Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Mol Cell ; 68(1): 185-197.e6, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28943315

ABSTRACT

Many infections and stress signals can rapidly activate the NLRP3 inflammasome to elicit robust inflammatory responses. This activation requires a priming step, which is thought to be mainly for upregulating NLRP3 transcription. However, recent studies report that the NLRP3 inflammasome can be activated independently of transcription, suggesting that the priming process has unknown essential regulatory steps. Here, we report that JNK1-mediated NLRP3 phosphorylation at S194 is a critical priming event and is essential for NLRP3 inflammasome activation. We show that NLRP3 inflammasome activation is disrupted in NLRP3-S194A knockin mice. JNK1-mediated NLRP3 S194 phosphorylation is critical for NLRP3 deubiquitination and facilitates its self-association and the subsequent inflammasome assembly. Importantly, we demonstrate that blocking S194 phosphorylation prevents NLRP3 inflammasome activation in cryopyrin-associated periodic syndromes (CAPS). Thus, our study reveals a key priming molecular event that is a prerequisite for NLRP3 inflammasome activation. Inhibiting NLRP3 phosphorylation could be an effective treatment for NLRP3-related diseases.


Subject(s)
Inflammasomes/genetics , Macrophages/immunology , Mitogen-Activated Protein Kinase 8/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Shock, Septic/genetics , Amino Acid Sequence , Animals , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/immunology , Escherichia coli/chemistry , Female , Gene Expression Regulation , HEK293 Cells , Humans , Inflammasomes/immunology , Lipopolysaccharides/pharmacology , Macrophages/pathology , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Phosphorylation , Sequence Alignment , Sequence Homology, Amino Acid , Shock, Septic/chemically induced , Shock, Septic/mortality , Shock, Septic/pathology , Signal Transduction , Survival Analysis
2.
J Cell Mol Med ; 24(7): 4298-4311, 2020 04.
Article in English | MEDLINE | ID: mdl-32108992

ABSTRACT

Multidrug resistance is one of the reasons for low survival of advanced hepatocellular carcinoma (HCC). Our previous studies indicate that the hedgehog signalling is involved in hepatic carcinogenesis, metastasis and chemo-resistance. The present study aims to uncover molecular mechanisms underlying hepatoma chemo-resistance. TAP1 and GLI1/2 gene expression was assessed in both poorly differentiated hepatoma cells and HCC specimens. Potential GLI-binding site in the TAP1 promoter sequence was validated by molecular assays. Approximately 75% HCC specimens exhibited an elevated expression of hedgehog GLI1 transcription factor compared with adjacent liver tissue. Both GLI1/2 and TAP1 protein levels were significantly elevated in poorly differentiated hepatoma cells. Both Huh-7-trans and Huh-7-DN displayed more karyotypic abnormalities and differential gene expression profiles than their native Huh-7 cells. Sensitivity to Sorafenib, doxorubicin and cisplatin was remarkably improved after either GLI1 or TAP1 gene was inhibited by an RNAi approach or by a specific GLI1/2 inhibitor, GANT61. Further experiments confirmed that hedgehog transcription factor GLI1/2 binds to the TAP1 promoter, indicating that TAP1 is one of GLI1/2 target genes. In conclusion, TAP1 is under direct transcriptional control of the hedgehog signalling. Targeting hedgehog signalling confers a novel insight into alleviating drug resistance in the treatment of refractory HCC.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein Gli2/antagonists & inhibitors , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/genetics
3.
Nat Immunol ; 9(5): 533-41, 2008 May.
Article in English | MEDLINE | ID: mdl-18362886

ABSTRACT

Despite rapid progress in elucidating the molecular mechanisms of activation of the kinase IKK, the processes that regulate IKK deactivation are still unknown. Here we demonstrate that CUE domain-containing 2 (CUEDC2) interacted with IKKalpha and IKKbeta and repressed activation of the transcription factor NF-kappaB by decreasing phosphorylation and activation of IKK. Notably, CUEDC2 also interacted with GADD34, a regulatory subunit of protein phosphatase 1 (PP1). We found that IKK, CUEDC2 and PP1 existed in a complex and that IKK was released from the complex in response to inflammatory stimuli such as tumor necrosis factor. CUEDC2 deactivated IKK by recruiting PP1 to the complex. Therefore, CUEDC2 acts as an adaptor protein to target IKK for dephosphorylation and inactivation by recruiting PP1.


Subject(s)
Carrier Proteins/metabolism , I-kappa B Kinase/metabolism , Membrane Proteins/metabolism , Protein Phosphatase 1/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Carrier Proteins/immunology , Catalytic Domain , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Female , Humans , I-kappa B Kinase/chemistry , Inflammation/immunology , Interleukin-6/biosynthesis , Interleukin-6/genetics , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Protein Binding , Repressor Proteins/immunology , Up-Regulation
4.
Med Teach ; 41(5): 603-605, 2019 05.
Article in English | MEDLINE | ID: mdl-30462549

ABSTRACT

Objective: To reflectively look at the present methods by which the clinical competence of 5th-year medical students (i.e. interns) in Sun Yat-Sen University (SYSU) are assessed upon finishing internship rotation in internal medicine (IM). Methods: Current procedures for the competence assessment of end-of-rotation IM interns in the First Affiliated Hospital of SYSU were reviewed, along with a point-by-point appraisal based on the PROFILE approach to structured assessment, and, whenever possible, suggestions for future improvement. Results and discussions: On a scale of 1-10, with 10 being the best or the most ideal, our marks for current methods to assess end-of-rotation IM interns in terms of being Programmatic, Real-World, Outcome-based, Formative, Impactful, Learner-engaged, and Evaluation-guaranteed were 7, 9, 3, 4, 6, 8, and 1, respectively. The strengths, weaknesses as well as potential solutions in each of the seven aspects are also discussed separately. Conclusions: Current assessment program for IM internship is strong in being programmatic, real-world, educationally impactful and learner engaged, and has room for further improvement in its time-based arrangements, relative shortage of feedback provision, as well as a systematic lack of quality control measures.


Subject(s)
Clinical Competence , Educational Measurement/methods , Internal Medicine/education , Internship and Residency , Humans , Problem-Based Learning , Program Evaluation , Schools, Medical , Students, Medical
5.
J Biol Chem ; 290(16): 10395-405, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25681446

ABSTRACT

Excessive nuclear factor κB (NF-κB) activation should be precisely controlled as it contributes to multiple immune and inflammatory diseases. However, the negative regulatory mechanisms of NF-κB activation still need to be elucidated. Various types of polyubiquitin chains have proved to be involved in the process of NF-κB activation. Many negative regulators linked to ubiquitination, such as A20 and CYLD, inhibit IκB kinase activation in the NF-κB signaling pathway. To find new NF-κB signaling regulators linked to ubiquitination, we used a small scale siRNA library against 51 ubiquitin-associated domain-containing proteins and screened out UBXN1, which contained both ubiquitin-associated and ubiquitin regulatory X (UBX) domains as a negative regulator of TNFα-triggered NF-κB activation. Overexpression of UBXN1 inhibited TNFα-triggered NF-κB activation, although knockdown of UBXN1 had the opposite effect. UBX domain-containing proteins usually act as valosin-containing protein (VCP)/p97 cofactors. However, knockdown of VCP/p97 barely affected UBXN1-mediated NF-κB inhibition. At the same time, we found that UBXN1 interacted with cellular inhibitors of apoptosis proteins (cIAPs), E3 ubiquitin ligases of RIP1 in the TNFα receptor complex. UBXN1 competitively bound to cIAP1, blocked cIAP1 recruitment to TNFR1, and sequentially inhibited RIP1 polyubiquitination in response to TNFα. Therefore, our findings demonstrate that UBXN1 is an important negative regulator of the TNFα-triggered NF-κB signaling pathway by mediating cIAP recruitment independent of VCP/p97.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation , NF-kappa B/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Genes, Reporter , HEK293 Cells , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Proteolysis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Small Molecule Libraries , Tumor Necrosis Factor-alpha/pharmacology , Valosin Containing Protein
6.
Proc Natl Acad Sci U S A ; 110(27): 11017-22, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776205

ABSTRACT

DNA damage triggers cell cycle arrest to provide a time window for DNA repair. Failure of arrest could lead to genomic instability and tumorigenesis. DNA damage-induced G1 arrest is generally achieved by the accumulation of Cyclin-dependent kinase inhibitor 1 (p21). However, p21 is degraded and does not play a role in UV-induced G1 arrest. The mechanism of UV-induced G1 arrest thus remains elusive. Here, we have identified a critical role for CUE domain-containing protein 2 (CUEDC2) in this process. CUEDC2 binds to and inhibits anaphase-promoting complex/cyclosome-Cdh1 (APC/C(Cdh1)), a critical ubiquitin ligase in G1 phase, thereby stabilizing Cyclin A and promoting G1-S transition. In response to UV irradiation, CUEDC2 undergoes ERK1/2-dependent phosphorylation and ubiquitin-dependent degradation, leading to APC/C(Cdh1)-mediated Cyclin A destruction, Cyclin-dependent kinase 2 inactivation, and G1 arrest. A nonphosphorylatable CUEDC2 mutant is resistant to UV-induced degradation. Expression of this stable mutant effectively overrides UV-induced G1-S block. These results establish CUEDC2 as an APC/C(Cdh1) inhibitor and indicate that regulated CUEDC2 degradation is critical for UV-induced G1 arrest.


Subject(s)
Carrier Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Membrane Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Adaptor Proteins, Signal Transducing , Anaphase-Promoting Complex-Cyclosome , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Tumor , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , G1 Phase Cell Cycle Checkpoints/radiation effects , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Phosphorylation/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ultraviolet Rays
7.
Cochrane Database Syst Rev ; (10): CD007744, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25340915

ABSTRACT

BACKGROUND: Nonsteroidal anti-inflammatory drugs (NSAIDs) are used to reduce inflammatory pain and swelling in inflammatory bowel disease (IBD) patients with rheumatological manifestations. While these drugs effectively reduce musculoskeletal pain and stiffness, long-term use is limited by gastrointestinal (GI) adverse effects (AEs) and disease exacerbation. As an alternative to NSAIDs, selective cyclooxygenase 2 (COX-2) inhibitors were developed to improve GI safety and tolerability. COX-2 inhibitors include drugs such as celecoxib, rofecoxib, valdecoxib, etoricoxib, and lumiracoxib. Rofecoxib and valdecoxib have been withdrawn from the market worldwide due to safety concerns (most importantly for cardiovascular adverse events) and lumiracoxib has been withdrawn in many countries due to liver toxicity. However, celecoxib and etoricoxib continue to be available for use in many countries. Several studies have examined whether COX-2 inhibitors can be safely used for the treatment of rheumatological manifestations of IBD with inconsistent results. Some investigators report acceptable safety profiles associated with these drugs while others found that COX-2 inhibitors are associated with high rates of disease exacerbation. OBJECTIVES: The objective of this systematic review was to evaluate the tolerability and safety of COX-2 inhibitors used for the treatment of rheumatological manifestations of IBD. SEARCH METHODS: We searched the following databases from inception to 19 September 2013: PubMed, EMBASE, MEDLINE and CENTRAL. The search was not limited by language. Additional trials were identified by manually searching the reference lists of relevant papers and conference proceedings and through correspondence with experts and pharmaceutical companies. SELECTION CRITERIA: Randomized controlled trials (RCTs) that compared COX-2 inhibitors to placebo were considered for inclusion. Participants were adult patients with IBD presenting with rheumatological manifestations of at least two weeks duration. DATA COLLECTION AND ANALYSIS: Two authors independently assessed trial eligibility and extracted data. Methodological quality was assessed using the Cochrane risk of bias tool. The primary outcome measure was the proportion of patients with disease exacerbation as defined by the included studies. Secondary outcomes included GI adverse effects, renal toxicity, cardiovascular and thrombotic events. Data were analysed on an intention-to-treat basis where patients with missing final outcomes were assumed to have had an exacerbation of IBD. We calculated the risk ratio (RR) and corresponding 95% confidence interval (95% CI) for dichotomous outcomes. The overall quality of the evidence was assessed using the GRADE criteria. MAIN RESULTS: There were no RCTs that assessed the tolerability or safety of the withdrawn COX-2 inhibitors rofecoxib, valdecoxib, or lumiracoxib. Two RCTs (n = 381 IBD patients with rheumatological manifestations) were included in the review. One study (n = 159) compared etoricoxib (60 to 120 mg/day) to placebo in IBD patients with quiescent or active ulcerative colitis or Crohn's disease. The other study (n = 222) compared celecoxib (200 mg twice daily) to placebo in patients with quiescent ulcerative colitis. Both studies were judged to be at low risk of bias. The two included studies were not pooled for meta-analysis due to differences in patient populations and treatment duration. There was no statistically significant difference in exacerbation of IBD between etoricoxib and placebo. After 12 weeks of treatment the IBD exacerbation rate was 17% (14/82) in the etoricoxib group compared to 19% (15/77) in the placebo group (RR 0.88, 95% CI 0.45 to 1.69). A GRADE analysis indicated that the overall quality of the evidence supporting this outcome was low due to very sparse data (29 events). There was no statistically significant difference in exacerbation of ulcerative colitis between celecoxib and placebo. After two weeks of treatment 4% (5/112) of celecoxib patients experienced an exacerbation of ulcerative colitis compared to 6% (7/110) of patients in the placebo group (RR 0.70, 95% CI 0.23 to 2.14). A GRADE analysis indicated that the overall quality of the evidence supporting this outcome was low due to very sparse data (12 events). The study comparing etoricoxib to placebo documented but did not report on AEs. The proportion of patients who experienced AEs was similar in the celecoxib and placebo groups (21% and 17%, respectively, P > 0.20). No patients in either group died or experienced serious adverse events. Eleven percent of patients in the celecoxib and placebo groups experienced GI AEs (RR 0.97, 95% CI 0.46 to 2.07). A GRADE analysis indicated that the overall quality of the evidence supporting this outcome was low due to very sparse data (24 events). GI AEs led to premature withdrawal from the study in 3% of patients in celecoxib and placebo groups respectively. GI AEs included increased stool frequency, rectal bleeding, and inflamed mucosa. No patients experienced any cardiovascular adverse events. Renal toxicity or thrombotic AEs were not reported. AUTHORS' CONCLUSIONS: The results for disease exacerbation and AEs between the COX-2 inhibitors celecoxib and etoricoxib and placebo were uncertain. Thus no definitive conclusions regarding the tolerability and safety of the short term use of celecoxib and etoricoxib in patients with IBD can be drawn. The two included studies suggest that celecoxib and etoricoxib do not exacerbate IBD symptoms. However, it should be noted that both studies had relatively small sample sizes and short follow-up durations. Clinicians need to continue to weigh the risks and benefits of these drugs when treating patients IBD patients with rheumatological manifestations in order to avoid disease exacerbation and other adverse effects. Further RCTs are needed to determine the tolerability and safety of celecoxib and etoricoxib in these patients.


Subject(s)
Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Cyclooxygenase 2 Inhibitors/adverse effects , Pyrazoles/adverse effects , Pyridines/adverse effects , Sulfonamides/adverse effects , Sulfones/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Celecoxib , Colitis, Ulcerative/complications , Crohn Disease/complications , Diclofenac/adverse effects , Diclofenac/analogs & derivatives , Etoricoxib , Humans , Isoxazoles/adverse effects , Lactones/adverse effects , Randomized Controlled Trials as Topic , Safety-Based Drug Withdrawals
8.
BMC Complement Altern Med ; 14: 137, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24739144

ABSTRACT

BACKGROUND: Electroacupuncture (EA) stimulation has been shown to have a great therapeutic potential for treating gastrointestinal motility disorders. However, no evidence has clarified the mechanisms contributing to the effects of EA stimulation at the Zusanli acupoint (ST.36). This study was designed to investigate the regulative effect of EA stimulation at the ST.36 on gastric motility and to explore its possible mechanisms. METHODS: Thirty Sprague-Dawley rats were randomly divided into three groups: the ST.36 group, the non-acupoint group, and the control group. EA stimulation was set at 2 Hz, continuous mode, and 1 V for 30 min. The frequency and average peak amplitude of gastric motility were measured by electrogastrography. The protein kinase C (PKC) and mitogen-activated protein kinase (MAPK) signaling pathways were assessed using real-time polymerase chain reactions. Caldesmon (CaD) and calponin (CaP) protein expression in the gastric antrum were detected on Western blots. A Computed Video Processing System was used to evaluate morphological changes in smooth muscle cells (SMCs) from the gastric antrum. RESULTS: EA stimulation at ST.36 had a dual effect on the frequency and average peak amplitude. Additionally, EA stimulation at ST.36 regulated the expression of some genes in the PKC and MAPK signaling pathways, and it regulated the expression of the CaD and CaP proteins. EA serum induced SMC contractility. Promotion of gastric motility may correlate with up-regulation of MAPK6 (ERK3), MAPK13, and Prostaglandin-endoperoxide synthase 2 (PTGS2) gene expression, and the down-regulation of the collagen, type I, alpha 1 (COL1A1) gene and CaD and CaP protein expression. Inhibition of gastric motility may correlate with down-regulation of the Interleukin-1 receptor type 2 (IL1R2) and Matrix metalloproteinase-9 (MMP9) genes, and up-regulation of CaD and CaP protein expression. CONCLUSIONS: EA stimulation at ST.36 regulated gastric motility, and the effects were both promoting and inhibiting in rats. The possible mechanisms may correlate with the PKC and MAPK signal transduction pathways.


Subject(s)
Acupuncture Points , Electroacupuncture , Gastrointestinal Motility , Gastrointestinal Tract/enzymology , MAP Kinase Signaling System , Protein Kinase C/metabolism , Animals , Gene Expression , Male , Protein Kinase C/genetics , Rats , Rats, Sprague-Dawley
9.
PeerJ Comput Sci ; 10: e2167, 2024.
Article in English | MEDLINE | ID: mdl-38983239

ABSTRACT

Adaptive gradient algorithms have been successfully used in deep learning. Previous work reveals that adaptive gradient algorithms mainly borrow the moving average idea of heavy ball acceleration to estimate the first- and second-order moments of the gradient for accelerating convergence. However, Nesterov acceleration which uses the gradient at extrapolation point can achieve a faster convergence speed than heavy ball acceleration in theory. In this article, a new optimization algorithm which combines adaptive gradient algorithm with Nesterov acceleration by using a look-ahead scheme, called NALA, is proposed for deep learning. NALA iteratively updates two sets of weights, i.e., the 'fast weights' in its inner loop and the 'slow weights' in its outer loop. Concretely, NALA first updates the fast weights k times using Adam optimizer in the inner loop, and then updates the slow weights once in the direction of Nesterov's Accelerated Gradient (NAG) in the outer loop. We compare NALA with several popular optimization algorithms on a range of image classification tasks on public datasets. The experimental results show that NALA can achieve faster convergence and higher accuracy than other popular optimization algorithms.

10.
Cancer Lett ; 588: 216768, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38453045

ABSTRACT

Hedgehog signaling is activated in response to liver injury, and modulates organogenesis. However, the role of non-canonical hedgehog activation via TGF-ß1/SMAD3 in hepatic carcinogenesis is poorly understood. TGF-ß1/SMAD3-mediated non-canonical activation was found in approximately half of GLI2-positive hepatocellular carcinoma (HCC), and two new GLI2 isoforms with transactivating activity were identified. Phospho-SMAD3 interacted with active GLI2 isoforms to transactivate downstream genes in modulation of stemness, epithelial-mesenchymal transition, chemo-resistance and metastasis in poorly-differentiated hepatoma cells. Non-canonical activation of hedgehog signaling was confirmed in a transgenic HBV-associated HCC mouse model. Inhibition of TGF-ß/SMAD3 signaling reduced lung metastasis in a mouse in situ hepatic xenograft model. In another cohort of 55 HCC patients, subjects with high GLI2 expression had a shorter disease-free survival than those with low expression. Moreover, co-positivity of GLI2 with SMAD3 was observed in 87.5% of relapsed HCC patients with high GLI2 expression, indicating an increased risk of post-resection recurrence of HCC. The findings underscore that suppressing the non-canonical hedgehog signaling pathway may confer a potential strategy in the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Carcinoma, Hepatocellular/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Liver Neoplasms/pathology , Mice, Transgenic , Nuclear Proteins/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
11.
J Biol Chem ; 287(1): 382-392, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22084247

ABSTRACT

Janus kinase 1/signal transducers and activators of transcription 3 (JAK1/STAT3) pathway is one of the recognized oncogenic signaling pathways that frequently overactivated in a variety of human tumors. Despite rapid progress in elucidating the molecular mechanisms of activation of JAK/STAT pathway, the processes that regulate JAK/STAT deactivation need to be further clarified. Here we demonstrate that CUE domain-containing 2 (CUEDC2) inhibits cytokine-induced phosphorylation of JAK1 and STAT3 and the subsequent STAT3 transcriptional activity. Further analysis by a yeast two-hybrid assay showed that CUEDC2 could engage in a specific interaction with a key JAK/STAT inhibitor, SOCS3 (suppressors of cytokine signaling 3). The interaction between CUEDC2 and SOCS3 is required for the inhibitory effect of CUEDC2 on JAK1 and STAT3 activity. Additionally, we found CUEDC2 functions collaboratively with SOCS3 to inhibit JAK1/STAT3 signaling by increasing SOCS3 stability via enhancing its association with Elongin C. Therefore, our findings revealed a new biological activity for CUEDC2 as the regulator of JAK1/STAT3 signaling and paved the way to a better understanding of the mechanisms by which SOCS3 has been linked to suppression of the JAK/STAT pathway.


Subject(s)
Carrier Proteins/metabolism , Janus Kinase 1/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing , Cell Line , Elongin , Enzyme Activation , Humans , Phosphorylation , Protein Stability , Proteolysis , Suppressor of Cytokine Signaling 3 Protein , Transcription Factors/metabolism , Transcription, Genetic
12.
Biochem Biophys Res Commun ; 431(4): 686-92, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23357418

ABSTRACT

RIG-I/MDA5 plays a pivotal role in innate immunity by detecting intracellular double-stranded RNA (dsRNA) and activating the transcription of type I interferons and proinflammatory factors, but the exactly regulating mechanism of RIG-I/MDA5 signaling remains elusive. In this study, UbL-UBA domain containing protein RAD23A was identified as a negative regulator of RIG-I/MDA5-mediated signaling activation through a small interfering RNA (siRNA)-based screening. Knockdown of RAD23A augmented the expression of RIG-I/MDA5-mediated expression of proinflammatory cytokines and IFN-ß whereas ectopic expression of RAD23A showed the converse effect. Moreover, we confirmed the interaction between RAD23A and tumor necrosis factor receptor-associated factor 2 (TRAF2), an essential mediator of RIG-I/MDA5 signaling, and found that RAD23A down-regulated TRAF2 protein level through ubiquitin-proteasome system. Therefore, this study identified RAD23A as a novel negative regulator of RIG-I/MDA5 mediated anti-virus response.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , TNF Receptor-Associated Factor 2/metabolism , Ubiquitination , DEAD Box Protein 58 , DNA Repair Enzymes/genetics , DNA, Viral/immunology , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon-Induced Helicase, IFIH1 , Interferon-beta/biosynthesis , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Proteolysis , RNA, Small Interfering/genetics , RNA, Viral/immunology , Receptors, Immunologic , Signal Transduction , Ubiquitin/metabolism
13.
Pak J Med Sci ; 29(4): 1065-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24353690

ABSTRACT

Rosai-Dorfman disease (RDD) is rare and characterized by histiocytic proliferation and massive cervical lymphadenopathy. About 40% of patients have extra-nodal involvement. Opthalmic involvement is seen in 10% of cases. A case of orbital Rosai Dorfman disease in a 58 years old woman is presented here, who was misdiagnosed as orbital inflammatory disease initially. The patient did not respond to a course of oral prednisolone. Then complete surgical excision of the mass was performed and the histopathological examination was consistent with a diagnosis of RDD.

14.
J Cancer ; 14(2): 200-218, 2023.
Article in English | MEDLINE | ID: mdl-36741265

ABSTRACT

Eyes absent homologue 4 (EYA4) is silenced in pancreatic ductal adenocarcinoma (PDAC) and functions as a tumor suppressor to restrain PDAC development, albeit the molecular mechanism underlying its downregulation remains enigmatic. Methods: Functional studies were determined by immunohistochemistry of PDAC samples from patients and Pdx1-Cre; LSL-KrasG12D/+; Trp53fl/+ (KPC) mice, three-dimensional spheroid culture, flow cytometry, MTT and subcutaneous xenograft experiments. Mechanistical studies were examined by cellular ubiquitination, cycloheximide (CHX) pulse-chase, co-immunoprecipitation, chromatin immunoprecipitation, GST-pulldown, in vitro protein kinase assay, immunofluorescence and luciferase reporter assays. Results: We screen E3 ligase that is negatively correlated with EYA4 and uncover a mutually exclusive interaction of tripartite motif containing 69 (TRIM69) with EYA4 in human PDAC. TRIM69 elicits EYA4 polyubiquitylation and turnover independent of P53 and impedes the EYA4-driven deactivation of ß-catenin/ID2 cascade, fueling PDAC cell proliferation in vitro and tumor development in mice. Expression of TRIM69 is upregulated in PDAC samples from independent cohorts of patients and the Pdx1-Cre; LSL-KrasG12D/+; Trp53fl/+ (KPC) mice, and associated with unfavorable prognosis. Depleting TRIM69 preferentially induces lethality in the EYA4-deficient PDAC cells. We further unearth that ERK2 directly binds to the D-site of mitogen-activated protein kinase (MAPK) docking groove in EYA4 Leu512/514 and phosphorylates EYA4 at Ser37, which is instrumental for EYA4 polyubiquitylation and turnover by TRIM69. Conclusion: Our results define a previously unappreciated role of TRIM69-EYA4 axis in pancreatic tumorigenesis and underscore that targeting TRIM69 might be an effective therapeutic approach for PDAC harboring EYA4 deficiency.

15.
Science ; 380(6648): 972-979, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37262147

ABSTRACT

The suprachiasmatic nucleus (SCN) drives circadian clock coherence through intercellular coupling, which is resistant to environmental perturbations. We report that primary cilia are required for intercellular coupling among SCN neurons to maintain the robustness of the internal clock in mice. Cilia in neuromedin S-producing (NMS) neurons exhibit pronounced circadian rhythmicity in abundance and length. Genetic ablation of ciliogenesis in NMS neurons enabled a rapid phase shift of the internal clock under jet-lag conditions. The circadian rhythms of individual neurons in cilia-deficient SCN slices lost their coherence after external perturbations. Rhythmic cilia changes drive oscillations of Sonic Hedgehog (Shh) signaling and clock gene expression. Inactivation of Shh signaling in NMS neurons phenocopied the effects of cilia ablation. Thus, cilia-Shh signaling in the SCN aids intercellular coupling.


Subject(s)
Cilia , Circadian Clocks , Circadian Rhythm , Hedgehog Proteins , Suprachiasmatic Nucleus Neurons , Animals , Mice , Cilia/metabolism , Cilia/physiology , Circadian Clocks/genetics , Circadian Rhythm/physiology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Suprachiasmatic Nucleus Neurons/physiology , Signal Transduction , Gene Expression Regulation , Mice, Transgenic
16.
Proc Natl Acad Sci U S A ; 106(10): 3788-93, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19234109

ABSTRACT

DNA damage response (DDR) acts as a tumorigenesis barrier, and any defects in the DDR machinery may lead to cancer. SOX4 expression is elevated in many types of tumors; however, its role in DDR is still largely unknown. Here, we show that SOX4, a new DNA damage sensor, is required for the activation of p53 tumor suppressor in response to DNA damage. Notably, SOX4 interacts with and stabilizes p53 protein by blocking Mdm2-mediated p53 ubiquitination and degradation. Furthermore, SOX4 enhances p53 acetylation by interacting with p300/CBP and facilitating p300/CBP/p53 complex formation. In concert with these results, SOX4 promotes cell cycle arrest and apoptosis, and it inhibits tumorigenesis in a p53-dependent manner. Therefore, these findings highlight SOX4 as a potential key factor in regulating DDR-associated cancer.


Subject(s)
DNA Damage , SOXC Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Humans , Protein Binding , Protein Stability , Proto-Oncogene Proteins c-mdm2/metabolism , Ubiquitination
17.
J Cell Biol ; 221(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34813648

ABSTRACT

Primary cilia transduce diverse signals in embryonic development and adult tissues. Defective ciliogenesis results in a series of human disorders collectively known as ciliopathies. The CP110-CEP97 complex removal from the mother centriole is an early critical step for ciliogenesis, but the underlying mechanism for this step remains largely obscure. Here, we reveal that the linear ubiquitin chain assembly complex (LUBAC) plays an essential role in ciliogenesis by targeting the CP110-CEP97 complex. LUBAC specifically generates linear ubiquitin chains on CP110, which is required for CP110 removal from the mother centriole in ciliogenesis. We further identify that a pre-mRNA splicing factor, PRPF8, at the distal end of the mother centriole acts as the receptor of the linear ubiquitin chains to facilitate CP110 removal at the initial stage of ciliogenesis. Thus, our study reveals a direct mechanism of regulating CP110 removal in ciliogenesis and implicates the E3 ligase LUBAC as a potential therapy target of cilia-associated diseases, including ciliopathies and cancers.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Cilia/metabolism , Microtubule-Associated Proteins/metabolism , Organogenesis , Phosphoproteins/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Humans , Mice , Multiprotein Complexes , RNA-Binding Proteins/metabolism , Substrate Specificity , Ubiquitination , Zebrafish
18.
Cells ; 10(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33440657

ABSTRACT

Hedgehog (Hh) signaling is a classic morphogen in controlling embryonic development and tissue repairing. Aberrant activation of Hh signaling has been well documented in liver cancer, including hepatoblastoma, hepatocellular carcinoma (HCC) and cholangiocarcinoma. The present review aims to update the current understanding on how abnormal Hh signaling molecules modulate initiation, progression, drug resistance and metastasis of HCC. The latest relevant literature was reviewed with our recent findings to provide an overview regarding the molecular interplay and clinical relevance of the Hh signaling in HCC management. Hh signaling molecules are involved in the transformation of pre-carcinogenic lesions to malignant features in chronic liver injury, such as nonalcoholic steatohepatitis. Activation of GLI target genes, such as ABCC1 and TAP1, is responsible for drug resistance in hepatoma cells, with a CD133-/EpCAM- surface molecular profile, and GLI1 and truncated GLI1 account for the metastatic feature of the hepatoma cells, with upregulation of matrix metalloproteinases. A novel bioassay for the Sonic Hh ligand in tissue specimens may assist HCC diagnosis with negative α-fetoprotein and predict early microvascular invasion. In-depth exploration of the Hh signaling deepens our understanding of its molecular modulation in HCC initiation, drug sensitivity and metastasis, and guides precise management of HCC on an individual basis.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Hedgehog Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Signal Transduction , Animals , Bioengineering , Carcinogenesis/pathology , Humans
19.
Int J Biol Sci ; 17(11): 2772-2794, 2021.
Article in English | MEDLINE | ID: mdl-34345207

ABSTRACT

Autophagy and glycolysis are two catabolic processes that manipulate pancreatic ductal adenocarcinoma (PDAC) development in response to hypoxia sensing, yet the underlying mechanism of how they are interlinked remain elusive. Methods: The functional roles of Unc-51 like kinase 1 and 2 (ULK1/2) in pyruvate kinase M2 (PKM2) transcription and glycolysis under hypoxia were assessed by chromatin immunoprecipitation, luciferase reporter, glucose consumption and lactate production assay. Co-immunoprecipitation, cellular ubiquitination, His-pulldown, in vitro protein kinase assay, immunofluorescence, immunohistochemistry, CRISPR technology, in silico studies were adopted to determine the molecular mechanism. Correlation analyses were performed in KPC (Pdx1-Cre; LSL-KrasG12D/+; Trp53fl/+) mice and clinical samples from PDAC patients. Therapeutic potential of ULK1/2 inhibitor and 2-deoxyglucose (2-DG) or 3-bromopyruvate (3-BP) was evaluated in cell-derived xenograft (CDX) and the patient-derived xenograft (PDX) models of nude mice. Results: ULK1/2, but not ULK3, augments hypoxic glycolysis in PDAC cells mediated by PKM2 independent of BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3). Mechanistically, hypoxia stimulates ULK1 to translocate into nucleus, where it interacts with and phosphorylates yes-associated protein (YAP) at Ser227, resulting in YAP stabilization through blockade of ubiquitin-proteasome system (UPS), which in turn facilitates PKM2 transcription, glycolysis, cell proliferation in vitro as well as PDAC growth in mice. ULK1/2 is positively correlated with YAP and PKM2 in tumor tissues from KPC mice and clinical samples from PDAC patients. Pharmacological deactivation of ULK1/2 potentiates the antineoplastic efficacy of 2-DG and 3-BP in CDX and PDX models. Conclusion: Our findings underscore the Ser227 autophosphorylation-dependent nuclear YAP stabilization as a central node that couples ULK1/2-initiated autophagy to hypoxic glycolysis during PDAC development and propose that targeting ULK1/2 combined with 2-DG or 3-BP might be a feasible therapeutic strategy against PDAC.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/metabolism , YAP-Signaling Proteins/metabolism , Animals , Autophagy-Related Protein-1 Homolog/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Computer Simulation , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphorylation , Signal Transduction , Tumor Hypoxia , Xenograft Model Antitumor Assays , YAP-Signaling Proteins/genetics , Pancreatic Neoplasms
20.
Acta Biomater ; 119: 485-498, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33130305

ABSTRACT

Although various biodegradable materials have been investigated for ligament reconstruction fixation in the past decades, only few of them possess a combination of high mechanical properties, appropriate degradation rate, good biocompatibility, and osteogenic effect, thus limiting their clinical applications. A high-strength Zn-0.8Mn-0.4Mg alloy (i.e., Zn08Mn04Mg) with yield strength of 317 MPa was developed to address this issue. The alloy showed good biocompatibility and promising osteogenic effect in vitro. The degradation effects of Zn08Mn04Mg interference screws on the interface between soft tissue and bone were investigated in anterior cruciate ligament (ACL) reconstruction in rabbits. Compared to Ti6Al4V, the Zn alloy screws significantly accelerated the formation of new bone and further induced partial tendon mineralization, which promoted tendon-bone integration. The newly developed screws are believed to facilitate early joint function recovery and rehabilitation training and also avoid screw breakage during insertion, thereby contributing to an extensive clinical prospect.


Subject(s)
Anterior Cruciate Ligament Reconstruction , Anterior Cruciate Ligament , Alloys/pharmacology , Animals , Anterior Cruciate Ligament/surgery , Biomechanical Phenomena , Bone Screws , Rabbits , Zinc
SELECTION OF CITATIONS
SEARCH DETAIL