Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 619(7969): 363-370, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37407814

ABSTRACT

In mammalian cells, the decision to proliferate is thought to be irreversibly made at the restriction point of the cell cycle1,2, when mitogen signalling engages a positive feedback loop between cyclin A2/cyclin-dependent kinase 2 (CDK2) and the retinoblastoma protein3-5. Contrary to this textbook model, here we show that the decision to proliferate is actually fully reversible. Instead, we find that all cycling cells will exit the cell cycle in the absence of mitogens unless they make it to mitosis and divide first. This temporal competition between two fates, mitosis and cell cycle exit, arises because cyclin A2/CDK2 activity depends upon CDK4/6 activity throughout the cell cycle, not just in G1 phase. Without mitogens, mitosis is only observed when the half-life of cyclin A2 protein is long enough to sustain CDK2 activity throughout G2/M. Thus, cells are dependent on mitogens and CDK4/6 activity to maintain CDK2 activity and retinoblastoma protein phosphorylation throughout interphase. Consequently, even a 2-h delay in a cell's progression towards mitosis can induce cell cycle exit if mitogen signalling is lost. Our results uncover the molecular mechanism underlying the restriction point phenomenon, reveal an unexpected role for CDK4/6 activity in S and G2 phases and explain the behaviour of all cells following loss of mitogen signalling.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , G2 Phase , S Phase , Animals , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/deficiency , Cyclin-Dependent Kinase 4/metabolism , Mitogens/deficiency , Mitogens/metabolism , Mitosis , Phosphorylation , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Cyclin-Dependent Kinase 6/deficiency , Cyclin-Dependent Kinase 6/metabolism , G1 Phase
2.
Mol Cell ; 74(4): 758-770.e4, 2019 05 16.
Article in English | MEDLINE | ID: mdl-30982746

ABSTRACT

The cyclin-dependent kinases Cdk4 and Cdk6 form complexes with D-type cyclins to drive cell proliferation. A well-known target of cyclin D-Cdk4,6 is the retinoblastoma protein Rb, which inhibits cell-cycle progression until its inactivation by phosphorylation. However, the role of Rb phosphorylation by cyclin D-Cdk4,6 in cell-cycle progression is unclear because Rb can be phosphorylated by other cyclin-Cdks, and cyclin D-Cdk4,6 has other targets involved in cell division. Here, we show that cyclin D-Cdk4,6 docks one side of an alpha-helix in the Rb C terminus, which is not recognized by cyclins E, A, and B. This helix-based docking mechanism is shared by the p107 and p130 Rb-family members across metazoans. Mutation of the Rb C-terminal helix prevents its phosphorylation, promotes G1 arrest, and enhances Rb's tumor suppressive function. Our work conclusively demonstrates that the cyclin D-Rb interaction drives cell division and expands the diversity of known cyclin-based protein docking mechanisms.


Subject(s)
Cell Proliferation/genetics , Cyclin D/genetics , Protein Interaction Maps/genetics , Retinoblastoma Protein/genetics , Cell Cycle/genetics , Crk-Associated Substrate Protein/genetics , Cyclin D/chemistry , Cyclin-Dependent Kinase 4/chemistry , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/chemistry , Cyclin-Dependent Kinase 6/genetics , Cyclins/genetics , G1 Phase/genetics , Humans , Molecular Docking Simulation , Phosphorylation/genetics , Protein Binding/genetics , Protein Conformation, alpha-Helical/genetics , Retinoblastoma Protein/chemistry , Retinoblastoma-Like Protein p107/genetics , S Phase/genetics
3.
Nature ; 568(7753): 557-560, 2019 04.
Article in English | MEDLINE | ID: mdl-30971822

ABSTRACT

The cell cycle is a tightly regulated process that is controlled by the conserved cyclin-dependent kinase (CDK)-cyclin protein complex1. However, control of the G0-to-G1 transition is not completely understood. Here we demonstrate that p38 MAPK gamma (p38γ) acts as a CDK-like kinase and thus cooperates with CDKs, regulating entry into the cell cycle. p38γ shares high sequence homology, inhibition sensitivity and substrate specificity with CDK family members. In mouse hepatocytes, p38γ induces proliferation after partial hepatectomy by promoting the phosphorylation of retinoblastoma tumour suppressor protein at known CDK target residues. Lack of p38γ or treatment with the p38γ inhibitor pirfenidone protects against the chemically induced formation of liver tumours. Furthermore, biopsies of human hepatocellular carcinoma show high expression of p38γ, suggesting that p38γ could be a therapeutic target in the treatment of this disease.


Subject(s)
Carcinogenesis/pathology , Cell Cycle , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver/enzymology , Liver/pathology , Mitogen-Activated Protein Kinase 12/metabolism , Aged , Animals , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Female , Hepatocytes/cytology , Hepatocytes/pathology , Humans , Liver/surgery , Liver Neoplasms/chemically induced , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 12/antagonists & inhibitors , Phosphorylation , Pyridones/pharmacology , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Sequence Homology , Substrate Specificity
4.
Mol Cell ; 64(1): 25-36, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27642049

ABSTRACT

Control of the G1/S phase transition by the Retinoblastoma (RB) tumor suppressor is critical for the proliferation of normal cells in tissues, and its inactivation is one of the most fundamental events leading to cancer. Cyclin-dependent kinase (CDK) phosphorylation inactivates RB to promote cell cycle-regulated gene expression. Here we show that, upon stress, the p38 stress-activated protein kinase (SAPK) maximizes cell survival by downregulating E2F gene expression through the targeting of RB. RB undergoes selective phosphorylation by p38 in its N terminus; these phosphorylations render RB insensitive to the inactivation by CDKs. p38 phosphorylation of RB increases its affinity toward the E2F transcription factor, represses gene expression, and delays cell-cycle progression. Remarkably, introduction of a RB phosphomimetic mutant in cancer cells reduces colony formation and decreases their proliferative and tumorigenic potential in mice.


Subject(s)
Breast Neoplasms/genetics , Cyclin-Dependent Kinases/genetics , E2F Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Retinoblastoma Protein/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinases/metabolism , E2F Transcription Factors/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Molecular Mimicry , Phosphorylation , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Proc Natl Acad Sci U S A ; 117(52): 33628-33638, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318192

ABSTRACT

Retinoblastoma (Rb) is the most prevalent intraocular malignancy in children, with a worldwide survival rate <30%. We have developed a cancerous model of Rb in retinal organoids derived from genetically engineered human embryonic stem cells (hESCs) with a biallelic mutagenesis of the RB1 gene. These organoid Rbs exhibit properties highly consistent with Rb tumorigenesis, transcriptome, and genome-wide methylation. Single-cell sequencing analysis suggests that Rb originated from ARR3-positive maturing cone precursors during development, which was further validated by immunostaining. Notably, we found that the PI3K-Akt pathway was aberrantly deregulated and its activator spleen tyrosine kinase (SYK) was significantly up-regulated. In addition, SYK inhibitors led to remarkable cell apoptosis in cancerous organoids. In conclusion, we have established an organoid Rb model derived from genetically engineered hESCs in a dish that has enabled us to trace the cell of origin and to test novel candidate therapeutic agents for human Rb, shedding light on the development and therapeutics of other malignancies.


Subject(s)
Human Embryonic Stem Cells/pathology , Organoids/pathology , Retinoblastoma/pathology , Amino Acid Sequence , Animals , Base Sequence , Carcinogenesis/pathology , Human Embryonic Stem Cells/metabolism , Humans , Mice, Inbred NOD , Mutagenesis/genetics , Mutation/genetics , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcriptome/genetics
6.
PLoS Biol ; 17(7): e3000367, 2019 07.
Article in English | MEDLINE | ID: mdl-31323018

ABSTRACT

Human papillomaviruses (HPVs) are causative agents of various diseases associated with cellular hyperproliferation, including cervical cancer, one of the most prevalent tumors in women. E7 is one of the two HPV-encoded oncoproteins and directs recruitment and subsequent degradation of tumor-suppressive proteins such as retinoblastoma protein (pRb) via its LxCxE motif. E7 also triggers tumorigenesis in a pRb-independent pathway through its C-terminal domain, which has yet been largely undetermined, with a lack of structural information in a complex form with a host protein. Herein, we present the crystal structure of the E7 C-terminal domain of HPV18 belonging to the high-risk HPV genotypes bound to the catalytic domain of human nonreceptor-type protein tyrosine phosphatase 14 (PTPN14). They interact directly and potently with each other, with a dissociation constant of 18.2 nM. Ensuing structural analysis revealed the molecular basis of the PTPN14-binding specificity of E7 over other protein tyrosine phosphatases and also led to the identification of PTPN21 as a direct interacting partner of E7. Disruption of HPV18 E7 binding to PTPN14 by structure-based mutagenesis impaired E7's ability to promote keratinocyte proliferation and migration. Likewise, E7 binding-defective PTPN14 was resistant for degradation via proteasome, and it was much more effective than wild-type PTPN14 in attenuating the activity of downstream effectors of Hippo signaling and negatively regulating cell proliferation, migration, and invasion when examined in HPV18-positive HeLa cells. These results therefore demonstrated the significance and therapeutic potential of the intermolecular interaction between HPV E7 and host PTPN14 in HPV-mediated cell transformation and tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Uterine Cervical Neoplasms/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , HEK293 Cells , HeLa Cells , Humans , Models, Molecular , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/genetics , Protein Binding , Protein Domains , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Sequence Homology, Amino Acid , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
7.
Proc Natl Acad Sci U S A ; 114(19): 4942-4947, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439018

ABSTRACT

The retinoblastoma protein (Rb) and the homologous pocket proteins p107 and p130 negatively regulate cell proliferation by binding and inhibiting members of the E2F transcription factor family. The structural features that distinguish Rb from other pocket proteins have been unclear but are critical for understanding their functional diversity and determining why Rb has unique tumor suppressor activities. We describe here important differences in how the Rb and p107 C-terminal domains (CTDs) associate with the coiled-coil and marked-box domains (CMs) of E2Fs. We find that although CTD-CM binding is conserved across protein families, Rb and p107 CTDs show clear preferences for different E2Fs. A crystal structure of the p107 CTD bound to E2F5 and its dimer partner DP1 reveals the molecular basis for pocket protein-E2F binding specificity and how cyclin-dependent kinases differentially regulate pocket proteins through CTD phosphorylation. Our structural and biochemical data together with phylogenetic analyses of Rb and E2F proteins support the conclusion that Rb evolved specific structural motifs that confer its unique capacity to bind with high affinity those E2Fs that are the most potent activators of the cell cycle.


Subject(s)
E2F Transcription Factors/chemistry , Retinoblastoma Protein/chemistry , Retinoblastoma-Like Protein p107/chemistry , Crystallography, X-Ray , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Humans , Protein Domains , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism
8.
Genes Dev ; 26(11): 1156-66, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22569856

ABSTRACT

Cyclin-dependent kinase (Cdk) phosphorylation of the Retinoblastoma protein (Rb) drives cell proliferation through inhibition of Rb complexes with E2F transcription factors and other regulatory proteins. We present the first structures of phosphorylated Rb that reveal the mechanism of its inactivation. S608 phosphorylation orders a flexible "pocket" domain loop such that it mimics and directly blocks E2F transactivation domain (E2F(TD)) binding. T373 phosphorylation induces a global conformational change that associates the pocket and N-terminal domains (RbN). This first multidomain Rb structure demonstrates a novel role for RbN in allosterically inhibiting the E2F(TD)-pocket association and protein binding to the pocket "LxCxE" site. Together, these structures detail the regulatory mechanism for a canonical growth-repressive complex and provide a novel example of how multisite Cdk phosphorylation induces diverse structural changes to influence cell cycle signaling.


Subject(s)
Cell Cycle , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Crystallography, X-Ray , Cyclin-Dependent Kinases/metabolism , Models, Molecular , Phosphorylation
9.
Genes Dev ; 26(11): 1128-30, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22661226

ABSTRACT

In this issue of Genes & Development, Burke and colleagues (pp. 1156-1166) describe how the structure of retinoblastoma protein (pRb) is altered by phosphorylation at T373 or S608. These modifications cause specific conformational changes and alter pRb's interaction with E2F via two distinct mechanisms. The structures suggest that the panel of phosphorylation sites represents a versatile set of tools that are used to sculpt pRb in precise, but very different, ways.


Subject(s)
Cell Cycle , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism
10.
Int J Mol Sci ; 21(14)2020 Jul 12.
Article in English | MEDLINE | ID: mdl-32664691

ABSTRACT

The Retinoblastoma protein (pRb) is a key cell cycle regulator conserved in a wide variety of organisms. Experimental analysis of pRb's functions in animals and plants has revealed that this protein participates in cell proliferation and differentiation processes. In addition, pRb in animals and its orthologs in plants (RBR), are part of highly conserved protein complexes which suggest the possibility that analogies exist not only between functions carried out by pRb orthologs themselves, but also in the structure and roles of the protein networks where these proteins are involved. Here, we present examples of pRb/RBR participation in cell cycle control, cell differentiation, and in the regulation of epigenetic changes and chromatin remodeling machinery, highlighting the similarities that exist between the composition of such networks in plants and animals.


Subject(s)
Cell Cycle Proteins/physiology , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Plant Proteins/physiology , Retinoblastoma Protein/physiology , Animals , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/physiology , Cell Cycle/genetics , Cell Cycle Proteins/chemistry , Cell Differentiation/genetics , DNA Damage , Genes, Plant , Genes, Retinoblastoma , Homeostasis , Mammals/genetics , Mammals/metabolism , Models, Molecular , Multigene Family , Multiprotein Complexes , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , Plant Proteins/chemistry , Plants/genetics , Plants/metabolism , Protein Conformation , Protein Domains , Protein Processing, Post-Translational , Retinoblastoma Protein/chemistry , Species Specificity , Stem Cells/metabolism
11.
Nature ; 498(7454): 390-4, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23783631

ABSTRACT

Allostery is an intrinsic property of many globular proteins and enzymes that is indispensable for cellular regulatory and feedback mechanisms. Recent theoretical and empirical observations indicate that allostery is also manifest in intrinsically disordered proteins, which account for a substantial proportion of the proteome. Many intrinsically disordered proteins are promiscuous binders that interact with multiple partners and frequently function as molecular hubs in protein interaction networks. The adenovirus early region 1A (E1A) oncoprotein is a prime example of a molecular hub intrinsically disordered protein. E1A can induce marked epigenetic reprogramming of the cell within hours after infection, through interactions with a diverse set of partners that include key host regulators such as the general transcriptional coactivator CREB binding protein (CBP), its paralogue p300, and the retinoblastoma protein (pRb; also called RB1). Little is known about the allosteric effects at play in E1A-CBP-pRb interactions, or more generally in hub intrinsically disordered protein interaction networks. Here we used single-molecule fluorescence resonance energy transfer (smFRET) to study coupled binding and folding processes in the ternary E1A system. The low concentrations used in these high-sensitivity experiments proved to be essential for these studies, which are challenging owing to a combination of E1A aggregation propensity and high-affinity binding interactions. Our data revealed that E1A-CBP-pRb interactions have either positive or negative cooperativity, depending on the available E1A interaction sites. This striking cooperativity switch enables fine-tuning of the thermodynamic accessibility of the ternary versus binary E1A complexes, and may permit a context-specific tuning of associated downstream signalling outputs. Such a modulation of allosteric interactions is probably a common mechanism in molecular hub intrinsically disordered protein function.


Subject(s)
Adenovirus E1A Proteins/chemistry , Adenovirus E1A Proteins/metabolism , Allosteric Regulation , Amino Acid Motifs , Animals , Anisotropy , CREB-Binding Protein/chemistry , CREB-Binding Protein/metabolism , Fluorescence Resonance Energy Transfer , Humans , Mice , Models, Molecular , Protein Binding , Protein Folding , Protein Structure, Tertiary , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Thermodynamics , p300-CBP Transcription Factors/chemistry
12.
Nature ; 494(7437): 361-5, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23376950

ABSTRACT

Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1(-/-)Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression.


Subject(s)
Cellular Senescence/immunology , Cytokines/immunology , Neoplasms/immunology , Neoplasms/pathology , Th1 Cells/immunology , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Cell Cycle , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Interferon-gamma/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Oncogenes/genetics , Phosphoserine/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , STAT1 Transcription Factor/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/metabolism
13.
Mol Cell ; 43(4): 663-72, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855804

ABSTRACT

The LXCXE peptide motif facilitates interaction between the RB tumor suppressor and a large number of cellular proteins that are expected to impinge on diverse biological processes. In vitro and in vivo analyses demonstrated that LXCXE binding function is dispensable for RB promoter association and control of basal gene expression. Dependence on this function of RB is unmasked after DNA damage, wherein LXCXE binding is essential for exerting control over E2F3 and suppressing cell-cycle progression in the presence of genotoxic stress. Gene expression profiling revealed that the transcriptional program coordinated by this specific aspect of RB is associated with progression of human hepatocellular carcinoma and poor disease outcome. Consistent with these findings, biological challenge revealed a requirement for LXCXE binding in suppression of genotoxin-initiated hepatocellular carcinoma in vivo. Together, these studies establish an essential role of the LXCXE binding motif for RB-mediated transcriptional control, response to genotoxic insult, and tumor suppression.


Subject(s)
DNA Damage , Gene Expression Regulation , Retinoblastoma Protein/physiology , Transcription, Genetic , Amino Acid Motifs , Animals , Binding Sites , Cell Line , Chromatin/metabolism , Humans , Mice , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism
14.
Trends Biochem Sci ; 38(1): 12-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23218751

ABSTRACT

Multisite phosphorylation modulates the function of regulatory proteins with complex signaling properties and outputs. The retinoblastoma tumor suppressor protein (Rb) is inactivated by cyclin-dependent kinase (Cdk) phosphorylation in normal and cancer cell cycles, so understanding the molecular mechanisms and effects of Rb phosphorylation is imperative. Rb functions in diverse processes regulating proliferation, and it has been speculated that multisite phosphorylation might act as a code in which discrete phosphorylations control specific activities. The idea of an Rb phosphorylation code is evaluated here in light of recent studies of Rb structure and function. Rb inactivation is discussed with an emphasis on how multisite phosphorylation changes Rb structure and associations with protein partners.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinases/metabolism , Retinoblastoma Protein/metabolism , Animals , Humans , Phosphorylation , Protein Conformation , Retinoblastoma Protein/chemistry , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 111(31): 11341-6, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25049398

ABSTRACT

The retinoblastoma tumor suppressor protein pRb is a key regulator of cell cycle progression and mediator of the DNA damage response. Lysine methylation at K810, which occurs within a critical Cdk phosphorylation motif, holds pRb in the hypophosphorylated growth-suppressing state. We show here that methyl K810 is read by the tandem tudor domain containing tumor protein p53 binding protein 1 (53BP1). Structural elucidation of 53BP1 in complex with a methylated K810 pRb peptide emphasized the role of the 53BP1 tandem tudor domain in recognition of the methylated lysine and surrounding residues. Significantly, binding of 53BP1 to methyl K810 occurs on E2 promoter binding factor target genes and allows pRb activity to be effectively integrated with the DNA damage response. Our results widen the repertoire of cellular targets for 53BP1 and suggest a previously unidentified role for 53BP1 in regulating pRb tumor suppressor activity.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Retinoblastoma Protein/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cellular Senescence , Chromatin/metabolism , DNA Repair , Humans , Methylation , Mice , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Retinoblastoma Protein/chemistry , Tumor Suppressor p53-Binding Protein 1
16.
J Biol Chem ; 290(32): 19666-80, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26100623

ABSTRACT

The retinoblastoma (Rb) tumor suppressor restricts cell cycle progression by repressing E2F-responsive transcription. Cellular cyclin-dependent kinase (CDK)-mediated Rb inactivation through phosphorylation disrupts Rb-E2F complexes, stimulating transcription. The human cytomegalovirus (HCMV) UL97 protein is a viral CDK (v-CDK) that phosphorylates Rb. Here we show that UL97 phosphorylates 11 of the 16 consensus CDK sites in Rb. A cleft within Rb accommodates peptides with the amino acid sequence LXCXE. UL97 contains three such motifs. We determined that the first LXCXE motif (L1) of UL97 and the Rb cleft enhance UL97-mediated Rb phosphorylation. A UL97 mutant with a non-functional L1 motif (UL97-L1m) displayed significantly reduced Rb phosphorylation at multiple sites. Curiously, however, it efficiently disrupted Rb-E2F complexes but failed to relieve Rb-mediated repression of E2F reporter constructs. The HCMV immediate early 1 protein cooperated with UL97-L1m to inactivate Rb in transfection assays, likely indicating that cells infected with a UL97-L1m mutant virus show no defects in growth or E2F-responsive gene expression because of redundant viral mechanisms to inactivate Rb. Our data suggest that UL97 possesses a mechanism to elicit E2F-dependent gene expression distinct from disruption of Rb-E2F complexes and dependent upon both the L1 motif of UL97 and the cleft region of Rb.


Subject(s)
Cytomegalovirus/metabolism , E2F1 Transcription Factor/metabolism , Fibroblasts/metabolism , Host-Pathogen Interactions , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Retinoblastoma Protein/metabolism , Binding Sites , Cell Line, Tumor , Cytomegalovirus/genetics , E2F1 Transcription Factor/genetics , Fibroblasts/virology , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mutation , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/genetics , Signal Transduction , Transcription, Genetic
17.
J Virol ; 88(6): 3144-60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24371076

ABSTRACT

UNLABELLED: Interference with tumor suppressor pathways by polyomavirus-encoded tumor antigens (T-Ags) can result in transformation. Consequently, it is thought that T-Ags encoded by Merkel cell polyomavirus (MCPyV), a virus integrated in ∼90% of all Merkel cell carcinoma (MCC) cases, are major contributors to tumorigenesis. The MCPyV large T-Ag (LT-Ag) has preserved the key functional domains present in all family members but has also acquired unique regions that flank the LxCxE motif. As these regions may mediate unique functions, or may modulate those shared with T-Ags of other polyomaviruses, functional studies of MCPyV T-Ags are required. Here, we have performed a comparative study of full-length or MCC-derived truncated LT-Ags with regard to their biochemical characteristics, their ability to bind to retinoblastoma (Rb) and p53 proteins, and their transforming potential. We provide evidence that full-length MCPyV LT-Ag may not directly bind to p53 but nevertheless can significantly reduce p53-dependent transcription in reporter assays. Although early region expression constructs harboring either full-length or MCC-derived truncated LT-Ag genes can transform primary baby rat kidney cells, truncated LT-Ags do not bind to p53 or reduce p53-dependent transcription. Interestingly, shortened LT-Ags exhibit a very high binding affinity for Rb, as shown by coimmunoprecipitation and in vitro binding studies. Additionally, we show that truncated MCPyV LT-Ag proteins are expressed at higher levels than those for the wild-type protein and are able to partially relocalize Rb to the cytoplasm, indicating that truncated LT proteins may have gained additional features that distinguish them from the full-length protein. IMPORTANCE: MCPyV is one of the 12 known polyomaviruses that naturally infect humans. Among these, it is of particular interest since it is the only human polyomavirus known to be involved in tumorigenesis. MCPyV is thought to be causally linked to MCC, a rare skin tumor. In these tumors, viral DNA is monoclonally integrated into the genome of the tumor cells in up to 90% of all MCC cases, and the integrated MCV genomes, furthermore, harbor signature mutations in the so-called early region that selectively abrogate viral replication while preserving cell cycle deregulating functions of the virus. This study describes comparative studies of early region T-Ag protein characteristics, their ability to bind to Rb and p53, and their transforming potential.


Subject(s)
Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/metabolism , Merkel cell polyomavirus/metabolism , Polyomavirus Infections/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Virus Infections/metabolism , Amino Acid Motifs , Animals , Antigens, Viral, Tumor/chemistry , Antigens, Viral, Tumor/genetics , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/pathology , Carcinoma, Merkel Cell/virology , Cell Line, Tumor , Cell Transformation, Viral , Down-Regulation , Humans , Kinetics , Merkel cell polyomavirus/chemistry , Merkel cell polyomavirus/genetics , Polyomavirus Infections/genetics , Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Protein Binding , Protein Transport , Rats , Rats, Sprague-Dawley , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Virus Infections/genetics , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
18.
J Biol Chem ; 288(26): 18923-38, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23632018

ABSTRACT

The retinoblastoma tumor suppressor (Rb) controls the proliferation, differentiation, and survival of cells in most eukaryotes with a role in the fate of stem cells. Its inactivation by mutation or oncogenic viruses is required for cellular transformation and eventually carcinogenesis. The high conservation of the Rb cyclin fold prompted us to investigate the link between conformational stability and ligand binding properties of the RbAB pocket domain. RbAB unfolding presents a three-state transition involving cooperative secondary and tertiary structure changes and a partially folded intermediate that can oligomerize. The first transition corresponds to unfolding of the metastable B subdomain containing the binding site for the LXCXE motif present in cellular and viral targets, and the second transition corresponds to the stable A subdomain. The low thermodynamic stability of RbAB translates into a propensity to rapidly oligomerize and aggregate at 37 °C (T50 = 28 min) that is suppressed by human papillomavirus E7 and E2F peptide ligands, suggesting that Rb is likely stabilized in vivo through binding to target proteins. We propose that marginal stability and associated oligomerization may be conserved for function as a "hub" protein, allowing the formation of multiprotein complexes, which could constitute a robust mechanism to retain its cell cycle regulatory role throughout evolution. Decreased stability and oligomerization are shared with the p53 tumor suppressor, suggesting a link between folding and function in these two essential cell regulators that are inactivated in most cancers and operate within multitarget signaling pathways.


Subject(s)
Cyclins/chemistry , Protein Folding , Retinoblastoma Protein/chemistry , Binding Sites , Cell Differentiation , Circular Dichroism , DNA-Binding Proteins/chemistry , E2F Transcription Factors/chemistry , Humans , Ligands , Models, Molecular , Oncogene Proteins, Viral/chemistry , Papillomavirus E7 Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Signal Transduction , Temperature , Tumor Suppressor Protein p53/chemistry
19.
Theor Biol Med Model ; 10: 1, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23276293

ABSTRACT

BACKGROUND: Lung cancer is the major cause of mortality worldwide. Major signalling pathways that could play significant role in lung cancer therapy include (1) Growth promoting pathways (Epidermal Growth Factor Receptor/Ras/ PhosphatidylInositol 3-Kinase) (2) Growth inhibitory pathways (p53/Rb/P14ARF, STK11) (3) Apoptotic pathways (Bcl-2/Bax/Fas/FasL). Insilico strategy was implemented to solve the mystery behind selected lung cancer pathway by applying comparative modeling and molecular docking studies. RESULTS: YASARA [v 12.4.1] was utilized to predict structural models of P16-INK4 and RB1 genes using template 4ELJ-A and 1MX6-B respectively. WHAT CHECK evaluation tool demonstrated overall quality of predicted P16-INK4 and RB1 with Z-score of -0.132 and -0.007 respectively which showed a strong indication of reliable structure prediction. Protein-protein interactions were explored by utilizing STRING server, illustrated that CDK4 and E2F1 showed strong interaction with P16-INK4 and RB1 based on confidence score of 0.999 and 0.999 respectively. In order to facilitate a comprehensive understanding of the complex interactions between candidate genes with their functional interactors, GRAMM-X server was used. Protein-protein docking investigation of P16-INK4 revealed four ionic bonds illustrating Arg47, Arg80,Cys72 and Met1 residues as actively participating in interactions with CDK4 while docking results of RB1 showed four hydrogen bonds involving Glu864, Ser567, Asp36 and Arg861 residues which interact strongly with its respective functional interactor E2F1. CONCLUSION: This research may provide a basis for understanding biological insights of P16-INK4 and RB1 proteins which will be helpful in future to design a suitable drug to inhibit the disease pathogenesis as we have determined the interacting amino acids which can be targeted in order to design a ligand in-vitro to propose a drug for clinical trials. Protein -protein docking of candidate genes and their important interacting residues likely to be provide a gateway for developing computer aided drug designing.


Subject(s)
Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , E2F1 Transcription Factor/metabolism , Lung Neoplasms/genetics , Molecular Docking Simulation , Retinoblastoma Protein/genetics , Signal Transduction/genetics , Cyclin D1/chemistry , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Lung Neoplasms/metabolism , Protein Binding/genetics , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism
20.
J Biol Chem ; 286(49): 42381-42389, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22006926

ABSTRACT

Zcchc11 is a uridyltransferase protein with enzymatic activity directed against diverse RNA species. On the basis of its known uridylation targets, we hypothesized that Zcchc11 might regulate cell proliferation. Confirming this, loss-of-function and complementary gain-of-function experiments consistently revealed that Zcchc11 promotes the transition from G(1) to S phase of the cell cycle. This activity takes place through both Rb-dependent and Rb-independent mechanisms by promoting the expression of multiple G(1)-associated proteins, including cyclins D(1) and A and CDK4. Surprisingly, a Zcchc11 construct with point mutations inactivating the uridyltransferase domain enhanced cell proliferation as effectively as wild-type Zcchc11. Furthermore, truncated mutant constructs revealed that the cell cycle effects of Zcchc11 were driven by the N-terminal region of the protein that lacks the RNA-binding domains and uridyltransferase activity of the full protein. Therefore, the N-terminal portion of Zcchc11, which lacks nucleotidyltransferase capabilities, is biologically active and mediates a previously unrecognized role for Zcchc11 in facilitating cell proliferation.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Enzymologic , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelial Cells , Flow Cytometry , HeLa Cells , Humans , Mutation , Protein Structure, Tertiary , RNA/chemistry , Retinoblastoma Protein/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL