Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Gene ; 840: 146772, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35905845

ABSTRACT

The expression of ULK1, a core protein of autophagy, is closely related to autophagic activity. Numerous studies have shown that pathological abnormal expression of ULK1 is associated with various human diseases such as neurological disorders, infections, cardiovascular diseases, liver diseases and cancers. In addition, new advances in the regulation of ULK1 have been identified. Furthermore, targeting ULK1 as a therapeutic strategy for diseases is gaining attention as new corresponding activators or inhibitors are being developed. In this review, we describe the structure and regulation of ULK1 as well as the current targeted activators and inhibitors. Moreover, we highlight the pathological disorders of ULK1 expression and its critical role in human diseases.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Neoplasms/genetics
2.
Molecules ; 27(9)2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35565977

ABSTRACT

There is an urgent need to develop new effective therapies for HCC. Our previous study identified ULK1 as the potential target for HCC therapy and screened the compound XST-14 as a specific inhibitor of ULK1 to suppress HCC progression. However, the poor manufacturability of XST-14 impeded the process of its clinical translation. In this study, we first generated pharmacophore models of ULK1 based on the X-ray structure of UKL1 in complex with ligands. We then screened the Specs chemical library for potential UKL1 inhibitors. By molecular docking, we screened out the 19 compounds through structure-based virtual screening. Through CCK8 activity screening on HCC cells, we found that ZZY-19 displayed obvious cell killing effects on HCC cells. SPR assay indicated that ZZY-19 had a higher binding affinity for ULK1 than XST-14. Moreover, ZZY-19 induced the effects of anti-proliferation, anti-invasion and anti-migration in HCC cells. Mechanistically, ZZY-19 induces autophagy inhibition by reducing the expression of ULK1 on HCC cells. Especially, the combination of ZZY-19 with sorafenib synergistically suppresses the progression of HCC in vivo. Taken together, ZZY-19 was a potential candidate compound that targeted ULK1 and possessed promising anti-HCC activities by inhibiting autophagy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Autophagy , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins/pharmacology , Liver Neoplasms/drug therapy , Molecular Docking Simulation
3.
Sci Rep ; 10(1): 14874, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913252

ABSTRACT

Autophagy is a key clearance process to recycle damaged cellular components. One important upstream regulator of autophagy is ULK1 kinase. Several three-dimensional structures of the ULK1 catalytic domain are available, but a comprehensive study, including molecular dynamics, is missing. Also, an exhaustive description of ULK1 alterations found in cancer samples is presently lacking. We here applied a framework which links -omics data to structural protein ensembles to study ULK1 alterations from genomics data available for more than 30 cancer types. We predicted the effects of mutations on ULK1 function and structural stability, accounting for protein dynamics, and the different layers of changes that a mutation can induce in a protein at the functional and structural level. ULK1 is down-regulated in gynecological tumors. In other cancer types, ULK2 could compensate for ULK1 downregulation and, in the majority of the cases, no marked changes in expression have been found. 36 missense mutations of ULK1, not limited to the catalytic domain, are co-occurring with mutations in a large number of ULK1 interactors or substrates, suggesting a pronounced effect of the upstream steps of autophagy in many cancer types. Moreover, our results pinpoint that more than 50% of the mutations in the kinase domain of ULK1, here investigated, are predicted to affect protein stability. Three mutations (S184F, D102N, and A28V) are predicted with only impact on kinase activity, either modifying the functional dynamics or the capability to exert effects from distal sites to the functional and catalytic regions. The framework here applied could be extended to other protein targets to aid the classification of missense mutations from cancer genomics studies, as well as to prioritize variants for experimental validation, or to select the appropriate biological readouts for experiments.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/enzymology , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Molecular Dynamics Simulation , Mutation , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Protein Structural Elements , Signal Transduction , Structure-Activity Relationship
4.
Eur J Med Chem ; 208: 112782, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32961380

ABSTRACT

UNC51-like kinase1 (ULK1) recruits its binding partners and initiates the autophagy process in cancer. ULK1 is significantly overexpressed in Non-small cell lung cancer (NSCLC) and negatively correlated with clinical prognosis in NSCLC patients. Based upon the binding features of ULK1, we explored the pharmacophore modeling to discover the common anchoring features. It was verified by synthesizing 5-bromo-4-phenoxy-N-phenylpyrimidin-2-amine derivatives, as well as subsequently elucidating the structure-activity relationships (SAR). Among all the obtained ULK1 inhibitors, 5-bromo-4-(2-fluoro-4-nitrophenoxy)-N-(3,4,5-trimethoxyphenyl) pyrimidin-2-amine (3s), was the most active one. The docking analysis was conducted to compare 3s and SBI-0206965, which further elucidated the roles of the H-bond donor. This compound inhibited the proliferation of A549 cells and showed strong inhibitory activity against ULK1 kinase. Moreover, we found that compound 3s could induce apoptosis while simultaneously blocking autophagy. Collectively, these findings shed new light on compound 3s that would be utilized as a promising candidate drug for the future NSCLC therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pyrimidines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Binding , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Structure-Activity Relationship
5.
Structure ; 28(11): 1184-1196.e6, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32814032

ABSTRACT

Unc-51-like kinase 4 (ULK4) is a pseudokinase that has been linked to the development of several diseases. Even though sequence motifs required for ATP binding in kinases are lacking, ULK4 still tightly binds ATP and the presence of the co-factor is required for structural stability of ULK4. Here, we present a high-resolution structure of a ULK4-ATPγS complex revealing a highly unusual ATP binding mode in which the lack of the canonical VAIK motif lysine is compensated by K39, located N-terminal to αC. Evolutionary analysis suggests that degradation of active site motifs in metazoan ULK4 has co-occurred with an ULK4-specific activation loop, which stabilizes the C helix. In addition, cellular interaction studies using BioID and biochemical validation data revealed high confidence interactors of the pseudokinase and armadillo repeat domains. Many of the identified ULK4 interaction partners were centrosomal and tubulin-associated proteins and several active kinases suggesting interesting regulatory roles for ULK4.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/analogs & derivatives , Autophagy-Related Protein-1 Homolog/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Magnesium/chemistry , Protein Serine-Threonine Kinases/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Arabidopsis/chemistry , Arabidopsis/enzymology , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Binding Sites , Cations, Divalent , Crystallography, X-Ray , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Magnesium/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Substrate Specificity , Trypanosoma/chemistry , Trypanosoma/enzymology
6.
Sci Rep ; 10(1): 10940, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616830

ABSTRACT

Macroautophagy, the degradation of cytoplasmic content by lysosomal fusion, is an evolutionary conserved process promoting homeostasis and intracellular defence. Macroautophagy is initiated primarily by a complex containing ULK1 or ULK2 (two paralogs of the yeast Atg1 protein). To understand the differences between ULK1 and ULK2, we compared the human ULK1 and ULK2 proteins and their regulation. Despite the similarity in their enzymatic domain, we found that ULK1 and ULK2 have major differences in their autophagy-related interactors and their post-translational and transcriptional regulators. We identified 18 ULK1-specific and 7 ULK2-specific protein motifs serving as different interaction interfaces. We found that interactors of ULK1 and ULK2 all have different tissue-specific expressions partially contributing to diverse and ULK-specific interaction networks in various tissues. We identified three ULK1-specific and one ULK2-specific transcription factor binding sites, and eight sites shared by the regulatory region of both genes. Importantly, we found that both their post-translational and transcriptional regulators are involved in distinct biological processes-suggesting separate functions for ULK1 and ULK2. Unravelling differences between ULK1 and ULK2 could lead to a better understanding of how ULK-type specific dysregulation affects autophagy and other cellular processes that have been implicated in diseases such as inflammatory bowel disease and cancer.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/metabolism , Autophagy , Computational Biology/methods , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Proteins/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Lysosomes , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Serine-Threonine Kinases/chemistry
7.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32516362

ABSTRACT

The autophagy-initiating human ULK complex consists of the kinase ULK1/2, FIP200, ATG13, and ATG101. Hydrogen-deuterium exchange mass spectrometry was used to map their mutual interactions. The N-terminal 640 residues (NTD) of FIP200 interact with the C-terminal IDR of ATG13. Mutations in these regions abolish their interaction. Negative stain EM and multiangle light scattering showed that FIP200 is a dimer, while a single molecule each of the other subunits is present. The FIP200NTD is flexible in the absence of ATG13, but in its presence adopts the shape of the letter C ∼20 nm across. The ULK1 EAT domain interacts loosely with the NTD dimer, while the ATG13:ATG101 HORMA dimer does not contact the NTD. Cryo-EM of the NTD dimer revealed a structural similarity to the scaffold domain of TBK1, suggesting an evolutionary similarity between the autophagy-initiating TBK1 kinase and the ULK1 kinase complex.


Subject(s)
Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Proteins/chemistry , Autophagy/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Vesicular Transport Proteins/chemistry , Amino Acid Sequence , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Binding Sites , Cloning, Molecular , Cryoelectron Microscopy , Deuterium Exchange Measurement , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
8.
Cell Death Dis ; 10(8): 583, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31378785

ABSTRACT

ULK1, the upper-most protein of the ULK1 complex, is emerging as a crucial node in autophagy induction. However, the regulation of ULK1 is not fully understood. In this study, we identified TOPK (T-LAK cell-originated protein kinase), an oncokinase, as a novel upstream kinase to phosphorylate ULK1. We found that TOPK could directly bind with and phosphorylate ULK1 at Ser469, Ser495, and Ser533. The phosphorylation of ULK1 at Ser469, Ser495, and Ser533 by TOPK decreased the activity and stability of ULK1. In addition, we want to examine the initiation of autophagy because the reduction activity of ULK1 reduces the occurrence of autophagy. We demonstrated that TOPK could inhibit the initiation and progression of autophagy in glioma cells. Furthermore, TOPK inhibition increased the sensitivity of glioma cells to temozolomide (TMZ). This discovery provides insight into the problem of TMZ-resistance in GBM treatment.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Temozolomide/therapeutic use , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Cell Line, Tumor , Glioblastoma/pathology , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Phosphorylation/genetics , Protein Domains , Protein Stability , Transfection
9.
J Recept Signal Transduct Res ; 39(2): 122-133, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31311432

ABSTRACT

Autophagy is a self-catabolic mechanism employed by cancer cells to acquire nutrients and energy in times of stress conditions, thereby leading to its progression and survival. Thus, autophagy inhibition has emerged as a new paradigm in the area of cancer treatment. Here, we leverage multi-dimensional screening campaigns aim to identify potent inhibitors against an early and an essential autophagic kinase, ULK1 from DrugBank database. In particular, receptor-based hypothesis, pharmacophore hypothesis, e-pharmacophore hypothesis and shape similarity-based screening algorithm were employed. Of note, the results of the different algorithm were then integrated to eliminate the false positive prediction. Moreover, the inhibitory activities and PK/PD parameters of the leads were tested by Glide and Qikprop algorithm. This resulted in a set of four hits namely; DB12686, DB08341, DB07936, and DB07163. Finally, molecular dynamics simulation was performed using the GROMACS package, to validate the binding kinetics of the hit compound. The compound activity in vitro was assessed by PASS algorithm, highlights the anti-cancer activities of the hits. The structural insights reveal existence of functional moieties such as piperidine carboxamide, benzenesulfonamide, benzamide, and isoindolone in the resultant hits which plays a major role in the anti-cancer activity. Overall, we strongly believe that these ULK1 antagonists could be novel and potent drug candidates for future cancer therapeutics.


Subject(s)
Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Algorithms , Autophagy-Related Protein-1 Homolog/chemistry , Computer Simulation , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Neoplasms/chemistry , Neoplasms/pathology , Quantitative Structure-Activity Relationship
10.
Autophagy ; 15(12): 2076-2090, 2019 12.
Article in English | MEDLINE | ID: mdl-30929559

ABSTRACT

Imatinib is the first molecularly targeted compound for chronic myeloid leukemia (CML) capable to inhibit BCR-ABL kinase activity. However, recent clinical evidence indicates that a substantial proportion of CML patients exhibit BCR-ABL-dependent or independent resistance to imatinib. Despite the importance of imatinib resistance in CML, the underlying molecular mechanisms of this resistance are largely unknown. Here, we identified GCA (grancalcin) as a critical regulator of imatinib resistance in chronic phase CML via activation of autophagy. Mechanistically, we demonstrated that GCA activates TRAF6 ubiquitin ligase activity to induce Lys63 ubiquitination of ULK1, a crucial regulator of autophagy, resulting in its stabilization and activation. We also highlighted the role of GCA-TRAF6-ULK1 autophagy regulatory axis in imatinib resistance. Our findings represent the basis for novel therapeutic strategies against CML.Abbreviation: ACTB/ß-actin: actin beta; ADM: adrenomedullin; AMBRA1: autophagy and beclin 1 regulator 1; AMPK: AMP-activated protein kinase; ANXA5: annexin A5; CP: cytogenetic response; CML: chronic myeloid leukemia; CUL3: cullin 3; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GCA: grancalcin; Dx: at diagnosis; E-64-d: (2S,3S)-trans-Epoxysuccinyl-L-leucylamido-3-methylbutane ethyl ester; IMres: Imatinib resistance; KLHL20: Kelch-like protein 20; LRMP: lymphoid-restricted membrane protein; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MMR: major molecular response; NH4Cl: ammonium chloride; PBMCs: peripheral blood mononuclear cells; PTPRC: protein tyrosine phosphatase, receptor type, C; SQSTM1/p62: sequestosome 1; SYK: spleen associated tyrosine kinase; TAP1: transporter 1, ATP binding cassette subfamily B member; TKIs: ABL-specific tyrosine kinase inhibitors; TLR9: toll- like receptor 9; TRAF6: TNF receptor associated factor 6; ULK1: unc-51 like autophagy activating kinase 1.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy/genetics , Calcium-Binding Proteins/metabolism , Imatinib Mesylate/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Calcium-Binding Proteins/genetics , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/drug effects , Female , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/genetics , Transplantation, Heterologous , Ubiquitination/drug effects , Ubiquitination/genetics
11.
Biochem J ; 476(5): 875-887, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30782972

ABSTRACT

Autophagy is essential for cellular homeostasis and when deregulated this survival mechanism has been associated with disease development. Inhibition of autophagy initiation by inhibiting the kinase ULK1 (Unc-51-like autophagy activating kinase 1) has been proposed as a potential cancer therapy. While inhibitors and crystal structures of ULK1 have been reported, little is known about the other closely related kinase ULK2 (Unc-51-like autophagy activating kinase 2). Here, we present the crystal structure of ULK2 in complex with ATP competitive inhibitors. Surprisingly, the ULK2 structure revealed a dimeric assembly reminiscent of dimeric arrangements of auto-activating kinases suggesting a role for this association in ULK activation. Screening of a kinase focused library of pre-clinical and clinical compounds revealed several potent ULK1/2 inhibitors and good correlation of inhibitor-binding behavior with both ULK kinases. Aurora A was identified as a major off-target of currently used ULK1 inhibitors. Autophagic flux assays demonstrated that this off-target activity by strongly inducing autophagy in different cellular systems conferred an additional layer of complexity in the interpretation of cellular data. The data presented here provide structural models and chemical starting points for the development of ULK1/2 dual inhibitors with improved selectivity for future exploitation of autophagy inhibition.


Subject(s)
Autophagic Cell Death/drug effects , Autophagy-Related Protein-1 Homolog , Intracellular Signaling Peptides and Proteins , Models, Molecular , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Aurora Kinase A/metabolism , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism
12.
Autophagy ; 15(7): 1150-1162, 2019 07.
Article in English | MEDLINE | ID: mdl-30724690

ABSTRACT

Understanding of the mechanism for myeloid differentiation provides important insights into the hematopoietic developmental processes. By using an ESC-derived myeloid progenitor cell model, we found that CSF2/GM-CSF triggered macrophage differentiation and activation of the MTOR signaling pathway. Activation or inhibition of the MTOR signaling enhanced or attenuated macrophage differentiation, respectively, suggesting a critical function. We further showed that macroautophagy/autophagy was inhibited with the addition of CSF2. Furthermore, pharmacological inhibition and genetic modification of autophagy enhanced macrophage differentiation and rescued the inhibitory effect on differentiation caused by MTOR inhibition. Thus, the MTOR signaling pathway regulates macrophage differentiation of myeloid progenitors by inhibiting autophagy. Our results provide new insights into the mechanisms for myeloid differentiation and may prove useful for therapeutic applications of hematopoietic and myeloid progenitor cells. Abbreviations: 2-DG: 2-deoxy-D-glucose; ADGRE1/F4/80: adhesion G protein-coupled receptor E1; BM: bone marrow; CQ: chloroquine; ECAR: extracellular acidification rate; ESC: embryonic stem cell; CSF2/GM-CSF: colony stimulating factor 2; CSF3/G-CSF: colony stimulating factor 3; HPC: hematopoietic progenitor cell; ITGAM/CD11b: integrin alpha M; LPS: lipopolysaccharide; MFI: median fluorescence intensity; MTOR: mechanistic target of rapamycin kinase; RPS6KB1/p70S6K1: ribosomal protein S6 kinase, polypeptide 1; shRNA: short hairpin RNA; SQSTM1/p62: sequestosome 1.


Subject(s)
Autophagy , Cell Differentiation , Macrophages/metabolism , Myeloid Progenitor Cells/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Phosphorylation , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
13.
Autophagy ; 15(4): 613-630, 2019 04.
Article in English | MEDLINE | ID: mdl-30335599

ABSTRACT

ULK1 (unc-51 like autophagy activating kinase 1) is a core component at multiple steps of canonical macroautophagy/autophagy. The activity of ULK1 is tightly regulated by several post-translational modifications, including ubiquitination, yet the deubiquitinase (DUB) responsible for its reversible deubiquitination has not been described. Here, we identified USP1 (ubiquitin specific peptidase 1) as a key player in the modulation of ULK1 K63-linked deubiquitination. Moreover, both USP1 depletion and its chemical inhibition by pimozide are coupled to a reduction of ULK1 in Triton X-100 soluble cellular lysates, and its compartmentalization to a fraction that can be solubilized in 5 M urea. In USP1-depleted cells this fraction is also enriched in SQSTM1 (sequestosome 1), the aggresome marker HDAC6 (histone deacetylase 6), and the prototype of USP1 targets FANCD2 (FA complementation group D2). Consistently, in USP1-depleted and pimozide-treated cells, ULK1 forms protein aggregates enriched in SQSTM1, as detected by both immummunofluorescence and co-immunoprecipitation studies. Notably, depletion of USP1 inhibits canonical autophagic flux and promotes an alternative route leading to lysosomal-mediated degradation of SQSTM1. Our findings reveal a novel function of the USP1-ULK1 axis as a modulator of the switch between canonical and unconventional autophagy. Further, we provide the first evidence supporting the existence of a subset of breast tumors co-expressing ULK1 and MAP1LC3B (microtubule associated protein 1 light chain 3 beta) proteins. Because the USP1 inhibitor pimozide affects breast cancer cell growth, targeting USP1 in those tumors relying on autophagy for growth might prove to be a convenient therapeutic strategy. Abbreviations: ATG13: autophagy related 13; BECN1: beclin 1; BZ: bortezomib; CAPN1: calpain 1; DUB: deubiquitinase; FANCI: FA complementation group I; FANCD2: FA complementation group D2; FZR1: fizzy and cell division cycle 20 related 1; HDAC6: histone deacetylase 6; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; PMZ: pimozide; SH3GLB1: SH3 domain containing GRB2 like, endophilin B1; SQSTM1: sequestosome 1; TRAF6: TNF receptor associated factor 6; ULK1: unc-51 like autophagy activating kinase 1; USP1: ubiquitin specific peptidase 1; WDR48: WD repeat domain 48.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Breast Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Beclin-1/genetics , Beclin-1/metabolism , Breast Neoplasms/genetics , Cell Compartmentation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Microtubule-Associated Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination/drug effects , Ubiquitination/genetics
14.
Autophagy ; 14(12): 2104-2116, 2018.
Article in English | MEDLINE | ID: mdl-30081750

ABSTRACT

The initiation of macroautophagy/autophagy is tightly regulated by the upstream ULK1 kinase complex, which affects many downstream factors including the PtdIns3K complex. The phosphorylation of the right position at the right time on downstream molecules is governed by proper complex formation. One component of the ULK1 complex, ATG101, known as an accessory protein, is a stabilizer of ATG13 in cells. The WF finger region of ATG101 plays an important role in the recruitment of WIPI1 (WD repeat domain, phosphoinositide interacting protein 1) and ZFYVE1 (zinc finger FYVE-type containing 1). Here, we report that the C-terminal region identified in the structure of the human ATG101-ATG13HORMA complex is responsible for the binding of the PtdIns3K complex. This region adopts a ß-strand conformation in free ATG101, but either an α-helix or random coil in our ATG101-ATG13HORMA complex, which protrudes from the core and interacts with other molecules. The C-terminal deletion of ATG101 shows a significant defect in the interaction with PtdIns3K components and subsequently impairs autophagosome formation. This result clearly presents an additional role of ATG101 for bridging the ULK1 and PtdIns3K complexes in the mammalian autophagy process. Abbreviations: ATG: autophagy related; BECN1: beclin 1; GFP: green fluorescent protein; HORMA: Hop1p/Rev7p/MAD2; HsATG13HORMA: HORMA domain of ATG13 from Homo sapiens; KO: knockout; MAD2: mitotic arrest deficient 2 like 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PIK3R4/VPS15: phosphoinositide-3-kinase regulatory subunit 4; PtdIns3K: phosphatidylinositol 3-kinase; RB1CC1/FIP200: RB1 inducible coiled-coil 1; SAXS: small-angle X-ray scattering; ScAtg13HORMA: HORMA domain of Atg13 from Sccharomyces cerevisiae; SEC-SAXS: size-exclusion chromatography with small-angle X-ray scattering; SpAtg13HORMA: HORMA domain of Atg13 from Schizosaccharomyces pombe; SQSTM1/p62: sequestosome 1; ULK1: unc51-like autophagy activating kinase 1; UVRAG: UV radiation resistance associated; WIPI1: WD repeat domain: phosphoinositide interacting 1; ZFYVE1/DFCP1: zinc finger FYVE-type containing 1.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/metabolism , Autophagy/physiology , Class III Phosphatidylinositol 3-Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Vacuolar Sorting Protein VPS15/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Autophagy-Related Protein-1 Homolog/chemistry , Class III Phosphatidylinositol 3-Kinases/chemistry , Crystallography, X-Ray , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Male , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Interaction Domains and Motifs/physiology , Scattering, Small Angle , Tumor Cells, Cultured , X-Ray Diffraction
15.
J Med Chem ; 61(15): 6491-6500, 2018 Aug 09.
Article in English | MEDLINE | ID: mdl-29509411

ABSTRACT

UNC-51-like kinase 1 (ULK1), known as an ortholog of the yeast Atg1, is the serine-threonine kinase and the autophagic initiator in mammals. Accumulating evidence has recently revealed the kinase domain structure of ULK1 and its post-translational modifications, as well as further elucidated its regulatory autophagic pathways and associations with diverse human diseases. Interestingly, a series of small molecules have been recently reported to target ULK1 or ULK1-modulating autophagy, which may provide a clue on exploiting them as novel candidate drugs. Taken together, this review discusses how ULK1 acts as an autophagic initiator for modulation of its intricate mechanisms, as well as how ULK1 becomes a multifunctional target for potential therapeutic applications.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Molecular Targeted Therapy/methods , Animals , Autophagy/drug effects , Autophagy-Related Protein-1 Homolog/chemistry , Humans , Protein Processing, Post-Translational/drug effects
16.
Nat Commun ; 8(1): 548, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28916822

ABSTRACT

Mitochondrial health is critical for skeletal muscle function and is improved by exercise training through both mitochondrial biogenesis and removal of damaged/dysfunctional mitochondria via mitophagy. The mechanisms underlying exercise-induced mitophagy have not been fully elucidated. Here, we show that acute treadmill running in mice causes mitochondrial oxidative stress at 3-12 h and mitophagy at 6 h post-exercise in skeletal muscle. These changes were monitored using a novel fluorescent reporter gene, pMitoTimer, that allows assessment of mitochondrial oxidative stress and mitophagy in vivo, and were preceded by increased phosphorylation of AMP activated protein kinase (Ampk) at tyrosine 172 and of unc-51 like autophagy activating kinase 1 (Ulk1) at serine 555. Using mice expressing dominant negative and constitutively active Ampk in skeletal muscle, we demonstrate that Ulk1 activation is dependent on Ampk. Furthermore, exercise-induced metabolic adaptation requires Ulk1. These findings provide direct evidence of exercise-induced mitophagy and demonstrate the importance of Ampk-Ulk1 signaling in skeletal muscle.Exercise is associated with biogenesis and removal of dysfunctional mitochondria. Here the authors use a mitochondrial reporter gene to demonstrate the occurrence of mitophagy following exercise in mice, and show this is dependent on AMPK and ULK1 signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Exercise , Lysosomes/enzymology , Mitochondria/enzymology , Mitophagy , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Amino Acid Motifs , Animals , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Humans , Lysosomes/genetics , Male , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Oxidative Stress , Phosphorylation
17.
Nat Commun ; 8: 15637, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28561066

ABSTRACT

Autophagy is controlled by AMPK and mTOR, both of which associate with ULK1 and control the production of phosphatidylinositol 3-phosphate (PtdIns3P), a prerequisite for autophagosome formation. Here we report that WIPI3 and WIPI4 scaffold the signal control of autophagy upstream of PtdIns3P production and have a role in the PtdIns3P effector function of WIPI1-WIPI2 at nascent autophagosomes. In response to LKB1-mediated AMPK stimulation, WIPI4-ATG2 is released from a WIPI4-ATG2/AMPK-ULK1 complex and translocates to nascent autophagosomes, controlling their size, to which WIPI3, in complex with FIP200, also contributes. Upstream, WIPI3 associates with AMPK-activated TSC complex at lysosomes, regulating mTOR. Our WIPI interactome analysis reveals the scaffold functions of WIPI proteins interconnecting autophagy signal control and autophagosome formation. Our functional kinase screen uncovers a novel regulatory link between LKB1-mediated AMPK stimulation that produces a direct signal via WIPI4, and we show that the AMPK-related kinases NUAK2 and BRSK2 regulate autophagy through WIPI4.


Subject(s)
Autophagy , Carrier Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Signal Transduction , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Proteins/chemistry , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Lysosomes/metabolism , Phagosomes/metabolism , Phosphatidylinositol Phosphates/chemistry , Protein Binding , Protein Conformation , Vesicular Transport Proteins/chemistry
18.
Curr Opin Cell Biol ; 45: 47-54, 2017 04.
Article in English | MEDLINE | ID: mdl-28292700

ABSTRACT

Mammalian Unc-51-like kinases 1 and 2 (ULK1 and ULK2) belong to the ULK/Atg1 family of serine/threonine kinases, which are conserved from yeast to mammals. Although ULK/Atg1 is best known for regulating flux through the autophagy pathway, it has evolutionarily conserved noncanonical functions in protein trafficking that are essential for maintaining cellular homeostasis. As a direct target of energy- and nutrient-sensing kinases, ULK/Atg1 is positioned to regulate the distribution and use of cellular resources in response to metabolic cues. In this review, we provide an overview of the molecular mechanisms through which ULK/Atg1 carries out its canonical and noncanonical functions and the signaling pathways that link its function to metabolism. We also highlight potential contributions of ULK/Atg1 in human diseases, including cancer and neurodegeneration.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Autophagy , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Biological Transport , Humans , Mammals/metabolism , Molecular Targeted Therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics
19.
Annu Rev Biochem ; 86: 225-244, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28301741

ABSTRACT

Autophagy is the process of cellular self-eating by a double-membrane organelle, the autophagosome. A range of signaling processes converge on two protein complexes to initiate autophagy: the ULK1 (unc51-like autophagy activating kinase 1) protein kinase complex and the PI3KC3-C1 (class III phosphatidylinositol 3-kinase complex I) lipid kinase complex. Some 90% of the mass of these large protein complexes consists of noncatalytic domains and subunits, and the ULK1 complex has essential noncatalytic activities. Structural studies of these complexes have shed increasing light on the regulation of their catalytic and noncatalytic activities in autophagy initiation. The autophagosome is thought to nucleate from vesicles containing the integral membrane protein Atg9 (autophagy-related 9), COPII (coat protein complex II) vesicles, and possibly other sources. In the wake of reconstitution and super-resolution imaging studies, we are beginning to understand how the ULK1 and PI3KC3-C1 complexes might coordinate the nucleation and fusion of Atg9 and COPII vesicles at the start of autophagosome biogenesis.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phagosomes/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , COP-Coated Vesicles/metabolism , COP-Coated Vesicles/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Class III Phosphatidylinositol 3-Kinases/chemistry , Class III Phosphatidylinositol 3-Kinases/genetics , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Gene Expression , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Phagosomes/ultrastructure , Phosphatidylinositol 3-Kinase/chemistry , Phosphatidylinositol 3-Kinase/genetics , Protein Binding , Protein Multimerization , Signal Transduction
20.
Protein Cell ; 4(9): 711-21, 2013 Sep.
Article in English | MEDLINE | ID: mdl-27023913

ABSTRACT

Mutations in LR RK2 (Leucine rich repeat kinase 2) are a major cause of Parkinson's disease (PD). We and others reported recently that expression of the pathogenic gainof-function mutant form of LRRK2, LRRK2 G2019S, induces mitochondrial fission in neurons through DLP1. Here we provide evidence that expression of LRRK2 G2019S stimulates mitochondria loss or mitophagy. We have characterized several LRRK2 interacting proteins and found that LRRK2 interacts with ULK1 which plays an essential role in autophagy. Knockdown of either ULK1 or DLP1 expression with shRNAs suppresses LRRK2 G2019S expression-induced mitochondrial clearance, suggesting that LRRK2 G2019S expression induces mitochondrial fission through DLP1 followed by mitophagy via an ULK1 dependent pathway. In addition to ULK1, we found that LRRK2 interacts with the endogenous MKK4/7, JIP3 and coordinates with them in the activation of JNK signaling. Interestingly, LRRK2 G2019S-induced loss of mitochondria can also be suppressed by 3 different JNK inhibitors, implying the involvement of the JNK pathway in the pathogenic mechanism of mutated LRRK2. Thus our findings may provide an insight into the complicated pathogenesis of PD as well as some clues to the development of novel therapeutic strategies.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , MAP Kinase Signaling System , Mitophagy/genetics , Mitophagy/physiology , Mutation , Amino Acid Substitution , Autophagosomes/metabolism , Autophagosomes/pathology , Autophagy-Related Protein-1 Homolog/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Dynamins , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/chemistry , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...