Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Oncogene ; 40(48): 6614-6626, 2021 12.
Article in English | MEDLINE | ID: mdl-34621020

ABSTRACT

KIT/PDGFRA oncogenic tyrosine kinase signaling is the central oncogenic event in most gastrointestinal stromal tumors (GIST), which are human malignant mesenchymal neoplasms that often feature myogenic differentiation. Although targeted inhibition of KIT/PDGFRA provides substantial clinical benefit, GIST cells adapt to KIT/PDGFRA driver suppression and eventually develop resistance. The specific molecular events leading to adaptive resistance in GIST remain unclear. By using clinically representative in vitro and in vivo GIST models and GIST patients' samples, we found that the E3 ubiquitin ligase Atrogin-1 (FBXO32)-the main effector of muscular atrophy in cachexia-resulted in the most critical gene derepressed in response to KIT inhibition, regardless the type of KIT primary or secondary mutation. Atrogin-1 in GISTs is transcriptionally controlled by the KIT-FOXO3a axis, thus indicating overlap with Atrogin-1 regulation mechanisms in nonneoplastic muscle cells. Further, Atrogin-1 overexpression was a GIST-cell-specific pro-survival mechanism that enabled the adaptation to KIT-targeted inhibition by apoptosis evasion through cell quiescence. Buttressed on these findings, we established in vitro and in vivo the preclinical proof-of-concept for co-targeting KIT and the ubiquitin pathway to maximize the therapeutic response to first-line imatinib treatment.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Gastrointestinal Stromal Tumors/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Imatinib Mesylate/pharmacology , Muscle Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Sulfides/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Drug Therapy, Combination , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Humans , Mice , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Balkan Med J ; 38(1): 43-49, 2021 01.
Article in English | MEDLINE | ID: mdl-32975519

ABSTRACT

BACKGROUND: Multiple myeloma remains a virtually incurable hematologic malignancy, which is featured with the aberrant growth of malignant plasma cells. AIMS: To elucidate the functions of miR-19a-3p in multiple myeloma. STUDY DESIGN: Cell study. METHODS: Cell counting kit-8 assay was performed to detect cell viability, and flow cytometry was conducted to detect cell apoptosis. Bioinformatics analysis predicted miR-19a-3p-associated biological function, pathway, core regulatory network, and target genes. Luciferase reporter assay verified the target sequence of miR-19a-3p regulating FBXO32. RESULTS: miR-19a-3p is upregulated in multiple myeloma cells (p<0.01) and patients with multiple myeloma (p<0.001). Overexpressed miR-19a-3p significantly increased cell viability (p<0.05) and inhibited cell apoptosis (p<0.01). FBXO32 is a target gene of miR-19a-3p (p<0.01). Moreover, FBXO32 is downregulated in MM, and it significantly decreased cell viability (p<0.05) and promoted cell apoptosis (p<0.01). FBXO32 significantly rescued the influence of miR-19a-3p-inhibiting cell apoptosis (p<0.05). CONCLUSION: miR-19a-3p promoted cell proliferation and inhibited cell apoptosis by degrading the target FBXO32 mRNA in multiple myeloma.


Subject(s)
MicroRNAs/pharmacology , Multiple Myeloma/drug therapy , Muscle Proteins/antagonists & inhibitors , Oncogenes/drug effects , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Adult , Apoptosis/drug effects , Female , Humans , Male , MicroRNAs/therapeutic use , Middle Aged , Multiple Myeloma/physiopathology , Muscle Proteins/therapeutic use , Oncogenes/physiology , SKP Cullin F-Box Protein Ligases/therapeutic use
3.
Chem Biol Interact ; 304: 43-51, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30849338

ABSTRACT

Accumulations of uremic toxins has been widely recognized as the major trigger of skeletal muscle loss in chronic kidney disease (CKD), which is defined as uremic sarcopenia. Current study was aimed to examine the effects of representative uremic toxin, indoxyl sulfate (IS), on C2C12 myotubes. The incubation of IS (from 0.1 mM to 1.2 mM) exerted the reduction in myotube diameter without cell survival impairment. Elevated oxidative stress and mitogen-activated protein kinase (MAPKs) phosphorylation were observed after IS stimulation for 1 and 24 h. After N-acetylcysteine (NAC) treatment as antioxidants, the recovery in IS-induced decrease myotube diameter and ERK phosphorylation was observed. This findings were implicit the transduction of p-ERK in IS-induced ROS toxicity. Moreover, the increase of LC3ß was found closely with IS treatment in C2C12 myotubes. The reverse effect of NAC on LC3ß expression revealed the ROS-responsibility in autophagy regulation of CKD myopathy. The evaluation of IS-treated proteasome system showed increased phospho-myosin light chain, along with the upregulation of muscle atrophy F-box (MAFbx) mRNA and protein. This alteration in MAFbx was also identified in nephrectomy-induced CKD model. Besides, the inhibition of p-JNK was capable to attenuate IS-induced upward change in MAFbx protein expression. These findings indicated that IS-mediated myotube atrophy may manipulate through ROS-ERK axis and JNK-MAFbx regulation in C2C12 cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Indican/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins/antagonists & inhibitors , Muscular Atrophy/chemically induced , Reactive Oxygen Species/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Muscle Proteins/metabolism , Muscular Atrophy/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Structure-Activity Relationship
4.
Biol Reprod ; 100(4): 896-906, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30535233

ABSTRACT

The mechanism of maternal protein degradation during preimplantation development has not been clarified yet. It is thought that a lot of maternal proteins are degraded by the ubiquitin-proteasome system. In this study, we focused on the role of the SCF (Skp1-Cullin-F-box) complexes during early bovine embryogenesis. We inhibited them using MLN4924, an inhibitor of SCF complex ligases controlled by neddylation. Oocytes maturated in MLN4924 could be fertilized, but we found no cumulus cell expansion and a high number of polyspermy after in vitro fertilization. We also found a statistically significant deterioration of development after MLN4924 treatment. After treatment with MLN4924 from the four-cell to late eight-cell stage, we found a statistically significant delay in their development; some of the treated embryos were, however, able to reach the blastocyst stage later. We found reduced levels of mRNA of EGA markers PAPOLA and U2AF1A, which can be related to this developmental delay. The cultivation with MLN4924 caused a significant increase in protein levels in MLN4924-treated oocytes and embryos; no such change was found in cumulus cells. To detect the proteins affected by MLN4924 treatment, we performed a Western blot analysis of selected proteins (SMAD4, ribosomal protein S6, centromeric protein E, P27, NFKB inhibitor alpha, RNA-binding motif protein 19). No statistically significant increase in protein levels was detected in either treated embryos or oocytes. In summary, our study shows that SCF ligases are necessary for the correct maturation of oocytes, cumulus cell expansion, fertilization, and early preimplantation development of cattle.


Subject(s)
Blastocyst/drug effects , Cyclopentanes/pharmacology , Embryonic Development/drug effects , Oocytes/drug effects , Oogenesis/drug effects , Pyrimidines/pharmacology , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Animals , Blastocyst/cytology , Blastocyst/physiology , Cattle , Cells, Cultured , Embryo, Mammalian , Female , In Vitro Oocyte Maturation Techniques/methods , In Vitro Oocyte Maturation Techniques/veterinary , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Oocytes/cytology , Oocytes/physiology , SKP Cullin F-Box Protein Ligases/metabolism , SKP Cullin F-Box Protein Ligases/physiology , Time Factors
5.
Nature ; 563(7733): 652-656, 2018 11.
Article in English | MEDLINE | ID: mdl-30464344

ABSTRACT

The strigolactones, a class of plant hormones, regulate many aspects of plant physiology. In the inhibition of shoot branching, the α/ß hydrolase D14-which metabolizes strigolactone-interacts with the F-box protein D3 to ubiquitinate and degrade the transcription repressor D53. Despite the fact that multiple modes of interaction between D14 and strigolactone have recently been determined, how the hydrolase functions with D3 to mediate hormone-dependent D53 ubiquitination remains unknown. Here we show that D3 has a C-terminal α-helix that can switch between two conformational states. The engaged form of this α-helix facilitates the binding of D3 and D14 with a hydrolysed strigolactone intermediate, whereas the dislodged form can recognize unmodified D14 in an open conformation and inhibits its enzymatic activity. The D3 C-terminal α-helix enables D14 to recruit D53 in a strigolactone-dependent manner, which in turn activates the hydrolase. By revealing the structural plasticity of the SCFD3-D14 ubiquitin ligase, our results suggest a mechanism by which the E3 coordinates strigolactone signalling and metabolism.


Subject(s)
Heterocyclic Compounds, 3-Ring/metabolism , Lactones/metabolism , Oryza/enzymology , Oryza/metabolism , Plant Growth Regulators/metabolism , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Heterocyclic Compounds, 3-Ring/chemistry , Lactones/chemistry , Models, Molecular , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Plant Growth Regulators/chemistry , Plant Proteins/chemistry , Plant Proteins/metabolism , Protein Binding , Protein Structure, Secondary , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Structure-Activity Relationship , Ubiquitin , Ubiquitination
6.
Structure ; 26(9): 1226-1236.e3, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30033217

ABSTRACT

Skp1-Cul1-F-box (SCF) E3 ligases constitute the largest and best-characterized family of the multisubunit E3 ligases with important cellular functions and numerous disease links. The specificity of an SCF E3 ligase is established by one of the 69 human F-box proteins that are recruited to Cul1 through the Skp1 adaptor. We previously reported generation of ubiquitin variants (UbVs) targeting Fbw7 and Fbw11, which inhibit ligase activity by binding at the F-box-Skp1 interface to competitively displace Cul1. In the present study, we employed an optimized engineering strategy to generate specific binding UbVs against 17 additional Skp1-F-box complexes. We validated our design strategy and uncovered the structural basis of binding specificity by crystallographic analyses of representative UbVs bound to Skp1-Fbl10 and Skp1-Fbl11. Our study highlights the power of combining phage display with structure-based design to develop UbVs targeting specific protein surfaces.


Subject(s)
SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitins/chemistry , Ubiquitins/pharmacology , Binding Sites , Cell Surface Display Techniques , Crystallography, X-Ray , Cullin Proteins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , F-Box Proteins/metabolism , Female , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Models, Molecular , Protein Binding , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
7.
Life Sci ; 205: 91-96, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29753766

ABSTRACT

AIMS: Muscle mass is regulated by the balance between the synthesis and degradation of muscle proteins. Loss of skeletal muscle mass is associated with an increased risk of developing metabolic diseases such as obesity and type 2 diabetes mellitus. The aim of this study was to clarify the effects of licorice flavonoid oil on muscle mass in KK-Ay/Ta mice. MAIN METHODS: Male genetically type II diabetic KK-Ay/Ta mice received 0, 1, or 1.5 g/kg BW of licorice flavonoid oil by mouth once daily for 4 weeks. After 4 weeks, the femoral and soleus muscles were collected for western blotting for evaluation of the mTOR/p70 S6K, p38/FoxO3a, and Akt/FoxO3a signaling pathways. KEY FINDINGS: Ingestion of licorice flavonoid oil significantly enhanced femoral muscle mass without affecting body weight in KK-Ay/Ta mice. Licorice flavonoid oil also decreased expression of MuRF1 and atrogin-1, which are both markers of muscle atrophy. The mechanisms by which licorice flavonoid oil enhances muscle mass include activation of mTOR and p70 S6K, and regulation of phosphorylation of FoxO3a. SIGNIFICANCE: Ingestion of licorice flavonoids may help to prevent muscle atrophy.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Flavonoids/pharmacology , Glycyrrhiza/chemistry , Muscle, Skeletal/drug effects , Plant Oils/pharmacology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/metabolism , Forkhead Box Protein O3/biosynthesis , Forkhead Box Protein O3/genetics , Intracellular Signaling Peptides and Proteins , Male , Mice , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Organ Size/drug effects , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/biosynthesis , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/biosynthesis , Tripartite Motif Proteins , p38 Mitogen-Activated Protein Kinases/biosynthesis
8.
Open Biol ; 8(2)2018 02.
Article in English | MEDLINE | ID: mdl-29445034

ABSTRACT

Deregulation of centriole duplication has been implicated in cancer and primary microcephaly. Accordingly, it is important to understand how key centriole duplication factors are regulated. E3 ubiquitin ligases have been implicated in controlling the levels of several duplication factors, including PLK4, STIL and SAS-6, but the precise mechanisms ensuring centriole homeostasis remain to be fully understood. Here, we have combined proteomics approaches with the use of MLN4924, a generic inhibitor of SCF E3 ubiquitin ligases, to monitor changes in the cellular abundance of centriole duplication factors. We identified human STIL as a novel substrate of SCF-ßTrCP. The binding of ßTrCP depends on a DSG motif within STIL, and serine 395 within this motif is phosphorylated in vivo SCF-ßTrCP-mediated degradation of STIL occurs throughout interphase and mutations in the DSG motif causes massive centrosome amplification, attesting to the physiological importance of the pathway. We also uncover a connection between this new pathway and CDK2, whose role in centriole biogenesis remains poorly understood. We show that CDK2 activity protects STIL against SCF-ßTrCP-mediated degradation, indicating that CDK2 and SCF-ßTrCP cooperate via STIL to control centriole biogenesis.


Subject(s)
Centrioles/metabolism , Cyclin-Dependent Kinase 2/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Cyclopentanes/pharmacology , HEK293 Cells , Homeostasis , Humans , Interphase , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Phosphorylation , Proteolysis , Proteomics , Pyrimidines/pharmacology , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Serine/metabolism
9.
PLoS Genet ; 14(1): e1007165, 2018 01.
Article in English | MEDLINE | ID: mdl-29370161

ABSTRACT

Intellectual disability (ID), one of the most common human developmental disorders, can be caused by genetic mutations in Cullin 4B (Cul4B) and cereblon (CRBN). CRBN is a substrate receptor for the Cul4A/B-DDB1 ubiquitin ligase (CRL4) and can target voltage- and calcium-activated BK channel for ER retention. Here we report that ID-associated CRL4CRBN mutations abolish the interaction of the BK channel with CRL4, and redirect the BK channel to the SCFFbxo7 ubiquitin ligase for proteasomal degradation. Glioma cell lines harbouring CRBN mutations record density-dependent decrease of BK currents, which can be restored by blocking Cullin ubiquitin ligase activity. Importantly, mice with neuron-specific deletion of DDB1 or CRBN express reduced BK protein levels in the brain, and exhibit similar impairment in learning and memory, a deficit that can be partially rescued by activating the BK channel. Our results reveal a competitive targeting of the BK channel by two ubiquitin ligases to achieve exquisite control of its stability, and support changes in neuronal excitability as a common pathogenic mechanism underlying CRL4CRBN-associated ID.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Learning/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Proteolysis , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
10.
J Nutr Sci Vitaminol (Tokyo) ; 63(5): 339-348, 2017.
Article in English | MEDLINE | ID: mdl-29225319

ABSTRACT

Insulin resistance reduces insulin-induced muscle protein synthesis and accelerates muscle protein degradation. Ginseng ingestion has been reported to improve insulin resistance through the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. We hypothesized that panaxatriol (PT) derived from ginseng in combination with aerobic exercise (EX) may further promote protein synthesis and suppress protein degradation, and subsequently maintain muscle mass through the amelioration of insulin resistance. KKAy insulin-resistant mice were divided into control, panaxatriol only (PT), exercise only (EX), and EX+PT groups. EX and EX+PT ran on the treadmill for 45 min at 15 m/min 5 d/wk for 6 wk. PT and EX+PT groups were fed a standard diet containing 0.2% PT for 6 wk. Homeostasis model assessment for insulin resistance (HOMA-R) values was significantly improved after exercise for 6 wk. Moreover, EX+PT mice showed improved HOMA-R as compared to EX mice. p70S6K phosphorylation after a 4 h fast was significantly higher in EX than in the non-exercise control, and it was higher in EX+PT mice than in EX mice. Atrogin1 mRNA expression was significantly lower in EX than in the non-exercise control, and was significantly lowered further by PT treatment. EX and EX+PT mice showed higher soleus muscle mass and cross-sectional area (CSA) of the soleus myofibers than control animals, with higher values noted for both parameters in EX+PT than in EX. These results suggest that aerobic exercise and PT ingestion may contribute to maintain skeletal muscle mass through the amelioration of insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Dietary Supplements , Ginsenosides/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Muscular Atrophy/prevention & control , Physical Conditioning, Animal , Animals , Biomarkers/blood , Biomarkers/metabolism , Combined Modality Therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diet therapy , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Inulin/blood , Male , Mice, Mutant Strains , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/complications , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Panax/chemistry , Phosphorylation , Plant Roots/chemistry , Protein Processing, Post-Translational , Proteolysis , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism
11.
Nutrients ; 9(9)2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28930190

ABSTRACT

Stimulation of the ubiquitin-proteasome pathway-especially E3 ubiquitin ligases Atrogin-1 and MuRF1-is associated with muscle loss in diabetes. Elevated lipid metabolites impair myogenesis. Oligonol, a low molecular weight polyphenol derived from lychee, exhibited anti-diabetic and anti-obesity properties, suggesting it could be a proper supplement for attenuating muscle loss. Dietary (10 weeks) oligonol supplementation (20 or 200 mg/kg diet) on the skeletal muscle loss was investigated in diabetic db/db mice. Transcription factors NF-κB and FoxO3a involved in regulation of Atrogin-1 and MuRF1 were also investigated. Attenuation of muscle loss by oligonol (both doses) was associated with down-regulation of Atrogin-1 and MuRF1 gene expression. Oligonol supplementation decreased NF-κB expression in the nuclear fraction compared with db/db mice without oligonol supplement. Upregulation of sirtuin1 (SIRT1) expression prevented FoxO3a nuclear localization in db/db mice supplemented with oligonol. Marked increases in AMPKα activity and Ppara mRNA expression leading to lower lipid accumulation by oligonol provided additional benefits for attenuating muscle loss. Oligonol limited palmitate-induced senescent phenotype and cell cycle arrest and suppressed Atrogin-1 and MuRF1 mRNA expression in palmitate-treated C2C12 muscle cells, thus contributing to improving the impaired myotube formation. In conclusion, oligonol-mediated downregulation of Atrogin-1 and MuRF1 gene expression alleviates muscle loss and improves the impaired myotube formation, indicating that oligonol supplementation may be useful for the attenuation of myotube loss.


Subject(s)
Catechin/analogs & derivatives , Diabetes Mellitus, Experimental/physiopathology , Litchi/chemistry , Muscle Proteins/antagonists & inhibitors , Phenols/pharmacology , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Sarcopenia/drug therapy , Tripartite Motif Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Catechin/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Disease Models, Animal , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Fruit/chemistry , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Molecular Weight , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Polyphenols/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics
12.
Am J Physiol Endocrinol Metab ; 312(6): E495-E507, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28246104

ABSTRACT

Cushing's syndrome is caused by overproduction of the adrenocorticotropic hormone (ACTH), which stimulates the adrenal grand to make cortisol. Skeletal muscle wasting occurs in pathophysiological response to Cushing's syndrome. The forkhead box (FOX) protein family has been implicated as a key regulator of muscle loss under conditions such as diabetes and sepsis. However, the mechanistic role of the FOXO family in ACTH-induced muscle atrophy is not understood. We hypothesized that FOXO3a plays a role in muscle atrophy through expression of the E3 ubiquitin ligases, muscle RING finger protein-1 (MuRF-1), and atrogin-1 in Cushing's syndrome. For establishment of a Cushing's syndrome animal model, Sprague-Dawley rats were implanted with osmotic minipumps containing ACTH (40 ng·kg-1·day-1). ACTH infusion significantly reduced muscle weight. In ACTH-infused rats, MuRF-1, atrogin-1, and FOXO3a were upregulated and the FOXO3a promoter was targeted by the glucocorticoid receptor (GR). Transcriptional activity and expression of FOXO3a were significantly decreased by the GR antagonist RU486. Treatment with RU486 reduced MuRF-1 and atrogin-1 expression in accordance with reduced enrichment of FOXO3a and Pol II on the promoters. Knockdown of FOXO3a prevented dexamethasone-induced MuRF-1 and atrogin-1 expression. These results indicate that FOXO3a plays a role in muscle atrophy through expression of MuRF-1 and atrogin-1 in Cushing's syndrome.


Subject(s)
Cushing Syndrome/metabolism , Disease Models, Animal , Forkhead Box Protein O3/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Chromatin Immunoprecipitation , Cushing Syndrome/pathology , Cushing Syndrome/physiopathology , Forkhead Box Protein O3/agonists , Forkhead Box Protein O3/antagonists & inhibitors , Forkhead Box Protein O3/genetics , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Glucocorticoids/pharmacology , Hormone Antagonists/pharmacology , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Promoter Regions, Genetic/drug effects , RNA Interference , Rats, Sprague-Dawley , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Response Elements/drug effects , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins/agonists , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
13.
EMBO J ; 36(3): 260-273, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28007894

ABSTRACT

The F-box protein FBXW7 is the substrate-recruiting subunit of an SCF ubiquitin ligase and a major tumor-suppressor protein that is altered in several human malignancies. Loss of function of FBXW7 results in the stabilization of numerous proteins that orchestrate cell proliferation and survival. Little is known about proteins that directly regulate the function of this protein. In the current work, we have mapped the interactome of the enigmatic pseudophosphatase STYX We reasoned that a catalytically inactive phosphatase might have adopted novel mechanisms of action. The STYX interactome contained several F-box proteins, including FBXW7. We show that STYX binds to the F-box domain of FBXW7 and disables its recruitment into the SCF complex. Therefore, STYX acts as a direct inhibitor of FBXW7, affecting the cellular levels of its substrates. Furthermore, we find that levels of STYX and FBXW7 are anti-correlated in breast cancer patients, which affects disease prognosis. We propose the STYX-FBXW7 interaction as a promising drug target for future investigations.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , F-Box Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , F-Box-WD Repeat-Containing Protein 7 , Humans
14.
Appl Physiol Nutr Metab ; 41(12): 1240-1247, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27841025

ABSTRACT

l-Carnitine was recently found to downregulate the ubiquitin proteasome pathway (UPP) and increase insulin-like growth factor 1 concentrations in animal models. However, the effect of l-carnitine administration on disuse muscle atrophy induced by hindlimb suspension has not yet been studied. Thus, we hypothesized that l-carnitine may have a protective effect on muscle atrophy induced by hindlimb suspension via the Akt1/mTOR and/or UPP. Male Wistar rats were assigned to 3 groups: hindlimb suspension group, hindlimb suspension with l-carnitine administration (1250 mg·kg-1·day-1) group, and pair-fed group adjusted hindlimb suspension. l-Carnitine administration for 2 weeks of hindlimb suspension alleviated the decrease in weight and fiber size in the soleus muscle. In addition, l-carnitine suppressed atrogin-1 mRNA expression, which has been reported to play a pivotal role in muscle atrophy. The present study shows that l-carnitine has a protective effect against soleus muscle atrophy caused by hindlimb suspension and decreased E3 ligase messenger RNA expression, suggesting the possibility that l-carnitine protects against muscle atrophy, at least in part, through the inhibition of the UPP. These observations suggest that l-carnitine could serve as an effective supplement in the decrease of muscle atrophy caused by weightlessness in the fields of clinical and rehabilitative research.


Subject(s)
Carnitine/therapeutic use , Dietary Supplements , Enzyme Repression , Muscle Proteins/antagonists & inhibitors , Muscle, Skeletal/metabolism , Muscular Disorders, Atrophic/prevention & control , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Biomarkers/metabolism , Hindlimb Suspension/adverse effects , Immunohistochemistry , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/etiology , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/pathology , Proteasome Endopeptidase Complex , Proteasome Inhibitors/therapeutic use , Random Allocation , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Weightlessness/adverse effects
15.
PLoS Genet ; 12(10): e1006361, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27776126

ABSTRACT

SKN-1/Nrf are the primary antioxidant/detoxification response transcription factors in animals and they promote health and longevity in many contexts. SKN-1/Nrf are activated by a remarkably broad-range of natural and synthetic compounds and physiological conditions. Defining the signaling mechanisms that regulate SKN-1/Nrf activation provides insights into how cells coordinate responses to stress. Nrf2 in mammals is regulated in part by the redox sensor repressor protein named Keap1. In C. elegans, the p38 MAPK cascade in the intestine activates SKN-1 during oxidative stress by promoting its nuclear accumulation. Interestingly, we find variation in the kinetics of p38 MAPK activation and tissues with SKN-1 nuclear accumulation among different pro-oxidants that all trigger strong induction of SKN-1 target genes. Using genome-wide RNAi screening, we identify new genes that are required for activation of the core SKN-1 target gene gst-4 during exposure to the natural pro-oxidant juglone. Among 10 putative activators identified in this screen was skr-1/2, highly conserved homologs of yeast and mammalian Skp1, which function to assemble protein complexes. Silencing of skr-1/2 inhibits induction of SKN-1 dependent detoxification genes and reduces resistance to pro-oxidants without decreasing p38 MAPK activation. Global transcriptomics revealed strong correlation between genes that are regulated by SKR-1/2 and SKN-1 indicating a high degree of specificity. We also show that SKR-1/2 functions upstream of the WD40 repeat protein WDR-23, which binds to and inhibits SKN-1. Together, these results identify a novel p38 MAPK independent signaling mechanism that activates SKN-1 via SKR-1/2 and involves WDR-23.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Inactivation, Metabolic/genetics , Longevity/genetics , SKP Cullin F-Box Protein Ligases/genetics , Activin Receptors, Type I/genetics , Animals , Antioxidants/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/biosynthesis , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental , Humans , Kelch-Like ECH-Associated Protein 1/biosynthesis , Kelch-Like ECH-Associated Protein 1/genetics , Phosphorylation , RNA Interference , Reactive Oxygen Species/metabolism , S-Phase Kinase-Associated Proteins/genetics , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Acta Biochim Biophys Sin (Shanghai) ; 48(9): 827-32, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27521792

ABSTRACT

Control of cardiac muscle mass is thought to be determined by a dynamic balance of protein synthesis and degradation. Recent studies have demonstrated that atrophy-related forkhead box O 3a (FOXO3a)/muscle atrophy F-box (MAFbx) signaling pathway plays a central role in the modulation of proteolysis and exert inhibitory effect on cardiomyocyte hypertrophy. In this study, we tested the hypothesis that adenosine monophosphate-activated protein kinase (AMPK) activation attenuates cardiomyocyte hypertrophy by regulating FOXO3a/MAFbx signaling pathway and its downstream protein degradation. The results showed that activation of AMPK with 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) attenuated cardiomyocyte hypertrophy induced by angiotensin II (Ang II). The antihypertrophic effects of AICAR were blunted by AMPK inhibitor Compound C. In addition, AMPK dramatically increased the activity of transcription factor FOXO3a, up-regulated the expression of its downstream ubiquitin ligase MAFbx, and enhanced cardiomyocyte proteolysis. Meanwhile, the effects of AMPK on protein degradation and cardiomyocyte hypertrophy were blocked after MAFbx was silenced by transfection of cardiomyocytes with MAFbx-siRNA. These results indicate that AMPK plays an important role in the inhibition of cardiomyocyte hypertrophy by activating protein degradation via FOXO3a/MAFbx signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Forkhead Box Protein O3/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Enlargement/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Myocytes, Cardiac/drug effects , Proteolysis/drug effects , RNA, Small Interfering/genetics , Rats , Ribonucleotides/pharmacology , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics , Signal Transduction/drug effects
17.
Pharmacol Res ; 111: 34-42, 2016 09.
Article in English | MEDLINE | ID: mdl-27238229

ABSTRACT

In the last decade, the ubiquitin proteasome system (UPS), in general, and E3 ubiquitin ligases, in particular, have emerged as valid drug targets for the development of novel anti-cancer therapeutics. Cullin RING Ligases (CRLs), which can be classified into eight groups (CRL1-8) and comprise approximately 200 members, represent the largest family of E3 ubiquitin ligases which facilitate the ubiquitination-derived proteasomal degradation of a myriad of functionally and structurally diverse substrates. S phase kinase-associated protein 1 (Skp1)-Cullin1-F-Box protein (SCF) complexes are the best characterized among CRLs, which play crucial roles in numerous cellular processes and physiological dysfunctions, such as in cancer biology. Currently, there is growing interest in developing SCF-targeting anti-cancer therapies for clinical application. Indeed, the research in this field has seen some progress in the form of cullin neddylation- and Skp2-inhibitors. However, it still remains an underdeveloped area and needs to design new strategies for developing improved form of therapy. In this review, we venture a novel strategy that rational pharmacological targeting of Skp1, a central regulator of SCF complexes, may provide a novel avenue for SCF-oriented anti-cancer therapy, expected: (i) to simultaneously address the critical roles that multiple SCF oncogenic complexes play in cancer biology, (ii) to selectively target cancer cells with minimal normal cell toxicity, and (iii) to offer multiple chemical series, via therapeutic interventions at the Skp1 binding interfaces in SCF complex, thereby maximizing chances of success for drug discovery. In addition, we also discuss the challenges that might be posed regarding rational pharmacological interventions against Skp1.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Discovery/methods , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Animals , Cloning, Molecular , Drug Design , Humans , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Conformation , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
18.
Biosci Biotechnol Biochem ; 80(8): 1499-504, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27055887

ABSTRACT

To investigate the intracellular signaling mechanisms by which clenbuterol reduces muscle protein degradation, we examined the phosphorylation level and intracellular localization of FOXO1 in the sartorius muscle of neonatal chicks. One-day-old chicks were given a single intraperitoneal injection of clenbuterol (0.1 mg/kg body weight). Three hours after injection, AKT protein was phosphorylated in the sartorius muscle by clenbuterol injection. Coincidentally, clenbuterol increased cytosolic level of phosphorylated FOXO1 protein, while it decreased nuclear level of FOXO1 protein in the sartorius muscle. Furthermore, clenbuterol decreased the expression of mRNAs for muscle-specific ubiquitin ligases (atrogin-1/MAFbx and MuRF1) in the sartorius muscle accompanied by decreased plasma 3-methylhistidine concentration, an index of muscle protein degradation, at 3 h after injection. These results suggested that, in the sartorius muscle of the chicks, clenbuterol changed the intracellular localization of phosphorylated FOXO1, and consequently decreased protein degradation via suppressing the expression of genes encoding muscle-specific ubiquitin ligases.


Subject(s)
Avian Proteins/genetics , Clenbuterol/pharmacology , Forkhead Box Protein O1/genetics , Muscle, Skeletal/drug effects , SKP Cullin F-Box Protein Ligases/genetics , Sympathomimetics/pharmacology , Ubiquitin-Protein Ligases/genetics , Animals , Animals, Newborn , Avian Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chickens , Cytoplasm/drug effects , Cytoplasm/metabolism , Forkhead Box Protein O1/metabolism , Gene Expression Regulation , Injections, Intraperitoneal , Methylhistidines/blood , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism
19.
Proc Natl Acad Sci U S A ; 113(13): 3527-32, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976582

ABSTRACT

Skp1-Cul1-F-box (SCF) E3 ligases play key roles in multiple cellular processes through ubiquitination and subsequent degradation of substrate proteins. Although Skp1 and Cul1 are invariant components of all SCF complexes, the 69 different human F-box proteins are variable substrate binding modules that determine specificity. SCF E3 ligases are activated in many cancers and inhibitors could have therapeutic potential. Here, we used phage display to develop specific ubiquitin-based inhibitors against two F-box proteins, Fbw7 and Fbw11. Unexpectedly, the ubiquitin variants bind at the interface of Skp1 and F-box proteins and inhibit ligase activity by preventing Cul1 binding to the same surface. Using structure-based design and phage display, we modified the initial inhibitors to generate broad-spectrum inhibitors that targeted many SCF ligases, or conversely, a highly specific inhibitor that discriminated between even the close homologs Fbw11 and Fbw1. We propose that most F-box proteins can be targeted by this approach for basic research and for potential cancer therapies.


Subject(s)
Cullin Proteins/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitins/pharmacology , Amino Acid Sequence , Binding Sites , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cullin Proteins/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/chemistry , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Genetic Variation , Humans , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Engineering , Protein Interaction Domains and Motifs , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/genetics , Sequence Homology, Amino Acid , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitins/chemistry , Ubiquitins/genetics , beta-Transducin Repeat-Containing Proteins/antagonists & inhibitors , beta-Transducin Repeat-Containing Proteins/chemistry , beta-Transducin Repeat-Containing Proteins/genetics
20.
PLoS One ; 10(8): e0135591, 2015.
Article in English | MEDLINE | ID: mdl-26295339

ABSTRACT

Alpha-1 antitrypsin deficiency is the leading cause of childhood liver failure and one of the most common lethal genetic diseases. The disease-causing mutant A1AT-Z fails to fold correctly and accumulates in the endoplasmic reticulum (ER) of the liver, resulting in hepatic fibrosis and hepatocellular carcinoma in a subset of patients. Furthermore, A1AT-Z sequestration in hepatocytes leads to a reduction in A1AT secretion into the serum, causing panacinar emphysema in adults. The purpose of this work was to elucidate the details by which A1AT-Z is degraded in hepatic cell lines. We identified the ubiquitin ligase FBG1, which has been previously shown to degrade proteins by both the ubiquitin proteasome pathway and autophagy, as being key to A1AT-Z degradation. Using chemical and genetic approaches we show that FBG1 degrades A1AT-Z through both the ubiquitin proteasome system and autophagy. Overexpression of FBG1 decreases the half-life of A1AT-Z and knocking down FBG1 in a hepatic cell line, and in mice results in an increase in ATAT. Finally, we show that FBG1 degrades A1AT-Z through a Beclin1-dependent arm of autophagy. In our model, FBG1 acts as a safety ubiquitin ligase, whose function is to re-ubiquitinate ER proteins that have previously undergone de-ubiquitination to ensure they are degraded.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , SKP Cullin F-Box Protein Ligases/genetics , alpha 1-Antitrypsin Deficiency/genetics , alpha 1-Antitrypsin/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line, Tumor , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Female , Gene Expression Regulation , Half-Life , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Protein Binding , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Ubiquitination , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin Deficiency/metabolism , alpha 1-Antitrypsin Deficiency/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...