Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Oncol ; 12: 1009345, 2022.
Article in English | MEDLINE | ID: mdl-36248967

ABSTRACT

Endometrial carcinoma (EC) is the most diagnosed gynecological malignancy in Western countries. Both incidence and mortality rates of EC have steadily risen in recent years. Despite generally favorable prognoses for patients with the endometrioid type of EC, a subset of patients has been identified with decreased progression-free survival. Patients in this group are distinguished from other endometrioid EC patients by the presence of exon 3 hotspot mutations in CTNNB1, the gene encoding for the ß-catenin protein. ß-catenin is an evolutionarily conserved protein with critical functions in both adherens junctions and Wnt-signaling. The exact mechanism by which exon 3 CTNNB1 mutations drive EC progression is not well understood. Further, the potential contribution of mutant ß-catenin to adherens junctions' integrity is not known. Additionally, the magnitude of worsened progression-free survival in patients with CTNNB1 mutations is context dependent, and therefore the importance of this subset of patients can be obscured by improper categorization. This review will examine the history and functions of ß-catenin, how these functions may change and drive EC progression in CTNNB1 mutant patients, and the importance of this patient group in the broader context of the disease.

2.
Cell Rep ; 36(1): 109340, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233186

ABSTRACT

E-cadherin junctions facilitate assembly and disassembly of cell contacts that drive development and homeostasis of epithelial tissues. In this study, using Xenopus embryonic kidney and Madin-Darby canine kidney (MDCK) cells, we investigate the role of the Wnt/planar cell polarity (PCP) formin Daam1 (Dishevelled-associated activator of morphogenesis 1) in regulating E-cadherin-based intercellular adhesion. Using live imaging, we show that Daam1 localizes to newly formed cell contacts in the developing nephron. Furthermore, analyses of junctional filamentous actin (F-actin) upon Daam1 depletion indicate decreased microfilament localization and slowed turnover. We also show that Daam1 is necessary for efficient and timely localization of junctional E-cadherin, mediated by Daam1's formin homology domain 2 (FH2). Finally, we establish that Daam1 signaling promotes organized movement of renal cells. This study demonstrates that Daam1 formin junctional activity is critical for epithelial tissue organization.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nephrons/embryology , Nephrons/metabolism , Xenopus Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Animals , Cadherins/metabolism , Cell Adhesion , Dogs , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Female , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Imaging, Three-Dimensional , Madin Darby Canine Kidney Cells , Male , Nephrons/ultrastructure , Protein Domains , Protein Transport , Xenopus Proteins/chemistry , Xenopus laevis/embryology
3.
PLoS One ; 14(8): e0221698, 2019.
Article in English | MEDLINE | ID: mdl-31469868

ABSTRACT

Kidneys are composed of numerous ciliated epithelial tubules called nephrons. Each nephron functions to reabsorb nutrients and concentrate waste products into urine. Defects in primary cilia are associated with abnormal formation of nephrons and cyst formation in a wide range of kidney disorders. Previous work in Xenopus laevis and zebrafish embryos established that loss of components that make up the Wnt/PCP pathway, Daam1 and ArhGEF19 (wGEF) perturb kidney tubulogenesis. Dishevelled, which activates both the canonical and non-canonical Wnt/PCP pathway, affect cilia formation in multiciliated cells. In this study, we investigated the role of the noncanoncial Wnt/PCP components Daam1 and ArhGEF19 (wGEF) in renal ciliogenesis utilizing polarized mammalian kidney epithelia cells (MDCKII and IMCD3) and Xenopus laevis embryonic kidney. We demonstrate that knockdown of Daam1 and ArhGEF19 in MDCKII and IMCD3 cells leads to loss of cilia, and Daam1's effect on ciliogenesis is mediated by the formin-activity of Daam1. Moreover, Daam1 co-localizes with the ciliary transport protein Ift88 and is present in cilia. Interestingly, knocking down Daam1 in Xenopus kidney does not lead to loss of cilia. These data suggests a new role for Daam1 in the formation of primary cilia.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/genetics , Cilia/metabolism , Epithelial Cells/metabolism , Kidney/cytology , Wnt Proteins/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Animals , Cells, Cultured , Ciliopathies/etiology , Ciliopathies/metabolism , Ciliopathies/pathology , Formins , Gene Knockdown Techniques , Phenotype , Wnt Signaling Pathway , Xenopus laevis
4.
Dev Biol ; 442(2): 301-314, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30118662

ABSTRACT

Disruption of endometrial gland formation or function can cause female infertility. Formation of endometrial glands via tubulogenesis of luminal epithelial cells requires the establishment and maintenance of cell polarity and cell adhesion. The FERM domain-containing protein Merlin coordinates epithelial cell polarity and cell adhesion and is critical for epithelial tissue function in the skin and kidney. We now demonstrate a requirement for Merlin in endometrial gland development. Conditional deletion of Merlin in the endometrium results in female infertility caused by the absence of gland formation. Interestingly, we observed glandular epithelial markers within discrete groups of cells in the Merlin-deficient luminal epithelium. Wnt signaling, a pathway necessary for endometrial gland development is maintained in Merlin-deficient endometrium, suggesting the glandular fate program is active. Instead, we observe increased levels of apical actin and markers indicative of high membrane tension on the basal surface of the Merlin-deficient luminal epithelium. These findings suggest that the structural integrity of the luminal epithelium during gland formation is required for appropriate endometrial tubulogenesis and tissue function. Moreover, our work implicates Merlin-dependent regulation of mechanical tension in the proper formation of endometrial gland architecture and function.


Subject(s)
Endometrium/growth & development , Neurofibromin 2/physiology , Animals , Cell Adhesion/physiology , Cell Polarity/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Endometrium/cytology , Endometrium/metabolism , Endometrium/pathology , Epithelial Cells/metabolism , Female , In Situ Hybridization , Infertility, Female , Mice , Mice, Inbred C57BL , Morphogenesis/physiology , Neurofibromin 2/deficiency , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/physiology
5.
PLoS One ; 12(12): e0189081, 2017.
Article in English | MEDLINE | ID: mdl-29206870

ABSTRACT

Cell adhesion and apicobasal polarity together maintain epithelial tissue organization and homeostasis. Loss of adhesion has been described as a prerequisite for the epithelial to mesenchymal transition. However, what role misregulation of apicobasal polarity promotes tumor initiation and/or early progression remains unclear. We find that human low-grade endometrial cancers are associated with disrupted localization of the apical polarity protein Par3 and Ezrin while, the adhesion molecule E-cadherin remains unchanged, accompanied by decreased Notch signaling, and altered Notch receptor localization. Depletion of Par3 or Ezrin, in a cell-based model, results in loss of epithelial architecture, differentiation, increased proliferation, migration and decreased Notch signaling. Re-expression of Par3 in endometrial cancer cell lines with disrupted Par3 protein levels blocks proliferation and reduces migration in a Notch dependent manner. These data uncover a function for apicobasal polarity independent of cell adhesion in regulating Notch-mediated differentiation signals in endometrial epithelial cells.


Subject(s)
Cell Differentiation , Cell Polarity/physiology , Cell Proliferation , Endometrial Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction , Animals , Dogs , Female , Humans , Madin Darby Canine Kidney Cells
6.
Proc Natl Acad Sci U S A ; 112(6): 1785-90, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25624491

ABSTRACT

Adherens junctions (AJs) and cell polarity complexes are key players in the establishment and maintenance of apical-basal cell polarity. Loss of AJs or basolateral polarity components promotes tumor formation and metastasis. Recent studies in vertebrate models show that loss of AJs or loss of the basolateral component Scribble (Scrib) cause deregulation of the Hippo tumor suppressor pathway and hyperactivation of its downstream effectors Yes-associated protein (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ). However, whether AJs and Scrib act through the same or independent mechanisms to regulate Hippo pathway activity is not known. Here, we dissect how disruption of AJs or loss of basolateral components affect the activity of the Drosophila YAP homolog Yorkie (Yki) during imaginal disc development. Surprisingly, disruption of AJs and loss of basolateral proteins produced very different effects on Yki activity. Yki activity was cell-autonomously decreased but non-cell-autonomously elevated in tissues where the AJ components E-cadherin (E-cad) or α-catenin (α-cat) were knocked down. In contrast, scrib knockdown caused a predominantly cell-autonomous activation of Yki. Moreover, disruption of AJs or basolateral proteins had different effects on cell polarity and tissue size. Simultaneous knockdown of α-cat and scrib induced both cell-autonomous and non-cell-autonomous Yki activity. In mammalian cells, knockdown of E-cad or α-cat caused nuclear accumulation and activation of YAP without overt effects on Scrib localization and vice versa. Therefore, our results indicate the existence of multiple, genetically separable inputs from AJs and cell polarity complexes into Yki/YAP regulation.


Subject(s)
Adherens Junctions/metabolism , Cell Polarity/physiology , Drosophila Proteins/metabolism , Imaginal Discs/growth & development , Intracellular Signaling Peptides and Proteins/metabolism , Morphogenesis/physiology , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Caco-2 Cells , Cadherins/genetics , Cell Adhesion Molecules/genetics , Crosses, Genetic , DNA Primers/genetics , Dogs , Drosophila , Drosophila Proteins/genetics , Gene Knockdown Techniques , Humans , Madin Darby Canine Kidney Cells , Membrane Proteins , RNA Interference , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , alpha Catenin/genetics
7.
Genes Dev ; 26(24): 2709-23, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23249734

ABSTRACT

The ability to generate asymmetry at the cell cortex underlies cell polarization and asymmetric cell division. Here we demonstrate a novel role for the tumor suppressor Merlin and closely related ERM proteins (Ezrin, Radixin, and Moesin) in generating cortical asymmetry in the absence of external cues. Our data reveal that Merlin functions to restrict the cortical distribution of the actin regulator Ezrin, which in turn positions the interphase centrosome in single epithelial cells and three-dimensional organotypic cultures. In the absence of Merlin, ectopic cortical Ezrin yields mispositioned centrosomes, misoriented spindles, and aberrant epithelial architecture. Furthermore, in tumor cells with centrosome amplification, the failure to restrict cortical Ezrin abolishes centrosome clustering, yielding multipolar mitoses. These data uncover fundamental roles for Merlin/ERM proteins in spatiotemporally organizing the cell cortex and suggest that Merlin's role in restricting cortical Ezrin may contribute to tumorigenesis by disrupting cell polarity, spindle orientation, and, potentially, genome stability.


Subject(s)
Cytoskeletal Proteins/metabolism , Neurofibromin 2/metabolism , Animals , Caco-2 Cells , Cell Cycle/physiology , Cell Line, Tumor , Cell Polarity , Centrosome/metabolism , Cytoskeletal Proteins/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Mice , Neurofibromin 2/genetics , Spindle Apparatus/metabolism
8.
Dev Cell ; 19(5): 727-39, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21074722

ABSTRACT

The neurofibromatosis type 2 (NF2) tumor suppressor, Merlin, is a FERM (Four point one, Ezrin, Radixin, Moesin) domain-containing protein whose loss results in defective morphogenesis and tumorigenesis in multiple tissues. Like the closely related ERM proteins (Ezrin, Radixin, and Moesin), Merlin may organize the plasma membrane by assembling membrane protein complexes and linking them to the cortical actin cytoskeleton. We previously found that Merlin is a critical mediator of contact-dependent inhibition of proliferation and is required for the establishment of stable adherens junctions (AJs) in cultured cells. Here, we delineate the molecular function of Merlin in AJ establishment in epidermal keratinocytes in vitro and confirm that a role in AJ establishment is an essential function of Merlin in vivo. Our studies reveal that Merlin can associate directly with α-catenin and link it to Par3, thereby providing an essential link between the AJ and the Par3 polarity complex during junctional maturation.


Subject(s)
Adherens Junctions/metabolism , Cell Polarity , Epidermis/embryology , Epidermis/growth & development , Neurofibromatosis 2/metabolism , Neurofibromin 2/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Epidermal Cells , Epidermis/physiology , Keratinocytes/cytology , Keratinocytes/physiology , Mice , Mice, Transgenic , Neurofibromatosis 2/genetics , Neurofibromin 2/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , alpha Catenin/genetics , alpha Catenin/metabolism
9.
Genes Dev ; 24(16): 1718-30, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20675406

ABSTRACT

The molecular signals that control the maintenance and activation of liver stem/progenitor cells are poorly understood, and the role of liver progenitor cells in hepatic tumorigenesis is unclear. We report here that liver-specific deletion of the neurofibromatosis type 2 (Nf2) tumor suppressor gene in the developing or adult mouse specifically yields a dramatic, progressive expansion of progenitor cells throughout the liver without affecting differentiated hepatocytes. All surviving mice eventually developed both cholangiocellular and hepatocellular carcinoma, suggesting that Nf2(-/-) progenitors can be a cell of origin for these tumors. Despite the suggested link between Nf2 and the Hpo/Wts/Yki signaling pathway in Drosophila, and recent studies linking the corresponding Mst/Lats/Yap pathway to mammalian liver tumorigenesis, our molecular studies suggest that Merlin is not a major regulator of YAP in liver progenitors, and that the overproliferation of Nf2(-/-) liver progenitors is instead driven by aberrant epidermal growth factor receptor (EGFR) activity. Indeed, pharmacologic inhibition of EGFR blocks the proliferation of Nf2(-/-) liver progenitors in vitro and in vivo, consistent with recent studies indicating that the Nf2-encoded protein Merlin can control the abundance and signaling of membrane receptors such as EGFR. Together, our findings uncover a critical role for Nf2/Merlin in controlling homeostasis of the liver stem cell niche.


Subject(s)
Homeostasis/physiology , Liver Neoplasms/physiopathology , Liver/physiopathology , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Stem Cells , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/physiopathology , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Cholangiocarcinoma/genetics , Cholangiocarcinoma/physiopathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Hepatomegaly/genetics , Hepatomegaly/physiopathology , Homeostasis/genetics , Liver/cytology , Liver Neoplasms/genetics , Male , Mice , Neurofibromatosis 2/genetics , Phosphoproteins/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , YAP-Signaling Proteins
10.
Genes Dev ; 21(22): 2908-22, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006686

ABSTRACT

Deregulation of cyclin D1 occurs in numerous human cancers through mutations, alternative splicing, and gene amplification. Although cancer-derived cyclin D1 mutants are potent oncogenes in vitro and in vivo, the mechanisms whereby they contribute to neoplasia are poorly understood. We now provide evidence derived from both mouse models and human cancer-derived cells revealing that nuclear accumulation of catalytically active mutant cyclin D1/CDK4 complexes triggers DNA rereplication, resulting from Cdt1 stabilization, which in turn triggers the DNA damage checkpoint and p53-dependent apoptosis. Loss of p53 through mutations or targeted deletion results in increased genomic instability and neoplastic growth. Collectively, the data presented reveal mechanistic insights into how uncoupling of critical cell cycle regulatory events will perturb DNA replication fidelity, thereby contributing to neoplastic transformation.


Subject(s)
Cell Nucleus/metabolism , Cyclin D1/metabolism , DNA Replication/genetics , S Phase , Tumor Suppressor Protein p53/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Cullin Proteins/metabolism , Cyclin D1/genetics , DNA/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Hydrolysis , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Mutation , NIH 3T3 Cells , Osteosarcoma/pathology , Spleen/cytology , Spleen/metabolism
11.
Blood ; 108(5): 1744-50, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16690963

ABSTRACT

The prognosis for patients with mantle cell lymphoma (MCL) is poor, and at present there is no truly effective therapy. Gene translocation-mediated constitutive expression of cyclin D1 seems to play the key role in the pathogenesis of MCL. Here we report that although 3 of 4 MCL cell lines expressed the recently identified, highly oncogenic cyclin D1b isoform, as well as the canonical cyclin D1a, 8 MCL patient samples expressed only the cyclin D1a protein despite expressing detectable cyclin D1b mRNA. Cell lines and tissue samples displayed constitutive activation of the cyclin D1 signaling cascade, as evidenced by strong expression of CDK4, Rb phosphorylation, and cyclin D1/CDK4 coassociation. All MCL cell lines and tissues examined displayed nondetectable to diminished expression of the cyclin D1 inhibitor p16. Novel small molecule CDK4/CDK6 inhibitor PD0332991 profoundly suppressed--at low nanomolar concentrations--Rb phosphorylation, proliferation, and cell cycle progression at the G0/G1 phase of MCL cells. These findings provide evidence that MCL should be very sensitive to targeted therapy aimed at functional inhibition of the cyclin D1/CDK4 complex.


Subject(s)
Cyclin D1/genetics , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Lymphoma, Mantle-Cell/genetics , Piperazines/pharmacology , Pyridines/pharmacology , Apoptosis , Cell Cycle , Cell Line, Tumor , Genes, bcl-1 , Humans , Lymph Nodes/pathology , Lymphoma, Mantle-Cell/parasitology , Protein Isoforms/genetics , Signal Transduction
12.
J Cell Biochem ; 96(5): 906-13, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16163738

ABSTRACT

The control of cell proliferation is crucial in maintaining cellular homeostasis and loss of this mechanism is a principle hallmark of cancer cells. A primary target of growth factor signaling is the cyclin D1-dependent kinase (D1-CDK4/6) whose activity promotes G1 phase progression by phosphorylating the retinoblastoma protein (Rb) along with related pocket proteins 107 and p130, relieving inhibition of E2F family transcription factors. Cyclin D1 accumulation is regulated at multiple levels including transcription, post-translational activation and cellular localization throughout the cell cycle. While overexpression of cyclin D1 has been observed in a number of human cancers, mouse cancer models overexpressing D1 have fallen short of establishing a role for cyclin D1 in the initiation of malignant phenotypes suggesting an additional regulatory mechanism exists that prevents cyclin D1-driven cancer. This article will present an overview of current data investigating the regulation of cyclin D1 nuclear localization and the prevalence of these aberrations in cancer. Finally, future avenues of research involving cyclin D1 cellular localization and its regulation in cancer will be addressed.


Subject(s)
Cyclin D1/physiology , Neoplasms/metabolism , Active Transport, Cell Nucleus , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Crk-Associated Substrate Protein/metabolism , Cyclin D1/biosynthesis , Cyclin D1/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , G1 Phase , Gene Expression Regulation , Humans , Mice , Models, Biological , Phosphorylation , RNA, Messenger/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/metabolism , Signal Transduction , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism
13.
Cancer Res ; 63(21): 7056-61, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14612495

ABSTRACT

Glycogen synthase kinase-3beta-dependent phosphorylation of cyclin D1 at a conserved COOH-terminal residue, Thr-286, promotes CRM1-dependent cyclin D1 nuclear export at the G(1)-S boundary. Mutations that perturb the phosphorylation of cyclin D1 at Thr-286 contribute to cell transformation, although to date, no such mutations have been found in human cancers. Cyclin D1 (CCND1) undergoes alternative splicing leading to the production of an mRNA predicted to encode a unique cyclin D1 isoform, cyclin D1b, which lacks Thr-286. We have cloned and expressed cyclin D1b, and find that it retains the ability to bind to and activate CDK4. Unlike canonical cyclin D1a, cyclin D1b remains nuclear through the cell cycle where its constitutive expression facilitates cellular transformation. Using antisera specific for cyclin D1b, the protein was detected in a high percentage of esophageal cancer-derived cell lines and in primary esophageal carcinomas. Therefore, alternative splicing leads to expression of a nuclear, oncogenic cyclin D1 isoform that is expressed in human cancer.


Subject(s)
Cyclin D1/genetics , Esophageal Neoplasms/genetics , Oncogenes/physiology , Proto-Oncogene Proteins , Alternative Splicing , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclin D1/biosynthesis , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Esophageal Neoplasms/metabolism , Humans , Male , Mice , Mice, SCID , Molecular Sequence Data , NIH 3T3 Cells , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
14.
Cancer Cell ; 4(3): 160-2, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14522248

ABSTRACT

G1 is the phase of the cell cycle wherein the cell is responsive to growth factor-dependent signals. As such, G1 regulation is frequently disrupted in cancer through deregulation of cyclin/CDK activity; deregulation of G1 phase provides tumorigenic cells with a growth advantage. Cyclin E, the regulatory cyclin for CDK2, is considered a requisite regulator of G1 progression. Cyclin E is overexpressed in cancer, suggesting that cyclin E/CDK2 deregulation contributes to tumorigenesis. Two papers now challenge both the concept that cyclin E/CDK2 is a requisite component of the cell cycle machine and efforts to develop cyclin E/CDK2 inhibitors as antiproliferative therapeutics.


Subject(s)
CDC2-CDC28 Kinases/metabolism , Cell Cycle/physiology , Cyclin E/metabolism , Proto-Oncogene Proteins , Animals , Cells, Cultured , Cyclin D , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Embryo, Mammalian , Fibroblasts/cytology , G1 Phase/physiology , Mice , Models, Molecular , S Phase/physiology
15.
J Biol Chem ; 278(11): 9754-60, 2003 Mar 14.
Article in English | MEDLINE | ID: mdl-12519773

ABSTRACT

The capacity of the cyclin D-dependent kinase to promote G(1) progression through modulation of RB.E2F is well documented. We now demonstrate that the cyclin D1/CDK4 kinase binds to components of the MCM complex. MCM7 and MCM3 were identified as cyclin D1-binding proteins. Catalytically active cyclin D1/CDK4 complexes were incorporated into chromatin-bound protein complexes with the same kinetics as MCM7 and MCM3, where they associated specifically with MCM7. Although the cyclin D1-dependent kinase did not phosphorylate MCM7, active cyclin D1/CDK4, but not cyclin E/CDK2, did catalyze the dissociation of an RB.MCM7 complex. Finally, expression of an active D1/CDK4 kinase but not cyclin E/CDK2 promoted the removal of RB from chromatin-bound protein complexes. Our data suggest that D1/CDK4 complexes play a direct role in altering an inhibitory RB.MCM7 complex possibly allowing for setting of the origin in preparation for DNA replication.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins , Retinoblastoma Protein/metabolism , 3T3 Cells , Animals , Catalysis , Cell Line , Chromatin/metabolism , Cyclin D , Cyclin-Dependent Kinase 4 , Cyclins/metabolism , DNA, Complementary/metabolism , Insecta , Kinetics , Mice , Microscopy, Fluorescence , Minichromosome Maintenance Complex Component 7 , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Time Factors , Transfection , Two-Hybrid System Techniques
16.
J Biol Chem ; 277(10): 8517-23, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11751903

ABSTRACT

There is increasing evidence that p21(Cip1) and p27(Kip1) are requisite positive regulators of cyclin D1.CDK4 assembly and nuclear accumulation. Both Cip and Kip proteins can promote nuclear accumulation of cyclin D1, but the underlying mechanism has not been elucidated. We now provide evidence that p21(Cip1) promotes the nuclear accumulation of cyclin D1 complexes via inhibition of cyclin D1 nuclear export. In vivo, we demonstrate that p21(Cip1) can inhibit glycogen synthase kinase 3 beta-triggered cyclin D1 nuclear export and phosphorylation-dependent nucleocytoplasmic shuttling. Furthermore, we find that cyclin D1 nuclear accumulation in p21/p27 null cells can be restored through inhibition of CRM1-dependent nuclear export. The ability of p21(Cip1) to inhibit cyclin D1 nuclear export correlates with its ability to bind to Thr-286-phosphorylated cyclin D1 and thereby prevents cyclin D1.CRM1 association.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Cyclins/metabolism , Tumor Suppressor Proteins/metabolism , 3T3 Cells , Active Transport, Cell Nucleus , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cell Line , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cytoplasm/metabolism , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Immunoblotting , Insecta , Mice , Microscopy, Fluorescence , Phosphorylation , Protein Binding , Threonine/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...