Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
1.
Mol Cell ; 83(1): 74-89.e9, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36528027

ABSTRACT

The GATOR2-GATOR1 signaling axis is essential for amino-acid-dependent mTORC1 activation. However, the molecular function of the GATOR2 complex remains unknown. Here, we report that disruption of the Ring domains of Mios, WDR24, or WDR59 completely impedes amino-acid-mediated mTORC1 activation. Mechanistically, via interacting with Ring domains of WDR59 and WDR24, the Ring domain of Mios acts as a hub to maintain GATOR2 integrity, disruption of which leads to self-ubiquitination of WDR24. Physiologically, leucine stimulation dissociates Sestrin2 from the Ring domain of WDR24 and confers its availability to UBE2D3 and subsequent ubiquitination of NPRL2, contributing to GATOR2-mediated GATOR1 inactivation. As such, WDR24 ablation or Ring deletion prevents mTORC1 activation, leading to severe growth defects and embryonic lethality at E10.5 in mice. Hence, our findings demonstrate that Ring domains are essential for GATOR2 to transmit amino acid availability to mTORC1 and further reveal the essentiality of nutrient sensing during embryonic development.


Subject(s)
Multiprotein Complexes , TOR Serine-Threonine Kinases , Animals , Mice , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Signal Transduction
2.
Nature ; 609(7927): 502-506, 2022 09.
Article in English | MEDLINE | ID: mdl-36104553

ABSTRACT

Hund's multiplicity rule states that a higher spin state has a lower energy for a given electronic configuration1. Rephrasing this rule for molecular excited states predicts a positive energy gap between spin-singlet and spin-triplet excited states, as has been consistent with numerous experimental observations over almost a century. Here we report a fluorescent molecule that disobeys Hund's rule and has a negative singlet-triplet energy gap of -11 ± 2 meV. The energy inversion of the singlet and triplet excited states results in delayed fluorescence with short time constants of 0.2 µs, which anomalously decrease with decreasing temperature owing to the emissive singlet character of the lowest-energy excited state. Organic light-emitting diodes (OLEDs) using this molecule exhibited a fast transient electroluminescence decay with a peak external quantum efficiency of 17%, demonstrating its potential implications for optoelectronic devices, including displays, lighting and lasers.

3.
Cell ; 150(1): 179-93, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770219

ABSTRACT

Aberrant Skp2 signaling has been implicated as a driving event in tumorigenesis. Although the underlying molecular mechanisms remain elusive, cytoplasmic Skp2 correlates with more aggressive forms of breast and prostate cancers. Here, we report that Skp2 is acetylated by p300 at K68 and K71, which is a process that can be antagonized by the SIRT3 deacetylase. Inactivation of SIRT3 leads to elevated Skp2 acetylation, which leads to increased Skp2 stability through impairment of the Cdh1-mediated proteolysis pathway. As a result, Skp2 oncogenic function is increased, whereby cells expressing an acetylation-mimetic mutant display enhanced cellular proliferation and tumorigenesis in vivo. Moreover, acetylation of Skp2 in the nuclear localization signal (NLS) promotes its cytoplasmic retention, and cytoplasmic Skp2 enhances cellular migration through ubiquitination and destruction of E-cadherin. Thus, our study identifies an acetylation-dependent regulatory mechanism governing Skp2 oncogenic function and provides insight into how cytoplasmic Skp2 controls cellular migration.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Cadherins/metabolism , Casein Kinase I/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Disease Models, Animal , Humans , Lysine/metabolism , Male , Mice , Molecular Sequence Data , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Protein Sorting Signals , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , Sequence Alignment , Ubiquitination
4.
Mol Cell ; 68(4): 645-658.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149593

ABSTRACT

Hajdu-Cheney syndrome (HCS), a rare autosomal disorder caused by heterozygous mutations in NOTCH2, is clinically characterized by acro-osteolysis, severe osteoporosis, short stature, neurological symptoms, cardiovascular defects, and polycystic kidneys. Recent studies identified that aberrant NOTCH2 signaling and consequent osteoclast hyperactivity are closely associated with the bone-related disorder pathogenesis, but the exact molecular mechanisms remain unclear. Here, we demonstrate that sustained osteoclast activity is largely due to accumulation of NOTCH2 carrying a truncated C terminus that escapes FBW7-mediated ubiquitination and degradation. Mice with osteoclast-specific Fbw7 ablation revealed osteoporotic phenotypes reminiscent of HCS, due to elevated Notch2 signaling. Importantly, administration of Notch inhibitors in Fbw7 conditional knockout mice alleviated progressive bone resorption. These findings highlight the molecular basis of HCS pathogenesis and provide clinical insights into potential targeted therapeutic strategies for skeletal disorders associated with the aberrant FBW7/NOTCH2 pathway as observed in patients with HCS.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Hajdu-Cheney Syndrome , Mutation , Osteoporosis , Proteolysis , Receptor, Notch2 , Animals , Cell Line , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Hajdu-Cheney Syndrome/genetics , Hajdu-Cheney Syndrome/metabolism , Mice, Knockout , Osteoporosis/genetics , Osteoporosis/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Ubiquitination/genetics
5.
J Biol Chem ; 299(4): 104572, 2023 04.
Article in English | MEDLINE | ID: mdl-36870680

ABSTRACT

Post-translational modifications (PTMs) regulate all aspects of protein function. Therefore, upstream regulators of PTMs, such as kinases, acetyltransferases, or methyltransferases, are potential therapeutic targets for human diseases, including cancer. To date, multiple inhibitors and/or agonists of these PTM upstream regulators are in clinical use, while others are still in development. However, these upstream regulators control not only the PTMs of disease-related target proteins but also other disease-irrelevant substrate proteins. Thus, nontargeted perturbing activities may introduce unwanted off-target toxicity issues that limit the use of these drugs in successful clinical applications. Therefore, alternative drugs that solely regulate a specific PTM of the disease-relevant protein target may provide a more precise effect in treating disease with relatively low side effects. To this end, chemically induced proximity has recently emerged as a powerful research tool, and several chemical inducers of proximity (CIPs) have been used to target and regulate protein ubiquitination, phosphorylation, acetylation, and glycosylation. These CIPs have a high potential to be translated into clinical drugs and several examples such as PROTACs and MGDs are now in clinical trials. Hence, more CIPs need to be developed to cover all types of PTMs, such as methylation and palmitoylation, thus providing a full spectrum of tools to regulate protein PTM in basic research and also in clinical application for effective cancer treatment.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Humans , Proteins , Ubiquitination , Phosphorylation , Glycosylation , Acetylation , Neoplasms/drug therapy
6.
J Am Chem Soc ; 146(11): 7584-7593, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38469801

ABSTRACT

Given the prevalent advancements in DNA- and RNA-based PROTACs, there remains a significant need for the exploration and expansion of more specific DNA-based tools, thus broadening the scope and repertoire of DNA-based PROTACs. Unlike conventional A- or B-form DNA, Z-form DNA is a configuration that exclusively manifests itself under specific stress conditions and with specific target sequences, which can be recognized by specific reader proteins, such as ADAR1 or ZBP1, to exert downstream biological functions. The core of our innovation lies in the strategic engagement of Z-form DNA with ADAR1 and its degradation is achieved by leveraging a VHL ligand conjugated to Z-form DNA to recruit the E3 ligase. This ingenious construct engendered a series of Z-PROTACs, which we utilized to selectively degrade the Z-DNA-binding protein ADAR1, a molecule that is frequently overexpressed in cancer cells. This meticulously orchestrated approach triggers a cascade of PANoptotic events, notably encompassing apoptosis and necroptosis, by mitigating the blocking effect of ADAR1 on ZBP1, particularly in cancer cells compared with normal cells. Moreover, the Z-PROTAC design exhibits a pronounced predilection for ADAR1, as opposed to other Z-DNA readers, such as ZBP1. As such, Z-PROTAC likely elicits a positive immunological response, subsequently leading to a synergistic augmentation of cancer cell death. In summary, the Z-DNA-based PROTAC (Z-PROTAC) approach introduces a modality generated by the conformational change from B- to Z-form DNA, which harnesses the structural specificity intrinsic to potentiate a selective degradation strategy. This methodology is an inspiring conduit for the advancement of PROTAC-based therapeutic modalities, underscoring its potential for selectivity within the therapeutic landscape of PROTACs to target undruggable proteins.


Subject(s)
DNA, Z-Form , Proteolysis Targeting Chimera , Proteolysis , Adenosine Deaminase/metabolism , RNA/metabolism , Ubiquitin-Protein Ligases/metabolism , DNA-Binding Proteins/metabolism
7.
J Am Chem Soc ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38597345

ABSTRACT

Deubiquitinase-targeting chimeras (DUBTACs) have been recently developed to stabilize proteins of interest, which is in contrast to targeted protein degradation (TPD) approaches that degrade disease-causing proteins. However, to date, only the OTUB1 deubiquitinase has been utilized to develop DUBTACs via an OTUB1 covalent ligand, which could unexpectedly compromise the endogenous function of OTUB1 owing to its covalent nature. Here, we show for the first time that deubiquitinase USP7 can be harnessed for DUBTAC development. Based on a noncovalent ligand of USP7, we developed USP7-based DUBTACs that stabilized the ΔF508-CFTR mutant protein as effectively as the previously reported OTUB1-based DUBTAC. Importantly, using two different noncovalent ligands of USP7, we developed the first AMPK DUBTACs that appear to selectively stabilize different isoforms of AMPKß, leading to elevated AMPK signaling. Overall, these results highlight that, in addition to OTUB1, USP7 can be leveraged to develop DUBTACs, thus significantly expanding the limited toolbox for targeted protein stabilization and the development of novel AMPK DUBTACs as potential therapeutics.

8.
Semin Cancer Biol ; 86(Pt 2): 269-279, 2022 11.
Article in English | MEDLINE | ID: mdl-35798235

ABSTRACT

Tumor microenvironment (TME) composes of multiple cell types and non-cellular components, which supports the proliferation, metastasis and immune surveillance evasion of tumor cells, as well as accounts for the resistance to therapies. Therefore, therapeutic strategies using small molecule inhibitors (SMIs) and antibodies to block potential targets in TME are practical for cancer treatment. Targeted protein degradation using PROteolysis-TArgeting Chimera (PROTAC) technic has several advantages over traditional SMIs and antibodies, including overcoming drug resistance. Thus many PROTACs are currently under development for cancer treatment. In this review, we summarize the recent progress of PROTAC development that target TME pathways and propose the potential direction of future PROTAC technique to advance as novel cancer treatment options.


Subject(s)
Drug Discovery , Neoplasms , Humans , Drug Discovery/methods , Proteolysis , Neoplasms/drug therapy , Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Tumor Microenvironment
9.
Semin Cancer Biol ; 86(Pt 3): 32-40, 2022 11.
Article in English | MEDLINE | ID: mdl-35908574

ABSTRACT

Necroptosis is a caspase-independent form of programmed cell death executed by the receptor interacting protein kinase 1 (RIPK1)-RIPK3-mixed lineage kinase domain-like protein (MLKL) signaling cascade, deregulation of which can cause various human diseases including cancer. Escape from programmed cell death is a hallmark of cancer, leading to uncontrolled growth and drug resistance. Therefore, it is crucial to further understand whether necroptosis plays a key role in therapeutic resistance. In this review, we summarize the recent findings of the link between necroptosis and cancer, and discuss that targeting necroptosis is a new strategy to overcome apoptosis resistance in tumor therapy.


Subject(s)
Necroptosis , Protein Kinases , Humans , Necroptosis/genetics , Apoptosis/genetics , Signal Transduction , Carcinogenesis/genetics
10.
J Am Chem Soc ; 145(40): 21871-21878, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37774414

ABSTRACT

Methyl-CpG-binding protein 2 (MeCP2), a reader of DNA methylation, has been extensively investigated for its function in neurological and neurodevelopmental disorders. Emerging evidence indicates that MeCP2 exerts an oncogenic function in cancer; however, the endeavor to develop a MeCP2-targeted therapy remains a challenge. This work attempts to address it by introducing a methylated nucleotide-based targeting chimera termed methyl-proteolysis-targeting chimera (methyl-PROTAC). The methyl-PROTAC incorporates a methylated cytosine into an oligodeoxynucleotide moiety to recruit MeCP2 for targeted degradation in a von Hippel-Lindau- and proteasome-dependent manner, thus displaying antiproliferative effects in cancer cells reliant on MeCP2 overexpression. This selective cytotoxicity endows methyl-PROTAC with the capacity to selectively eliminate cancer cells that are addicted to the overexpression of the MeCP2 oncoprotein. Furthermore, methyl-PROTAC-mediated MeCP2 degradation induces apoptosis in cancer cells. These findings underscore the therapeutic potential of methyl-PROTAC to degrade undruggable epigenetic regulatory proteins. In summary, the development of methyl-PROTAC introduces an innovative strategy by designing a modified nucleotide-based degradation approach for manipulating epigenetic factors, thereby representing a promising avenue for the advancement of PROTAC-based therapeutics.


Subject(s)
Methyl-CpG-Binding Protein 2 , Nucleotides , Methyl-CpG-Binding Protein 2/genetics , Nucleotides/metabolism , Proteolysis , Transcription Factors/metabolism , DNA Methylation
11.
Mol Cell ; 59(6): 917-30, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26344095

ABSTRACT

The ERG gene is fused to TMPRSS2 in approximately 50% of prostate cancers (PrCa), resulting in its overexpression. However, whether this is the sole mechanism underlying ERG elevation in PrCa is currently unclear. Here we report that ERG ubiquitination and degradation are governed by the Cullin 3-based ubiquitin ligase SPOP and that deficiency in this pathway leads to aberrant elevation of the ERG oncoprotein. Specifically, we find that truncated ERG (ΔERG), encoded by the ERG fusion gene, is stabilized by evading SPOP-mediated destruction, whereas prostate cancer-associated SPOP mutants are also deficient in promoting ERG ubiquitination. Furthermore, we show that the SPOP/ERG interaction is modulated by CKI-mediated phosphorylation. Importantly, we demonstrate that DNA damage drugs, topoisomerase inhibitors, can trigger CKI activation to restore the SPOP/ΔERG interaction and its consequent degradation. Therefore, SPOP functions as a tumor suppressor to negatively regulate the stability of the ERG oncoprotein in prostate cancer.


Subject(s)
Nuclear Proteins/physiology , Prostatic Neoplasms/metabolism , Repressor Proteins/physiology , Trans-Activators/metabolism , Ubiquitination , Amino Acid Sequence , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Movement , Cullin Proteins/metabolism , Disease Progression , Etoposide/pharmacology , HEK293 Cells , Humans , Male , Molecular Sequence Data , Neoplasm Invasiveness , Prostatic Neoplasms/pathology , Protein Interaction Domains and Motifs , Proteolysis , Transcriptional Regulator ERG , Tumor Suppressor Proteins/physiology
12.
J Am Chem Soc ; 144(16): 7302-7307, 2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35414173

ABSTRACT

Chirality-induced current-perpendicular-to-plane magnetoresistance (CPP-MR) originates from current-induced spin polarization in molecules. The current-induced spin polarization is widely recognized as a fundamental principle of chiral-induced spin selectivity (CISS). In this study, we investigate chirality-induced current-in-plane magnetoresistance (CIP-MR) in a chiral molecule/ferromagnetic metal bilayer at room temperature. In contrast to CPP-MR, CIP-MR observed in the present study requires no bias charge current through the molecule. The temperature dependence of CIP-MR suggests that thermally driven spontaneous spin polarization in chiral molecules is the key to the observed MR. The novel MR is consistent with recent CISS-related studies, that is, chiral molecules in contact with a metallic surface possess a finite spin polarization.

13.
Mol Cell ; 56(4): 595-607, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25458846

ABSTRACT

Oncogenic mutations in PIK3CA, the gene encoding the catalytic subunit of phosphoinositide 3-kinase (PI3K), occur with high frequency in breast cancer. The protein kinase Akt is considered to be the primary effector of PIK3CA, although mechanisms by which PI3K mediates Akt-independent tumorigenic signals remain obscure. We show that serum and glucocorticoid-regulated kinase 3 (SGK3) is amplified in breast cancer and activated downstream of PIK3CA in a manner dependent on the phosphoinositide phosphatase INPP4B. Expression of INPP4B leads to enhanced SGK3 activation and suppression of Akt phosphorylation. Activation of SGK3 downstream of PIK3CA and INPP4B is required for 3D proliferation, invasive migration, and tumorigenesis in vivo. We further show that SGK3 targets the metastasis suppressor NDRG1 for degradation by Fbw7. We propose a model in which breast cancers harboring oncogenic PIK3CA activate SGK3 signaling while suppressing Akt, indicative of oncogenic functions for both INPP4B and SGK3 in these tumors.


Subject(s)
Breast Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases , Enzyme Activation , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Mutation , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/physiology , Protein Processing, Post-Translational , Proteolysis , Signal Transduction
14.
J Biol Chem ; 295(45): 15328-15341, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32868297

ABSTRACT

Dental enamel, the hardest tissue in the human body, is derived from dental epithelial cell ameloblast-secreted enamel matrices. Enamel mineralization occurs in a strictly synchronized manner along with ameloblast maturation in association with ion transport and pH balance, and any disruption of these processes results in enamel hypomineralization. G protein-coupled receptors (GPCRs) function as transducers of external signals by activating associated G proteins and regulate cellular physiology. Tissue-specific GPCRs play important roles in organ development, although their activities in tooth development remain poorly understood. The present results show that the adhesion GPCR Gpr115 (Adgrf4) is highly and preferentially expressed in mature ameloblasts and plays a crucial role during enamel mineralization. To investigate the in vivo function of Gpr115, knockout (Gpr115-KO) mice were created and found to develop hypomineralized enamel, with a larger acidic area because of the dysregulation of ion composition. Transcriptomic analysis also revealed that deletion of Gpr115 disrupted pH homeostasis and ion transport processes in enamel formation. In addition, in vitro analyses using the dental epithelial cell line cervical loop-derived dental epithelial (CLDE) cell demonstrated that Gpr115 is indispensable for the expression of carbonic anhydrase 6 (Car6), which has a critical role in enamel mineralization. Furthermore, an acidic condition induced Car6 expression under the regulation of Gpr115 in CLDE cells. Thus, we concluded that Gpr115 plays an important role in enamel mineralization via regulation of Car6 expression in ameloblasts. The present findings indicate a novel function of Gpr115 in ectodermal organ development and clarify the molecular mechanism of enamel formation.


Subject(s)
Ameloblasts/metabolism , Dental Enamel/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cells, Cultured , Mice , Mice, Knockout , Rats , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics
15.
FASEB J ; 34(11): 14930-14945, 2020 11.
Article in English | MEDLINE | ID: mdl-32931083

ABSTRACT

Proteasome inhibitors exert an anabolic effect on bone formation with elevated levels of osteoblast markers. These findings suggest the important role of the proteasomal degradation of osteogenic regulators, while the underlying molecular mechanisms are not fully understood. Here, we report that the proteasome inhibitors bortezomib and ixazomib markedly increased protein levels of the osteoblastic key transcription factor osterix/Sp7 (Osx). Furthermore, we revealed that Osx was targeted by p38 and Fbw7 for proteasomal degradation. Mechanistically, p38-mediated Osx phosphorylation at S73/77 facilitated Fbw7 interaction to trigger subsequent Osx ubiquitination. Consistent with these findings, p38 knockdown or pharmacological p38 inhibition resulted in Osx protein stabilization. Treatment with p38 inhibitors following osteogenic stimulation efficiently induced osteoblast differentiation through Osx stabilization. Conversely, pretreatment of p38 inhibitor followed by osteogenic challenge impaired osteoblastogenesis via suppressing Osx expression, suggesting that p38 exerts dual but opposite effects in the regulation of Osx level to fine-tune its activity during osteoblast differentiation. Furthermore, Fbw7-depleted human mesenchymal stem cells and primary mouse calvarial cells resulted in increased osteogenic capacity. Together, our findings unveil the molecular mechanisms underlying the Osx protein stability control and suggest that targeting the Osx degradation pathway could help enhance efficient osteogenesis and bone matrix regeneration.


Subject(s)
Cell Differentiation , Osteoblasts/metabolism , Proteolysis , Sp7 Transcription Factor/metabolism , Animals , Boron Compounds/pharmacology , Bortezomib/pharmacology , Cells, Cultured , F-Box-WD Repeat-Containing Protein 7/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , HCT116 Cells , HEK293 Cells , Humans , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Sp7 Transcription Factor/genetics , Ubiquitination , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Int J Mol Sci ; 22(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33809261

ABSTRACT

Lipin2 is a phosphatidate phosphatase that plays critical roles in fat homeostasis. Alterations in Lpin2, which encodes lipin2, cause the autoinflammatory bone disorder Majeed syndrome. Lipin2 limits lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. However, little is known about the precise molecular mechanisms underlying its anti-inflammatory function. In this study, we attempted to elucidate the molecular link between the loss of lipin2 function and autoinflammatory bone disorder. Using a Lpin2 knockout murine macrophage cell line, we showed that lipin2 deficiency enhances innate immune responses to LPS stimulation through excessive activation of the NF-κB signaling pathway, partly because of TAK1 signaling upregulation. Lipin2 depletion also enhanced RANKL-mediated osteoclastogenesis and osteoclastic resorption activity accompanied by NFATc1 dephosphorylation and increased nuclear accumulation. These results suggest that lipin2 suppresses the development of autoinflammatory bone disorder by fine-tuning proinflammatory responses and osteoclastogenesis in macrophages. Therefore, this study provides insights into the molecular pathogenesis of monogenic autoinflammatory bone disorders and presents a potential therapeutic intervention.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , Immunologic Deficiency Syndromes/genetics , Inflammation/genetics , MAP Kinase Kinase Kinases/genetics , NFATC Transcription Factors/genetics , Nuclear Proteins/genetics , Osteomyelitis/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Anemia, Dyserythropoietic, Congenital/metabolism , Anemia, Dyserythropoietic, Congenital/pathology , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/genetics , Humans , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Osteomyelitis/metabolism , Osteomyelitis/pathology , RANK Ligand/genetics , Signal Transduction/genetics , Transcription Factor RelA/genetics
17.
Angew Chem Int Ed Engl ; 60(30): 16377-16381, 2021 07 19.
Article in English | MEDLINE | ID: mdl-33955147

ABSTRACT

Graphitic carbon nitride (GCN) has garnered broad research interest due to its unique catalytic properties. However, GCN, prepared by general methods, possesses myriad structural defects and it has been difficult to elucidate their intrinsic physical properties. We report the development of azacalix[3]triazines (AC3Ts), a substructure of triazine-based GCN (Tz-GCN). Despite the electron-deficient natures of triazine, AC3Ts capture protons as organic superbases. We reveal the unique anion-π interactions of AC3Ts that alters the ionization potentials of AC3Ts. To the best of our knowledge, these features have not yet been recognized for Tz-GCN. These unveiled features of AC3Ts are expected to expand the usage scope and possibilities for GCNs.

18.
Angew Chem Int Ed Engl ; 60(6): 3261-3267, 2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33098203

ABSTRACT

Polar materials attract wide research interest due to their unique properties, such as ferroelectricity and the bulk photovoltaic effect (BPVE), which are not accessible with nonpolar materials. However, in general, rationally designing polar materials is difficult because nonpolar materials are more favorable in terms of dipole-dipole interactions. Here, we report a rational strategy to form polar assemblies with bowl-shaped π-conjugated molecules and a molecular design principle for this strategy. We synthesized and thoroughly characterized 12 single crystals with the help of various theoretical calculations. Furthermore, we demonstrated that it can be possible to predict whether polar assemblies become more favorable or not by estimating their lattice energies. We believe that this study contributes to the development of organic polar materials and their related studies.

19.
Biochem Biophys Res Commun ; 524(2): 477-483, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32008742

ABSTRACT

Lipin-2 is a phosphatidate phosphatase with key roles in regulating lipid storage and energy homeostasis. LPIN2-genetic deficiency is associated with an autoinflammatory disorder, underscoring its critical role in innate immune signaling; however, the regulatory mechanisms underlying protein stability remain unknown. Here, we demonstrate that Lipin-2 interacts with ß-TRCP, a substrate receptor subunit of the SCFß-TRCP E3 ligase, and undergoes ubiquitination and proteasomal degradation. ß-TRCP-knockout in RAW264.7 macrophages resulted in Lipin-2 accumulation, leading to the suppression of LPS-induced MAPK activation and subsequent proinflammatory gene expression. Consistent with this, treatment with MLN4924, a Cullin-neddylation inhibitor that suppresses SCF E3 activity, increased Lipin-2 protein and concomitantly decreased Il1b expression. These findings suggested that ß-TRCP-mediated Lipin-2 degradation affects macrophage-elicited proinflammatory responses and could lead to new therapeutic approaches to treat inflammatory diseases.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , Phosphatidate Phosphatase/metabolism , Proteolysis , Animals , Gene Expression Regulation , Gene Knockout Techniques , HEK293 Cells , Humans , Inflammation/genetics , Mice , Phosphatidate Phosphatase/genetics , RAW 264.7 Cells , Ubiquitination , beta-Transducin Repeat-Containing Proteins/genetics , beta-Transducin Repeat-Containing Proteins/metabolism
20.
Nature ; 508(7497): 541-5, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24670654

ABSTRACT

Akt, also known as protein kinase B, plays key roles in cell proliferation, survival and metabolism. Akt hyperactivation contributes to many pathophysiological conditions, including human cancers, and is closely associated with poor prognosis and chemo- or radiotherapeutic resistance. Phosphorylation of Akt at S473 (ref. 5) and T308 (ref. 6) activates Akt. However, it remains unclear whether further mechanisms account for full Akt activation, and whether Akt hyperactivation is linked to misregulated cell cycle progression, another cancer hallmark. Here we report that Akt activity fluctuates across the cell cycle, mirroring cyclin A expression. Mechanistically, phosphorylation of S477 and T479 at the Akt extreme carboxy terminus by cyclin-dependent kinase 2 (Cdk2)/cyclin A or mTORC2, under distinct physiological conditions, promotes Akt activation through facilitating, or functionally compensating for, S473 phosphorylation. Furthermore, deletion of the cyclin A2 allele in the mouse olfactory bulb leads to reduced S477/T479 phosphorylation and elevated cellular apoptosis. Notably, cyclin A2-deletion-induced cellular apoptosis in mouse embryonic stem cells is partly rescued by S477D/T479E-Akt1, supporting a physiological role for cyclin A2 in governing Akt activation. Together, the results of our study show Akt S477/T479 phosphorylation to be an essential layer of the Akt activation mechanism to regulate its physiological functions, thereby providing a new mechanistic link between aberrant cell cycle progression and Akt hyperactivation in cancer.


Subject(s)
Cell Cycle/physiology , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Cell Proliferation , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enzyme Activation , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Olfactory Bulb/cytology , Olfactory Bulb/enzymology , Olfactory Bulb/metabolism , Oncogene Protein v-akt/chemistry , Oncogene Protein v-akt/metabolism , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL