted therapeutic strategies for prostate cancer.


Subject(s)
Endoplasmic Reticulum Stress , Prostatic Neoplasms/physiopathology , Protein Aggregation, Pathological , Receptors, Androgen/metabolism , Animals , Endoplasmic Reticulum Chaperone BiP , Gene Expression Profiling , Histocytochemistry , Immunohistochemistry , Male , Mice, Transgenic , Microscopy , Prostate/pathology , Transcriptional Regulator ERG/metabolism
16.
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
17.
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
18.
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.

19.
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
20.
Oncotarget ; 5(5): 1352-62, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24657880

ABSTRACT

The development of effective therapies inhibiting prostate cancer progression and metastasis may substantially impact prostate cancer mortality and potentially reduce the rates of invasive treatments by enhancing the safety of active surveillance strategies. Hepsin (HPN) is a cell surface serine protease amplified in a subset of human sarcomas (7.2%), as well as in ovarian (10%), lung adeno (5.4%), lung squamous cell (4.5%), adenoid cystic (5%), breast (2.6%), uterine (1.7%) and colon (1.4%) carcinomas. While HPN is not amplified in prostate cancer, it is one of the most prominently overexpressed genes in the majority of human prostate tumors and genetic experiments in mice indicate that Hepsin promotes prostate cancer metastasis, particularly metastasis to the bone marrow. We report here the development, analysis and animal trial of the small-molecule Hepsin inhibitor HepIn-13. Long-term exposure to HepIn-13 inhibited bone, liver and lung metastasis in a murine model of metastatic prostate cancer. These findings indicate that inhibition of Hepsin with small-molecule compounds could provide an effective tool for attenuation of prostate cancer progression and metastasis.


Subject(s)
Adenocarcinoma/drug therapy , Bone Neoplasms/prevention & control , Membrane Proteins/antagonists & inhibitors , Naphthalenes/pharmacokinetics , Naphthalenes/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Administration, Oral , Animals , Biological Availability , Bone Neoplasms/secondary , HEK293 Cells , Half-Life , Humans , Inhibitory Concentration 50 , Male , Mice , Models, Molecular , Naphthalenes/pharmacology , Prostatic Neoplasms/metabolism , Pyrimidines/pharmacology , Serine Proteinase Inhibitors/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...