Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Nat Commun ; 15(1): 2017, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38443376

ABSTRACT

HIV-1 infection elevates the risk of developing various cancers, including T-cell lymphoma. Whether HIV-1-encoded proteins directly contribute to oncogenesis remains unknown. We observe that approximately 1-5% of CD4+ T cells from the blood of people living with HIV-1 exhibit over-duplicated centrioles, suggesting that centrosome amplification underlies the development of HIV-1-associated cancers by driving aneuploidy. Through affinity purification, biochemical, and cellular analyses, we discover that Vpr, an accessory protein of HIV-1, hijacks the centriole duplication machinery and induces centrosome amplification and aneuploidy. Mechanistically, Vpr forms a cooperative ternary complex with an E3 ligase subunit, VprBP, and polo-like kinase 4 (Plk4). Unexpectedly, however, the complex enhances Plk4's functionality by promoting its relocalization to the procentriole assembly and induces centrosome amplification. Loss of either Vpr's C-terminal 17 residues or VprBP acidic region, the two elements required for binding to Plk4 cryptic polo-box, abrogates Vpr's capacity to induce these events. Furthermore, HIV-1 WT, but not its Vpr mutant, induces multiple centrosomes and aneuploidy in human primary CD4+ T cells. We propose that the Vpr•VprBP•Plk4 complex serves as a molecular link that connects HIV-1 infection to oncogenesis and that inhibiting the Vpr C-terminal motif may reduce the occurrence of HIV-1-associated cancers.


Subject(s)
HIV-1 , T-Lymphocytes , Humans , Centrosome , Carcinogenesis , Cell Transformation, Neoplastic , Aneuploidy , CD4-Positive T-Lymphocytes
2.
Res Sq ; 2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37645926

ABSTRACT

HIV-1 infection elevates the risk of developing various cancers, including T-cell lymphoma. Whether HIV-1-encoded proteins directly contribute to oncogenesis remains unknown. We observed that approximately 1-5% of CD4+ T cells from the blood of people living with HIV-1 exhibit over-duplicated centrioles, suggesting that centrosome amplification underlies the development of HIV-1-associated cancers by driving aneuploidy. Through affinity purification, biochemical, and cell biology analyses, we discovered that Vpr, an accessory protein of HIV-1, hijacks the centriole duplication machinery and induces centrosome amplification and aneuploidy. Mechanistically, Vpr formed a cooperative ternary complex with an E3 ligase subunit, VprBP, and polo-like kinase 4 (Plk4). Unexpectedly, however, the complex enhanced Plk4's functionality by promoting its relocalization to the procentriole assembly and induced centrosome amplification. Loss of either Vpr's C-terminal 17 residues or VprBP acidic region, the two elements required for binding to Plk4 cryptic polo-box, abrogated Vpr's capacity to induce all these events. Furthermore, HIV-1 WT, but not its Vpr mutant, induced multiple centrosomes and aneuploidy in primary CD4+ T cells. We propose that the Vpr•VprBP•Plk4 complex serves as a molecular link that connects HIV-1 infection to oncogenesis and that inhibiting the Vpr C-terminal motif may reduce the occurrence of HIV-1-associated cancers.

3.
Proc Natl Acad Sci U S A ; 120(35): e2305037120, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37603740

ABSTRACT

Polo-like kinase 1 (Plk1) is considered an attractive target for anticancer therapy. Over the years, studies on the noncatalytic polo-box domain (PBD) of Plk1 have raised the expectation of generating highly specific protein-protein interaction inhibitors. However, the molecular nature of the canonical PBD-dependent interaction, which requires extensive water network-mediated interactions with its phospholigands, has hampered efforts to identify small molecules suitable for Plk1 PBD drug discovery. Here, we report the identification of the first allosteric inhibitor of Plk1 PBD, called Allopole, a prodrug that can disrupt intracellular interactions between PBD and its cognate phospholigands, delocalize Plk1 from centrosomes and kinetochores, and induce mitotic block and cancer cell killing. At the structural level, its unmasked active form, Allopole-A, bound to a deep Trp-Phe-lined pocket occluded by a latch-like loop, whose adjoining region was required for securely retaining a ligand anchored to the phospho-binding cleft. Allopole-A binding completely dislodged the L2 loop, an event that appeared sufficient to trigger the dissociation of a phospholigand and inhibit PBD-dependent Plk1 function during mitosis. Given Allopole's high specificity and antiproliferative potency, this study is expected to open an unexplored avenue for developing Plk1 PBD-specific anticancer therapeutic agents.


Subject(s)
Cell Cycle Proteins , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Cell Nucleus Division , Polo-Like Kinase 1
4.
Commun Biol ; 6(1): 712, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37433832

ABSTRACT

Proper organization of intracellular assemblies is fundamental for efficient promotion of biochemical processes and optimal assembly functionality. Although advances in imaging technologies have shed light on how the centrosome is organized, how its constituent proteins are coherently architected to elicit downstream events remains poorly understood. Using multidisciplinary approaches, we showed that two long coiled-coil proteins, Cep63 and Cep152, form a heterotetrameric building block that undergoes a stepwise formation into higher molecular weight complexes, ultimately generating a cylindrical architecture around a centriole. Mutants defective in Cep63•Cep152 heterotetramer formation displayed crippled pericentriolar Cep152 organization, polo-like kinase 4 (Plk4) relocalization to the procentriole assembly site, and Plk4-mediated centriole duplication. Given that the organization of pericentriolar materials (PCM) is evolutionarily conserved, this work could serve as a model for investigating the structure and function of PCM in other species, while offering a new direction in probing the organizational defects of PCM-related human diseases.


Subject(s)
Centrioles , Centrosome , Protein Serine-Threonine Kinases , Humans , Cell Cycle , Molecular Weight , Protein Domains , Protein Serine-Threonine Kinases/metabolism
6.
ACS Pharmacol Transl Sci ; 6(3): 422-446, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36926457

ABSTRACT

Polo-like kinase 1 (Plk1), a mitotic kinase whose activity is widely upregulated in various human cancers, is considered an attractive target for anticancer drug discovery. Aside from the kinase domain, the C-terminal noncatalytic polo-box domain (PBD), which mediates the interaction with the enzyme's binding targets or substrates, has emerged as an alternative target for developing a new class of inhibitors. Various reported small molecule PBD inhibitors exhibit poor cellular efficacy and/or selectivity. Here, we report structure-activity relationship (SAR) studies on triazoloquinazolinone-derived inhibitors, such as 43 (a 1-thioxo-2,4-dihydrothieno[2,3-e][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-one) that effectively block Plk1, but not Plk2 and Plk3 PBDs, with improved affinity and drug-like properties. The range of prodrug moieties needed for thiol group masking of the active drugs has been expanded to increase cell permeability and mechanism-based cancer cell (L363 and HeLa) death. For example, a 5-thio-1-methyl-4-nitroimidazolyl prodrug 80, derived from 43, showed an improved cellular potency (GI50 4.1 µM). As expected, 80 effectively blocked Plk1 from localizing to centrosomes and kinetochores and consequently induced potent mitotic block and apoptotic cell death. Another prodrug 78 containing 9-fluorophenyl in place of the thiophene-containing heterocycle in 80 also induced a comparable degree of anti-Plk1 PBD effect. However, orally administered 78 was rapidly converted in the bloodstream to parent drug 15, which was shown be relatively stable toward in vivo oxidation due to its 9-fluorophenyl group in comparison to unsubstituted phenyl. Further derivatization of these inhibitors, particularly to improve the systemic prodrug stability, could lead to a new class of therapeutics against Plk1-addicted cancers.

7.
J Med Chem ; 65(3): 1915-1932, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35029981

ABSTRACT

The polo-box domain (PBD) of Plk1 is a promising target for cancer therapeutics. We designed and synthesized novel phosphorylated macrocyclic peptidomimetics targeting PBD based on acyclic phosphopeptide PMQSpTPL. The inhibitory activities of 16e on Plk1-PBD is >30-fold higher than those of PMQSpTPL. Both 16a and 16e possess excellent selectivity for Plk1-PBD over Plk2/3-PBD. Analysis of the cocrystal structure of Plk1-PBD in complex with 16a reveals that the 3-(trifluoromethyl)benzoyl group in 16a interacts with Arg516 through a π-stacking interaction. This π-stacking interaction, which has not been reported previously, provides insight into the design of novel and potent Plk1-PBD inhibitors. Furthermore, 16h, a PEGlyated macrocyclic phosphopeptide derivative, induces Plk1 delocalization and mitotic failure in HeLa cells. Also, the number of phospho-H3-positive cells in a zebrafish embryo increases in proportion to the amount of 16a. Collectively, the novel macrocyclic peptidomimetics should serve as valuable templates for the design of potent and novel Plk1-PBD inhibitors.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Peptidomimetics/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Docking Simulation , Molecular Structure , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Peptidomimetics/chemical synthesis , Peptidomimetics/metabolism , Protein Binding , Protein Domains , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Structure-Activity Relationship , Zebrafish , Polo-Like Kinase 1
8.
Exp Mol Med ; 53(11): 1769-1780, 2021 11.
Article in English | MEDLINE | ID: mdl-34837016

ABSTRACT

C1q/TNF-related protein 1 (CTRP1) is a CTRP family member that has collagenous and globular C1q-like domains. The secreted form of CTRP1 is known to be associated with cardiovascular and metabolic diseases, but its cellular roles have not yet been elucidated. Here, we showed that cytosolic CTRP1 localizes to the endoplasmic reticulum (ER) membrane and that knockout or depletion of CTRP1 leads to mitochondrial fission defects, as demonstrated by mitochondrial elongation. Mitochondrial fission events are known to occur through an interaction between mitochondria and the ER, but we do not know whether the ER and/or its associated proteins participate directly in the entire mitochondrial fission event. Interestingly, we herein showed that ablation of CTRP1 suppresses the recruitment of DRP1 to mitochondria and provided evidence suggesting that the ER-mitochondrion interaction is required for the proper regulation of mitochondrial morphology. We further report that CTRP1 inactivation-induced mitochondrial fission defects induce apoptotic resistance and neuronal degeneration, which are also associated with ablation of DRP1. These results demonstrate for the first time that cytosolic CTRP1 is an ER transmembrane protein that acts as a key regulator of mitochondrial fission, providing new insight into the etiology of metabolic and neurodegenerative disorders.


Subject(s)
Adipokines/metabolism , Dynamins/metabolism , Endoplasmic Reticulum/metabolism , Mitochondrial Dynamics , Adipokines/genetics , Animals , Cell Line , Humans , Male , Mice , Mice, Knockout , Protein Binding
10.
Curr Opin Struct Biol ; 66: 66-73, 2021 02.
Article in English | MEDLINE | ID: mdl-33176265

ABSTRACT

Pericentriolar material (PCM) present around a pair of centrioles functions as a platform for various cellular processes, including microtubule (MT) assembly. While PCM is known to be an electron-dense proteinaceous matrix made of long coiled-coil proteins and their client molecules, the molecular mechanism underlying PCM organization remains largely elusive. A growing body of evidence suggests that PCM is constructed in part by an interphase cylindrical self-assembly and the mitotic mesh-like architectures surrounding it. In this review, we will discuss how these higher-order structures are constructed to achieve the functional proficiency of the centrosome.


Subject(s)
Cell Cycle Proteins , Centrosome , Centrioles , Humans , Microtubules
11.
Cell Cycle ; 19(24): 3419-3436, 2020 12.
Article in English | MEDLINE | ID: mdl-33323015

ABSTRACT

Polo-like kinase 4 (Plk4) is a key regulator of centriole biogenesis. Studies have shown that Plk4 undergoes dynamic relocalization from a ring-like pattern around a centriole to a dot-like morphology at the procentriole assembly site and this event is central for inducing centriole biogenesis. However, the detailed mechanisms underlying Plk4's capacity to drive its symmetry-breaking ring-to-dot relocalization remain largely unknown. Here, we showed that Plk4 self-initiates this process in an autophosphorylation-dependent manner and that STIL, its downstream target, is not required for this event. Time-dependent analyses with mEOS-fused photoconvertible Plk4 revealed that a portion of ring-state Plk4 acquires a capacity, presumably through autophosphorylation, to linger around a centriole, ultimately generating a dot-state morphology. Interestingly, Plk4 WT, but not its catalytically inactive mutant, showed the ability to form a nanoscale spherical assembly in the cytosol of human cells or heterologous E. coli, demonstrating its autophosphorylation-dependent self-organizing capacity. At the biochemical level, Plk4 - unlike its N-terminal ßTrCP degron motif - robustly autophosphorylated the PC3 SSTT motif within its C-terminal cryptic polo-box, an event critical for inducing its physical clustering. Additional in vivo experiments showed that although STIL was not required for Plk4's initial ring-to-dot conversion, coexpressed STIL greatly enhanced Plk4's ability to generate a spherical condensate and recruit Sas6, a major component of the centriolar cartwheel structure. We propose that Plk4's autophosphorylation-induced clustering is sufficient to induce its ring-to-dot localization conversion and that subsequently recruited STIL potentiates this process to generate a procentriole assembly body critical for Plk4-dependent centriole biogenesis.


Subject(s)
Centrioles/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Biocatalysis , Cell Cycle/genetics , Cell Line, Tumor , Cytosol/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Osteosarcoma/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Protein Serine-Threonine Kinases/genetics , Proteolysis , RNA Interference , Transfection
12.
Cell Cycle ; 19(24): 3437-3457, 2020 12.
Article in English | MEDLINE | ID: mdl-33208041

ABSTRACT

The centrosome is a unique membraneless organelle that plays a pivotal role in the orderly progression of the cell cycle in animal cells. It has been shown that two pericentriolar scaffold proteins, Cep63 and Cep152, generate a heterotetrameric complex to self-assemble into a higher-order cylindrical architecture around a centriole. However, the mechanisms underlying how they reach their threshold concentrations in the vast intracellular space and generate a self-assembled architecture remain mysterious. Here we demonstrate that, like liquid-like assemblies, Cep63 and Cep152 cooperatively generate amorphous aggregates capable of undergoing dynamic turnover and inter-aggregate fusion in vivo and a significant level of internal rearrangemefnt within a condensate in vitro. Consistently, 1,6-hexanediol, a liquid-liquid phase separation disruptor, greatly diminished the ability of endogenous Cep63 and Cep152 to localize to centrosomes. Interestingly, a purified Cep63•Cep152 complex generated either a cylindrical structure or a vesicle-like hollow sphere in a spatially controlled manner. It also formed condensate-like solid spheres in the presence of a macromolecular crowder. At the molecular level, two hydrophobic motifs, one each from Cep63 and Cep152, were required for generating phase-separating condensates and a high molecular-weight assembly. Thus, we propose that the self-assembly of the Cep63•Cep152 complex is triggered by an intrinsic property of the complex undergoing density transition through the hydrophobic-motif-mediated phase separation. Abbreviations: PCM, pericentriolar material; LLPS, liquid-liquid phase separation; MW, molecular-weight; CLEM, correlative light and electron microscopy; WT, wild-type; CMV, cytomegalovirus; FRAP, fluorescence recovery after photobleaching; FITC, fluorescein isothiocyanate; PCR, polymerase chain reaction; 3D-SIM, three-dimensional structured illumination microscopy; DMEM, Dulbecco's Modified Eagle Medium; PEI Max, Polyethylenimine Max; PBS, phosphate-buffered saline; RT, room temperature; DAPI, 4', 6-diamidino-2-phenylindole; AOTF, acousto-optic tunable filter; LB, Luria broth; OD, optical density; IPTG, isopropyl ß-D-1-thiogalactopyranoside; SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Centrioles/metabolism , Liquid-Liquid Extraction/methods , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Weight , Plasmids/genetics , Plasmids/metabolism , Protein Aggregates/genetics , Protein Conformation, alpha-Helical , Protein Domains/genetics , Transfection
13.
J Med Chem ; 63(22): 14087-14117, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33175530

ABSTRACT

As a mitotic-specific target widely deregulated in various human cancers, polo-like kinase 1 (Plk1) has been extensively explored for anticancer activity and drug discovery. Although multiple catalytic domain inhibitors were tested in preclinical and clinical studies, their efficacies are limited by dose-limiting cytotoxicity, mainly from off-target cross reactivity. The C-terminal noncatalytic polo-box domain (PBD) of Plk1 has emerged as an attractive target for generating new protein-protein interaction inhibitors. Here, we identified a 1-thioxo-2,4-dihydro-[1,2,4]triazolo[4,3-a]quinazolin-5(1H)-one scaffold that efficiently inhibits Plk1 PBD but not its related Plk2 and Plk3 PBDs. Structure-activity relationship studies led to multiple inhibitors having ≥10-fold higher inhibitory activity than the previously characterized Plk1 PBD-specific phosphopeptide, PLHSpT (Kd ∼ 450 nM). In addition, S-methyl prodrugs effectively inhibited mitotic progression and cell proliferation and their metabolic stability was determined. These data describe a novel class of small-molecule inhibitors that offer a promising avenue for future drug discovery against Plk1-addicted cancers.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Drug Discovery , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasms/enzymology , Neoplasms/pathology , Protein Binding , Protein Domains , Protein Kinase Inhibitors/pharmacokinetics , Structure-Activity Relationship , Tissue Distribution , Polo-Like Kinase 1
14.
J Med Chem ; 63(23): 14905-14920, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33142063

ABSTRACT

Polo-like kinase-1 (Plk1) plays a key role in mitosis and has been identified as an attractive anticancer drug target. Plk1 consists of two drug-targeting sites, namely, N-terminal kinase domain (KD) and C-terminal polo-box domain (PBD). As KD-targeting inhibitors are associated with severe side effects, here we report on the pyrazole-based Plk1 PBD inhibitor, KBJK557, which showed a remarkable in vitro anticancer effect by inducing Plk1 delocalization, mitotic arrest, and apoptosis in HeLa cells. Further, in vivo optical imaging analysis and antitumorigenic activities in mouse xenograft models demonstrate that KBJK557 preferentially accumulates in cancer cells and selectively inhibits cancer cell proliferation. Pharmacokinetic profiles and partition coefficients suggest that KBJK557 was exposed in the blood and circulated through the organs with an intermediate level of clearance (t1/2, 7.73 h). The present investigation offers a strategy for specifically targeting cancer using a newly identified small-molecule inhibitor that targets the Plk1 PBD.


Subject(s)
Antineoplastic Agents/therapeutic use , Barbiturates/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Barbiturates/chemical synthesis , Barbiturates/metabolism , Barbiturates/pharmacokinetics , Carbocyanines/chemistry , Cell Cycle Proteins/metabolism , Drug Design , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemistry , G2 Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , Humans , Male , Mice, Inbred BALB C , Mice, Inbred ICR , Molecular Structure , Neoplasms/diagnosis , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
15.
Open Biol ; 10(8): 200102, 2020 08.
Article in English | MEDLINE | ID: mdl-32810424

ABSTRACT

The centrosome, a unique membraneless multiprotein organelle, plays a pivotal role in various cellular processes that are critical for promoting cell proliferation. Faulty assembly or organization of the centrosome results in abnormal cell division, which leads to various human disorders including cancer, microcephaly and ciliopathy. Recent studies have provided new insights into the stepwise self-assembly of two pericentriolar scaffold proteins, Cep63 and Cep152, into a near-micrometre-scale higher-order structure whose architectural properties could be crucial for proper execution of its biological function. The construction of the scaffold architecture appears to be centrally required for tight control of a Ser/Thr kinase called Plk4, a key regulator of centriole duplication, which occurs precisely once per cell cycle. In this review, we will discuss a new paradigm for understanding how pericentrosomal scaffolds are self-organized into a new functional entity and how, on the resulting structural platform, Plk4 undergoes physico-chemical conversion to trigger centriole biogenesis.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Centrioles/metabolism , Animals , Biomarkers , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Eukaryotic Cells/metabolism , Humans , Protein Binding , Protein Transport
16.
Mol Cell Biol ; 40(10)2020 04 28.
Article in English | MEDLINE | ID: mdl-32152252

ABSTRACT

Cep57 has been characterized as a component of a pericentriolar complex containing Cep63 and Cep152. Interestingly, Cep63 and Cep152 self-assemble into a pericentriolar cylindrical architecture, and this event is critical for the orderly recruitment of Plk4, a key regulator of centriole duplication. However, the way in which Cep57 interacts with the Cep63-Cep152 complex and contributes to the structure and function of Cep63-Cep152 self-assembly remains unknown. We demonstrate that Cep57 interacts with Cep63 through N-terminal motifs and associates with Cep152 via Cep63. Three-dimensional structured illumination microscopy (3D-SIM) analyses suggested that the Cep57-Cep63-Cep152 complex is concentrically arranged around a centriole in a Cep57-in and Cep152-out manner. Cep57 mutant cells defective in Cep63 binding exhibited improper Cep63 and Cep152 localization and impaired Sas6 recruitment for procentriole assembly, proving the significance of the Cep57-Cep63 interaction. Intriguingly, Cep63 fused to a microtubule (MT)-binding domain of Cep57 functioned in concert with Cep152 to assemble around stabilized MTs in vitro Thus, Cep57 plays a key role in architecting the Cep63-Cep152 assembly around centriolar MTs and promoting centriole biogenesis. This study may offer a platform to investigate how the organization and function of the pericentriolar architecture are altered by disease-associated mutations found in the Cep57-Cep63-Cep152 complex.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Cell Cycle Proteins/analysis , Cell Line, Tumor , HEK293 Cells , Humans , Microtubule-Associated Proteins/analysis , Nuclear Proteins/analysis , Protein Interaction Domains and Motifs , Protein Interaction Maps
17.
Nat Commun ; 10(1): 4959, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31672968

ABSTRACT

Tight control of centriole duplication is critical for normal chromosome segregation and the maintenance of genomic stability. Polo-like kinase 4 (Plk4) is a key regulator of centriole biogenesis. How Plk4 dynamically promotes its symmetry-breaking relocalization and achieves its procentriole-assembly state remains unknown. Here we show that Plk4 is a unique kinase that utilizes its autophosphorylated noncatalytic cryptic polo-box (CPB) to phase separate and generate a nanoscale spherical condensate. Analyses of the crystal structure of a phospho-mimicking, condensation-proficient CPB mutant reveal that a disordered loop at the CPB PB2-tip region is critically required for Plk4 to generate condensates and induce procentriole assembly. CPB phosphorylation also promotes Plk4's dissociation from the Cep152 tether while binding to downstream STIL, thus allowing Plk4 condensate to serve as an assembling body for centriole biogenesis. This study uncovers the mechanism underlying Plk4 activation and may offer strategies for anti-Plk4 intervention against genomic instability and cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Organelle Biogenesis , Protein Serine-Threonine Kinases/genetics , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Imaging, Three-Dimensional , Microscopy, Confocal , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Sf9 Cells , Spodoptera
18.
Nat Commun ; 10(1): 1151, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858376

ABSTRACT

The cell is constructed by higher-order structures and organelles through complex interactions among distinct structural constituents. The centrosome is a membraneless organelle composed of two microtubule-derived structures called centrioles and an amorphous mass of pericentriolar material. Super-resolution microscopic analyses in various organisms revealed that diverse pericentriolar material proteins are concentrically localized around a centriole in a highly organized manner. However, the molecular nature underlying these organizations remains unknown. Here we show that two human pericentriolar material scaffolds, Cep63 and Cep152, cooperatively generate a heterotetrameric α-helical bundle that functions in conjunction with its neighboring hydrophobic motifs to self-assemble into a higher-order cylindrical architecture capable of recruiting downstream components, including Plk4, a key regulator for centriole duplication. Mutations disrupting the self-assembly abrogate Plk4-mediated centriole duplication. Because pericentriolar material organization is evolutionarily conserved, this work may offer a paradigm for investigating the assembly and function of centrosomal scaffolds in various organisms.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Neoplasm Proteins/metabolism , Protein Multimerization/physiology , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/isolation & purification , Cell Line, Tumor , Crystallography, X-Ray , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Microscopy, Fluorescence , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/isolation & purification , Protein Conformation, alpha-Helical , Protein Serine-Threonine Kinases/isolation & purification , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Time-Lapse Imaging
19.
Respirology ; 23(3): 325-330, 2018 03.
Article in English | MEDLINE | ID: mdl-28960643

ABSTRACT

BACKGROUND AND OBJECTIVE: Cryptogenic organizing pneumonia (COP) is corticosteroid responsive but residual computed tomography (CT) chest changes are often noted. The present study examined clinical and HRCT features of COP in which there was incomplete resolution. METHODS: We studied 93 patients with histopathologically confirmed COP and serial HRCT imaging. Clinical features were assessed, and serial CT images were analysed. Uni- and multivariate analyses were performed to determine clinical or imaging factors related to incomplete resolution on CT. RESULTS: Complete resolution on CT imaging was seen in 21/93 patients (23%) and residual abnormalities were seen in 72/93 patients (77%). In univariate analysis, total lesion (P = 0.036), degree of consolidation (P = 0.011), treatment duration (P < 0.001) and single-breath carbon monoxide diffusing capacity of lung (P = 0.021) were significantly associated with residual imaging abnormalities. In multivariate analysis, extent of consolidation (P = 0.018; odds ratio (OR) = 14.92) and treatment duration (P = 0.011; OR = 1.32) remained as significant factors linked to residual abnormalities. CT images in unresolved COP were akin to fibrotic non-specific interstitial pneumonia (fNSIP) in 53/72 (74%) patients. CONCLUSION: Clinical, radiological and lung diffusion measurements were related to incomplete resolution on CT after COP. Imaging abnormalities on CT chest generally resembled fNSIP.


Subject(s)
Cryptogenic Organizing Pneumonia/diagnosis , Lung/diagnostic imaging , Radiography, Thoracic/methods , Tomography, X-Ray Computed/methods , Adult , Aged , Cryptogenic Organizing Pneumonia/physiopathology , Disease Progression , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Respiratory Function Tests , Severity of Illness Index , Time Factors
20.
J Biol Chem ; 293(3): 847-862, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29191835

ABSTRACT

Elevated expression of human enhancer filamentation 1 (HEF1; also known as NEDD9 or Cas-L) is an essential stimulus for the metastatic process of various solid tumors. This process requires HEF1 localization to focal adhesions (FAs). Although the association of HEF1 with FAs is considered to play a role in cancer cell migration, the mechanism targeting HEF1 to FAs remains unclear. Moreover, up-regulation of Polo-like kinase 1 (Plk1) positively correlates with human cancer metastasis, yet how Plk1 deregulation promotes metastasis remains elusive. Here, we report that casein kinase 1δ (CK1δ) phosphorylates HEF1 at Ser-780 and Thr-804 and that these phosphorylation events promote a physical interaction between Plk1 and HEF1. We found that this interaction is critical for HEF1 translocation to FAs and for inducing migration of HeLa cells. Plk1-docking phosphoepitopes were mapped/confirmed in HEF1 by various methods, including X-ray crystallography, and mutated for functional analysis in HeLa cells. In summary, our results reveal the role of a phosphorylation-dependent HEF1-Plk1 complex in HEF1 translocation to FAs to induce cell migration. Our findings provide critical mechanistic insights into the HEF1-Plk1 complex-dependent localization of HEF1 to FAs underlying the metastatic process and may therefore contribute to the development of new cancer therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Focal Adhesions/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation/genetics , Cell Proliferation/physiology , Focal Adhesions/genetics , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Phosphoproteins/genetics , Phosphorylation/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...