Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(5): 3307-3320, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38105611

ABSTRACT

Polo-like kinase 1 (PLK1), which is crucial in cell cycle regulation, is considered a promising anticancer drug target. Herein, we present the N-degron pathway-based proteolysis targeting chimera (PROTAC) for PLK1 degradation, targeting the Polo-box domain (PBD). We identified DD-2 as the most potent PROTAC that selectively induces PLK1 degradation in cancer cells, including HeLa and nonsmall cell lung cancer (NSCLC), through the N-degron pathway. DD-2 exhibited significant in vitro anticancer effects, inducing G2/M arrest and apoptosis in HeLa and NSCLC cell lines. DD-2 showed significant tumor growth inhibition in a xenograft mouse model using HeLa and NSCLC cell lines, highlighting its potential in cancer treatment. Furthermore, the combination of DD-2 with tyrosine kinase inhibitor (TKI), osimertinib, effectively suppressed tumor growth in double-mutated H1975 cell lines, emphasizing DD-2's potential in combination cancer therapies. Collectively, this study demonstrates the potential of the N-degron pathway, especially using DD-2, for targeted cancer therapies.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins , Proteolysis Targeting Chimera , Protein Serine-Threonine Kinases , Polo-Like Kinase 1 , Apoptosis , Degrons , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , G2 Phase Cell Cycle Checkpoints , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Methods Mol Biol ; 2620: 219-228, 2023.
Article in English | MEDLINE | ID: mdl-37010765

ABSTRACT

Protein arginylation is a unique and under-explored posttranslational modification, which governs many biological functions and the fate of affected proteins. Since ATE1 was discovered in 1963, a central tenet of protein arginylation is that arginylated proteins are destined for proteolysis. However, recent studies have shown that protein arginylation controls not only the half-life of a protein but also various signaling pathways. Here, we introduce a novel molecular tool to elucidate protein arginylation. This new tool, termed R-catcher, is derived from the ZZ domain of p62/sequestosome-1, an N-recognin of the N-degron pathway. The ZZ domain, which has been shown to strongly bind N-terminal arginine, has been modified at specific residues to increase specificity and affinity for N-terminal arginine. R-catcher is a powerful analysis tool allowing researchers to capture the cellular arginylation patterns under various stimuli and conditions, thereby identifying potential therapeutic targets in numerous diseases.


Subject(s)
Aminoacyltransferases , Aminoacyltransferases/chemistry , Protein Processing, Post-Translational , Proteolysis , Proteins/metabolism , Arginine/metabolism
4.
EMBO Rep ; 22(12): e51503, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34585824

ABSTRACT

ß-Catenin is a multifunctional protein and participates in numerous processes required for embryonic development, cell proliferation, and homeostasis through various molecular interactions and signaling pathways. To date, however, there is no direct evidence that ß-catenin contributes to cytokinesis. Here, we identify a novel p-S60 epitope on ß-catenin generated by Plk1 kinase activity, which can be found at the actomyosin contractile ring of early telophase cells and at the midbody of late telophase cells. Depletion of ß-catenin leads to cytokinesis-defective phenotypes, which eventually result in apoptotic cell death. In addition, phosphorylation of ß-catenin Ser60 by Plk1 is essential for the recruitment of Ect2 to the midbody, activation of RhoA, and interaction between ß-catenin, Plk1, and Ect2. Time-lapse image analysis confirmed the importance of ß-catenin phospho-Ser60 in furrow ingression and the completion of cytokinesis. Taken together, we propose that phosphorylation of ß-catenin Ser60 by Plk1 in cooperation with Ect2 is essential for the completion of cytokinesis. These findings may provide fundamental knowledge for the research of cytokinesis failure-derived human diseases.


Subject(s)
Actomyosin , Cytokinesis , Actomyosin/metabolism , Cell Cycle Proteins/metabolism , HeLa Cells , Humans , Phosphorylation , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/metabolism , beta Catenin/metabolism , Polo-Like Kinase 1
5.
Int J Mol Sci ; 22(15)2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34361089

ABSTRACT

UBR box E3 ligases, also called N-recognins, are integral components of the N-degron pathway. Representative N-recognins include UBR1, UBR2, UBR4, and UBR5, and they bind destabilizing N-terminal residues, termed N-degrons. Understanding the molecular bases of their substrate recognition and the biological impact of the clearance of their substrates on cellular signaling pathways can provide valuable insights into the regulation of these pathways. This review provides an overview of the current knowledge of the binding mechanism of UBR box N-recognin/N-degron interactions and their roles in signaling pathways linked to G-protein-coupled receptors, apoptosis, mitochondrial quality control, inflammation, and DNA damage. The targeting of these UBR box N-recognins can provide potential therapies to treat diseases such as cancer and neurodegenerative diseases.


Subject(s)
Apoptosis , DNA Damage , Inflammation/pathology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , Humans , Inflammation/metabolism
6.
Cell Mol Life Sci ; 78(7): 3725-3741, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33687501

ABSTRACT

Protein arginylation is a critical regulator of a variety of biological processes. The ability to uncover the global arginylation pattern and its associated signaling pathways would enable us to identify novel disease targets. Here, we report the development of a tool able to capture the N-terminal arginylome. This tool, termed R-catcher, is based on the ZZ domain of p62, which was previously shown to bind N-terminally arginylated proteins. Mutating the ZZ domain enhanced its binding specificity and affinity for Nt-Arg. R-catcher pulldown coupled to LC-MS/MS led to the identification of 59 known and putative arginylated proteins. Among these were a subgroup of novel ATE1-dependent arginylated ER proteins that are linked to diverse biological pathways, including cellular senescence and vesicle-mediated transport as well as diseases, such as Amyotrophic Lateral Sclerosis and Alzheimer's disease. This study presents the first molecular tool that allows the unbiased identification of arginylated proteins, thereby unlocking the arginylome and provide a new path to disease biomarker discovery.


Subject(s)
Aminoacyltransferases/metabolism , Arginine/metabolism , Endoplasmic Reticulum/metabolism , Genetic Vectors/genetics , Membrane Proteins/metabolism , Protein Processing, Post-Translational , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics , Arginine/chemistry , Arginine/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Substrate Specificity
7.
Cancers (Basel) ; 13(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670717

ABSTRACT

Cancer-associated fibroblasts (CAFs) are important in tumor progression. The autophagy adaptor protein, p62/SQSTM1/Sequestosome-1, is up-regulated in tumors, but down-regulated in CAFs in the early stages of lung adenocarcinoma. We investigated whether p62-induced autophagy might control CAF activation. Under CAF-inducing conditions, like hypoxia or cancer cell co-cultures, p62 ablation or autophagy inhibition with hydroxychloroquine (HCQ) impaired CAF activation and reduced transforming growth factor beta (TGFß) production, which impeded tumor growth. During CAF activation, p62-induced autophagy up-regulated the expression of the anti-oxidant signaling protein, nuclear factor erythroid 2-related factor 2 (Nrf2), and the ER-stress response regulator, activating transcription factor 6 (ATF6). Genetically or pharmacologically inhibiting the Nrf2-ATF6 pathway totally blocked CAF activation and tumor progression. These results demonstrate that p62 is a key modulator of primary lung adenocarcinoma progression. Thus, targeting the p62-Nrf2 autophagy signaling pathway might be a novel, stroma-focused, cancer prevention and/or treatment strategy.

8.
Int J Mol Sci ; 21(13)2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32645923

ABSTRACT

In the past, several microtubule targeting agents (MTAs) have been developed into successful anticancer drugs. However, the usage of these drugs has been limited by the acquisition of drug resistance in many cancers. Therefore, there is a constant demand for the development of new therapeutic drugs. Here we report the discovery of 5-5 (3-cchlorophenyl)-N-(3-pyridinyl)-2-furamide (CPPF), a novel microtubule targeting anticancer agent. Using both 2D and 3D culture systems, we showed that CPPF was able to suppress the proliferation of diverse cancer cell lines. In addition, CPPF was able to inhibit the growth of multidrug-resistant cell lines that are resistant to other MTAs, such as paclitaxel and colchicine. Our results showed that CPPF inhibited growth by depolymerizing microtubules leading to mitotic arrest and apoptosis. We also confirmed CPPF anticancer effects in vivo using both a mouse xenograft and a two-step skin cancer mouse model. Using established zebrafish models, we showed that CPPF has low toxicity in vivo. Overall, our study proves that CPPF has the potential to become a successful anticancer chemotherapeutic drug.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Microtubules/metabolism , Neoplasms/drug therapy , A549 Cells , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Colchicine/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor/methods , HeLa Cells , Hep G2 Cells , Humans , Jurkat Cells , K562 Cells , MCF-7 Cells , Male , Mice , Mitosis/drug effects , Neoplasms/metabolism , PC-3 Cells , Paclitaxel/pharmacology , U937 Cells , Xenograft Model Antitumor Assays/methods , Zebrafish
9.
Cell Rep ; 30(5): 1447-1462.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32023461

ABSTRACT

Primary cilium is an antenna-like microtubule-based cellular sensing structure. Abnormal regulation of the dynamic assembly and disassembly cycle of primary cilia is closely related to ciliopathy and cancer. The Wnt signaling pathway plays a major role in embryonic development and tissue homeostasis, and defects in Wnt signaling are associated with a variety of human diseases, including cancer. In this study, we provide direct evidence of Wnt3a-induced primary ciliogenesis, which includes a continuous pathway showing that the stimulation of Wnt3a, a canonical Wnt ligand, promotes the generation of ß-catenin p-S47 epitope by CK1δ, and these events lead to the reorganization of centriolar satellites resulting in primary ciliogenesis. We have also confirmed the application of our findings in MCF-7/ADR cells, a multidrug-resistant tumor cell model. Thus, our data provide a Wnt3a-induced primary ciliogenesis pathway and may provide a clue on how to overcome multidrug resistance in cancer treatment.


Subject(s)
Centrioles/metabolism , Cilia/metabolism , Organogenesis , Wnt3A Protein/metabolism , beta Catenin/metabolism , Amino Acid Sequence , Animals , Casein Kinases/metabolism , Centrosome/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Epitopes/metabolism , HEK293 Cells , HeLa Cells , Humans , Ligands , MCF-7 Cells , Mice , Phosphorylation , Phosphoserine/metabolism , Wnt3A Protein/chemistry
10.
Bone ; 131: 115153, 2020 02.
Article in English | MEDLINE | ID: mdl-31730830

ABSTRACT

Bone homeostasis is regulated by a balance of bone formation and bone resorption; dysregulation of bone homeostasis may cause bone-related diseases (e.g., osteoporosis, osteopetrosis, bone fracture). Members of the nuclear factor of activated T cells (NFAT) family of transcription factors play crucial roles in the regulation of immune system, inflammatory responses, cardiac formation, skeletal muscle development, and bone homeostasis. Of these, NFATc1 is a key transcription factor mediating osteoclast differentiation, which is regulated by phosphorylation by distinct NFAT kinases including casein kinase 1 (CK1), glycogen synthase kinase 3 (GSK3), and dual-specificity tyrosine-phosphorylation-regulated kinases (DYRKs). In this study, we report that cell division control protein 2 homolog (cdc2) is a novel NFAT protein kinase that inhibits NFATc1 activation by direct phosphorylation of the NFATc1 S263 residue. Cdc2 inhibitors such as Roscovitine and BMI-1026 induce reduction of phosphorylation of NFATc1, and this process leads to the inhibition of NFATc1 translocation from the nucleus to the cytoplasm, consequently increasing the nuclear pool of NFATc1. Additionally, the inhibition of cdc2-mediated NFATc1 phosphorylation causes an elevation of osteoclast differentiation or TRAP-positive staining in zebrafish scales. Our results suggest that cdc2 is a novel NFAT protein kinase that negatively regulates osteoclast differentiation.


Subject(s)
Bone Resorption , Osteoclasts , Animals , Cell Differentiation , Glycogen Synthase Kinase 3 , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Phosphorylation , RANK Ligand , Zebrafish/metabolism
11.
Mol Cell ; 75(5): 1058-1072.e9, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31375263

ABSTRACT

The endoplasmic reticulum (ER) is susceptible to wear-and-tear and proteotoxic stress, necessitating its turnover. Here, we show that the N-degron pathway mediates ER-phagy. This autophagic degradation initiates when the transmembrane E3 ligase TRIM13 (also known as RFP2) is ubiquitinated via the lysine 63 (K63) linkage. K63-ubiquitinated TRIM13 recruits p62 (also known as sequestosome-1), whose complex undergoes oligomerization. The oligomerization is induced when the ZZ domain of p62 is bound by the N-terminal arginine (Nt-Arg) of arginylated substrates. Upon activation by the Nt-Arg, oligomerized TRIM13-p62 complexes are separated along with the ER compartments and targeted to autophagosomes, leading to lysosomal degradation. When protein aggregates accumulate within the ER lumen, degradation-resistant autophagic cargoes are co-segregated by ER membranes for lysosomal degradation. We developed synthetic ligands to the p62 ZZ domain that enhance ER-phagy for ER protein quality control and alleviate ER stresses. Our results elucidate the biochemical mechanisms and pharmaceutical means that regulate ER homeostasis.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Proteolysis , Sequestosome-1 Protein/metabolism , Animals , Carrier Proteins/genetics , Endoplasmic Reticulum/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , Sequestosome-1 Protein/genetics , Ubiquitination
12.
Cell Death Dis ; 10(8): 570, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358734

ABSTRACT

The initiation of centrosome duplication is regulated by the Plk4/STIL/hsSAS-6 axis; however, the involvement of other centrosomal proteins in this process remains unclear. In this study, we demonstrate that Cep131 physically interacts with Plk4 following phosphorylation of residues S21 and T205. Localizing at the centriole, phosphorylated Cep131 has an increased capability to interact with STIL, leading to further activation and stabilization of Plk4 for initiating centrosome duplication. Moreover, we found that Cep131 overexpression resulted in centrosome amplification by excessive recruitment of STIL to the centriole and subsequent stabilization of Plk4, contributing to centrosome amplification. The xenograft mouse model also showed that both centrosome amplification and colon cancer growth were significantly increased by Cep131 overexpression. These findings demonstrate that Cep131 is a novel substrate of Plk4, and that phosphorylation or dysregulated Cep131 overexpression promotes Plk4 stabilization and therefore centrosome amplification, establishing a perspective in understanding a relationship between centrosome amplification and cancer development.


Subject(s)
Cell Cycle Proteins/genetics , Centrosome/metabolism , Colonic Neoplasms/genetics , Cytoskeletal Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Cell Line, Tumor , Colonic Neoplasms/pathology , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , HEK293 Cells , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Phosphorylation/genetics
13.
BMB Rep ; 52(5): 342-347, 2019 May.
Article in English | MEDLINE | ID: mdl-31068247

ABSTRACT

Methylation is a primary epigenetic mechanism regulating gene expression. 5-aza-2'-deoxycytidine is an FDA-approved drug prescribed for treatment of cancer by inhibiting DNA-Methyl-Transferase 1 (DNMT1). Results of this study suggest that prolonged treatment with 5-aza-2'-deoxycytidine could induce centrosome abnormalities in cancer cells and that CEP131, a centrosome protein, is regulated by DNMT1. Interestingly, cancer cell growth was attenuated in vitro and in vivo by inhibiting the expression of Cep131. Finally, Cep131-deficient cells were more sensitive to treatment with DNMT1 inhibitors. These findings suggest that Cep131 is a potential novel anti-cancer target. Agents that can inhibit this protein may be useful alone or in combination with DNMT1 inhibitors to treat cancer. [BMB Reports 2019; 52(5): 342-347].


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , Decitabine/pharmacology , Microtubule Proteins/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cytoskeletal Proteins , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/drug effects , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Epigenesis, Genetic , Female , HEK293 Cells , HeLa Cells , Humans , Microtubule Proteins/genetics , Microtubule Proteins/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
14.
Exp Mol Med ; 51(2): 1-14, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755586

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) mediates tumor cell adaptation to hypoxic conditions and is a potentially important anticancer therapeutic target. We previously developed a method for synthesizing a benzofuran-based natural product, (R)-(-)-moracin-O, and obtained a novel potent analog, MO-460 that suppresses the accumulation of HIF-1α in Hep3B cells. However, the molecular target and underlying mechanism of action of MO-460 remained unclear. In the current study, we identified heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) as a molecular target of MO-460. MO-460 inhibits the initiation of HIF-1α translation by binding to the C-terminal glycine-rich domain of hnRNPA2B1 and inhibiting its subsequent binding to the 3'-untranslated region of HIF-1α mRNA. Moreover, MO-460 suppresses HIF-1α protein synthesis under hypoxic conditions and induces the accumulation of stress granules. The data provided here suggest that hnRNPA2B1 serves as a crucial molecular target in hypoxia-induced tumor survival and thus offer an avenue for the development of novel anticancer therapies.


Subject(s)
Benzofurans/pharmacology , Biological Products/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , 3' Untranslated Regions , Benzofurans/chemistry , Biological Products/chemistry , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Structure , Protein Binding , Protein Biosynthesis/drug effects , Protein Interaction Domains and Motifs , Stress, Physiological/drug effects , Transcription, Genetic
16.
Exp Dermatol ; 27(11): 1304-1308, 2018 11.
Article in English | MEDLINE | ID: mdl-30092122

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin disease whose prevalence is increasing worldwide. Filaggrin (FLG) is essential for the development of the skin barrier, and its genetic mutations are major predisposing factors for AD. In this study, we developed a convenient and practical method to detect FLG mutations in AD patients using peptide nucleic acid (PNA) probes labelled with fluorescent markers for rapid analysis. Fluorescence melting curve analysis (FMCA) precisely identified FLG mutations based on the distinct difference in the melting temperatures of the wild-type and mutant allele. Moreover, PNA probe-based FMCA easily and accurately verified patient samples with both heterozygote and homozygote FLG mutations, providing a high-throughput method to reliable screen AD patients. Our method provides a convenient, rapid and accurate diagnostic tool to identify potential AD patients allowing for early preventive treatment, leading to lower incidence rates of AD, and reducing total healthcare expenses.


Subject(s)
DNA Mutational Analysis/methods , DNA Probes , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/genetics , Intermediate Filament Proteins/genetics , Alleles , Case-Control Studies , Filaggrin Proteins , Fluorescence , Heterozygote , High-Throughput Nucleotide Sequencing/methods , Homozygote , Humans , Mutation , Peptide Nucleic Acids/genetics , Transition Temperature
17.
Exp Dermatol ; 27(3): 285-288, 2018 03.
Article in English | MEDLINE | ID: mdl-29392819

ABSTRACT

Skin cancer is the most common type of cancer. The incidence rate of skin cancer has continuously increased over the past decades. In an effort to discover novel anticancer agents, we identified a novel tubulin inhibitor STK899704, which is structurally distinct from other microtubule-binding agents such as colchicine, vinca alkaloids and taxanes. STK899704 inhibited microtubule polymerization leading to mitotic arrest and suppressed the proliferation of various cancer cell lines as well as multidrug resistance cancer cell lines. In this study, our investigation is further extended into animal model to evaluate the effect of STK899704 on skin carcinogenesis in vivo. Surprisingly, almost 80% of the tumors treated with STK899704 were regressed with a one-fifth reduction in tumor volume. Furthermore, the efficacy of STK899704 was nearly 2 times higher than that of 5-fluorouracil, a widely used skin cancer therapeutic. Overall, our results suggest that STK899704 is a promising anticancer chemotherapeutic that may replace existing therapies, particularly for skin cancer.


Subject(s)
Benzofurans/therapeutic use , Skin Neoplasms/drug therapy , Tubulin Modulators/therapeutic use , 9,10-Dimethyl-1,2-benzanthracene , Animals , Antimetabolites, Antineoplastic/therapeutic use , Carcinogenesis , Colchicine/therapeutic use , Disease Models, Animal , Fluorouracil/therapeutic use , Male , Mice , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate , Tubulin/metabolism
18.
Sci Signal ; 11(511)2018 01 02.
Article in English | MEDLINE | ID: mdl-29295953

ABSTRACT

BiP and other endoplasmic reticulum (ER)-resident proteins are thought to be metabolically stable and to function primarily in the ER lumen. We sought to assess how the abundance of these proteins dynamically fluctuates in response to various stresses and how their subpopulations are relocated to non-ER compartments such as the cytosol. We showed that the molecular chaperone BiP (also known as GRP78) was short-lived under basal conditions and ER stress. The turnover of BiP was in part driven by its amino-terminal arginylation (Nt-arginylation) by the arginyltransferase ATE1, which generated an autophagic N-degron of the N-end rule pathway. ER stress elicited the formation of R-BiP, an effect that was increased when the proteasome was also inhibited. Nt-arginylation correlated with the cytosolic relocalization of BiP under the types of stress tested. The cytosolic relocalization of BiP did not require the functionality of the unfolded protein response or the Sec61- or Derlin1-containing translocon. A key inhibitor of the turnover and Nt-arginylation of BiP was HERP (homocysteine-responsive ER protein), a 43-kDa ER membrane-integrated protein that is an essential component of ER-associated protein degradation. Pharmacological inhibition of the ER-Golgi secretory pathway also suppressed R-BiP formation. Finally, we showed that cytosolic R-BiP induced by ER stress and proteasomal inhibition was routed to autophagic vacuoles and possibly additional metabolic fates. These results suggest that Nt-arginylation is a posttranslational modification that modulates the function, localization, and metabolic fate of ER-resident proteins.


Subject(s)
Aminoacyltransferases/metabolism , Arginine/metabolism , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Aminoacyltransferases/genetics , Autophagy/drug effects , Cytosol/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , HEK293 Cells , HeLa Cells , Heat-Shock Proteins/genetics , Humans , Hydrogen Peroxide/pharmacology , Leupeptins/pharmacology , Membrane Proteins/genetics , PC-3 Cells , Proteasome Endopeptidase Complex/metabolism
19.
Autophagy ; 14(2): 359-361, 2018.
Article in English | MEDLINE | ID: mdl-29261001

ABSTRACT

In macroautophagy/autophagy, cargoes are collected by specific receptors, such as SQSTM1/p62 (sequestosome 1), and delivered to phagophores for lysosomal degradation. To date, little is known about how cells modulate SQSTM1 activity and autophagosome biogenesis in response to accumulating cargoes. In this study, we show that SQSTM1 is an N-recognin whose ZZ domain binds N-terminal arginine (Nt-Arg) and other N-degrons (Nt-Lys, Nt-His, Nt-Trp, Nt-Phe, and Nt-Tyr) of the N-end rule pathway. The substrates of SQSTM1 include the endoplasmic reticulum (ER)-residing chaperone HSPA5/GRP78/BiP. Upon N-end rule interaction with the Nt-Arg of arginylated HSPA5 (R-HSPA5), SQSTM1 undergoes self-polymerization via disulfide bonds of Cys residues including Cys113, facilitating cargo collection. In parallel, Nt-Arg-bound SQSTM1 acts as an inducer of autophagosome biogenesis and autophagic flux. Through this dual regulatory mechanism, SQSTM1 plays a key role in the crosstalk between the ubiquitin (Ub)-proteasome system (UPS) and autophagy. Based on these results, we employed 3D-modeling of SQSTM1 and a virtual chemical library to develop small molecule ligands to the ZZ domain of SQSTM1. These autophagy inducers accelerated the autophagic removal of mutant HTT (huntingtin) aggregates. We suggest that SQSTM1 can be exploited as a novel drug target to modulate autophagic processes in pathophysiological conditions.


Subject(s)
Autophagy , Heat-Shock Proteins/metabolism , Huntingtin Protein/metabolism , Proteolysis , Sequestosome-1 Protein/metabolism , Ubiquitin/metabolism , Animals , Arginine/metabolism , Autophagosomes/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Humans , Lysosomes/metabolism , Polymerization , Protein Binding , Protein Domains , Signal Transduction
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
...