Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.497
1.
Int J Oral Sci ; 16(1): 38, 2024 May 11.
Article En | MEDLINE | ID: mdl-38734708

Periodontitis is a chronic inflammatory and immune reactive disease induced by the subgingival biofilm. The therapeutic effect for susceptible patients is often unsatisfactory due to excessive inflammatory response and oxidative stress. Sinensetin (Sin) is a nature polymethoxylated flavonoid with anti-inflammatory and antioxidant activities. Our study aimed to explore the beneficial effect of Sin on periodontitis and the specific molecular mechanisms. We found that Sin attenuated oxidative stress and inflammatory levels of periodontal ligament cells (PDLCs) under inflammatory conditions. Administered Sin to rats with ligation-induced periodontitis models exhibited a protective effect against periodontitis in vivo. By molecular docking, we identified Bach1 as a strong binding target of Sin, and this binding was further verified by cellular thermal displacement assay and immunofluorescence assays. Chromatin immunoprecipitation-quantitative polymerase chain reaction results also revealed that Sin obstructed the binding of Bach1 to the HMOX1 promoter, subsequently upregulating the expression of the key antioxidant factor HO-1. Further functional experiments with Bach1 knocked down and overexpressed verified Bach1 as a key target for Sin to exert its antioxidant effects. Additionally, we demonstrated that Sin prompted the reduction of Bach1 by potentiating the ubiquitination degradation of Bach1, thereby inducing HO-1 expression and inhibiting oxidative stress. Overall, Sin could be a promising drug candidate for the treatment of periodontitis by targeting binding to Bach1.


Basic-Leucine Zipper Transcription Factors , Oxidative Stress , Periodontitis , Ubiquitination , Oxidative Stress/drug effects , Periodontitis/drug therapy , Periodontitis/prevention & control , Periodontitis/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Ubiquitination/drug effects , Rats , Male , Disease Models, Animal , Antioxidants/pharmacology , Rats, Sprague-Dawley , Humans , Chromatin Immunoprecipitation , Blotting, Western , Real-Time Polymerase Chain Reaction , Molecular Docking Simulation , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Periodontal Ligament/cytology
2.
Chem Commun (Camb) ; 60(42): 5514-5517, 2024 May 21.
Article En | MEDLINE | ID: mdl-38693792

In this study, we propose a novel therapy system composed of UiO-66 nanoparticles, which contain quercetin combined with chloroquine (UQCNP), to achieve dual autophagy-ubiquitination blockade. Through UiO-66 NP drug loading, the solubility of quercetin (a proteasome inhibitor) was improved under physiological conditions, thereby increasing its effective concentration at the tumor site. The cell experiment results showed that UQCNP significantly increased the apoptosis rate of 4T1 cells by 73.6%, which was significantly higher than other groups. Transmission electron microscopy results showed that the autophagosome of cells in the UQCNP treatment group was significantly lower than that in other treatment groups. Moreover, western blot results showed that, compared with other groups, LC3 expression and proteasome activity (p < 0.01), as well as the tumor volume of mice treated with UQCNP (p < 0.01) were significantly reduced. These results indicate that UQCNP achieves effective tumor therapy by blocking the autophagy and proteasome pathways synchronously.


Autophagy , Chloroquine , Nanoparticles , Quercetin , Ubiquitination , Quercetin/pharmacology , Quercetin/chemistry , Chloroquine/pharmacology , Chloroquine/chemistry , Animals , Autophagy/drug effects , Mice , Nanoparticles/chemistry , Ubiquitination/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Mice, Inbred BALB C , Humans
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167220, 2024 Jun.
Article En | MEDLINE | ID: mdl-38718847

Glioblastoma is one of the most challenging malignancies with high aggressiveness and invasiveness and its development and progression of glioblastoma highly depends on branched-chain amino acid (BCAA) metabolism. The study aimed to investigate effects of inhibition of BCAA metabolism with cytosolic branched-chain amino acid transaminase (BCATc) Inhibitor 2 on glioblastoma, elucidate its underlying mechanisms, and explore therapeutic potential of targeting BCAA metabolism. The expression of BCATc was upregulated in glioblastoma and BCATc Inhibitor 2 precipitated apoptosis both in vivo and in vitro with the activation of Bax/Bcl2/Caspase-3/Caspase-9 axis. In addition, BCATc Inhibitor 2 promoted K63-linkage ubiquitination of mitofusin 2 (Mfn2), which subsequently caused lysosomal degradation of Mfn2, and then oxidative stress, mitochondrial fission and loss of mitochondrial membrane potential. Furthermore, BCATc Inhibitor 2 treatment resulted in metabolic reprogramming, and significant inhibition of expression of ATP5A, UQCRC2, SDHB and COX II, indicative of suppressed oxidative phosphorylation. Moreover, Mfn2 overexpression or scavenging mitochondria-originated reactive oxygen species (ROS) with mito-TEMPO ameliorated BCATc Inhibitor 2-induced oxidative stress, mitochondrial membrane potential disruption and mitochondrial fission, and abrogated the inhibitory effect of BCATc Inhibitor 2 on glioblastoma cells through PI3K/AKT/mTOR signaling. All of these findings indicate suppression of BCAA metabolism promotes glioblastoma cell apoptosis via disruption of Mfn2-mediated mitochondrial dynamics and inhibition of PI3K/AKT/mTOR pathway, and suggest that BCAA metabolism can be targeted for developing therapeutic agents to treat glioblastoma.


Amino Acids, Branched-Chain , Apoptosis , GTP Phosphohydrolases , Glioblastoma , Oxidative Stress , Humans , Oxidative Stress/drug effects , Apoptosis/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , GTP Phosphohydrolases/metabolism , Animals , Amino Acids, Branched-Chain/metabolism , Cell Line, Tumor , Mice , Mitochondrial Proteins/metabolism , Ubiquitin/metabolism , Signal Transduction/drug effects , Male , Ubiquitination/drug effects , Reactive Oxygen Species/metabolism
4.
Int J Oncol ; 64(6)2024 06.
Article En | MEDLINE | ID: mdl-38757343

Daunorubicin, also known as daunomycin, is a DNA­targeting anticancer drug that is used as chemotherapy, mainly for patients with leukemia. It has also been shown to have anticancer effects in monotherapy or combination therapy in solid tumors, but at present it has not been adequately studied in colorectal cancer (CRC). In the present study, from a screening using an FDA­approved drug library, it was found that daunorubicin suppresses GLI­dependent luciferase reporter activity. Daunorubicin also increased p53 levels, which contributed to both GLI1 suppression and apoptosis. The current detailed investigation showed that daunorubicin promoted the ß­TrCP­mediated ubiquitination and proteasomal degradation of GLI1. Moreover, a competition experiment using BODIPY­cyclopamine, a well­known Smo inhibitor, suggested that daunorubicin does not bind to Smo in HCT116 cells. Administration of daunorubicin (2 mg/kg, ip, qod, 15 days) into HCT116 xenograft mice profoundly suppressed tumor progress and the GLI1 level in tumor tissues. Taken together, the present results revealed that daunorubicin suppresses canonical Hedgehog pathways in CRC. Ultimately, the present study discloses a new mechanism of daunorubicin's anticancer effect and might provide a rationale for expanding the clinical application of daunorubicin.


Apoptosis , Colorectal Neoplasms , Daunorubicin , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1 , Humans , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Daunorubicin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Animals , Mice , Apoptosis/drug effects , HCT116 Cells , Smoothened Receptor/metabolism , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Signal Transduction/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Ubiquitination/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects
5.
Neurol India ; 72(2): 309-318, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38691475

BACKGROUND: Acute cerebral infarction (ACI) is a common neurological disease that is associated with high morbidity, disability and mortality rates. At present, antiplatelet therapy is a necessary treatment for ACI. The present study aimed to investigate the effects of omentin-1 on the intravenous thrombolysis of ACI. OBJECTIVE: The present study aimed to investigate the effects of omentin-1 on the intravenous thrombolysis of ACI. MATERIAL AND METHODS: The mouse model of ACI was induced using male C57BL/6 mice through middle cerebral artery occlusion (MCAO). Meanwhile, the murine BV2 microglial cells were pretreated with 0.1 mg/ml of lipopolysaccharide (LPS), and then induced with 2 mM of adenosine triphosphate (ATP). RESULTS: The omentin-1 mRNA expression in patients receiving intravenous thrombolysis for ACI was down-regulated compared with the normal group. Additionally, the serum level of omentin-1 was negatively correlated with National Institute of Health Stroke Scale (NIHSS) score or serum level of IL-1ß or MMP-2 in patients receiving intravenous thrombolysis for ACI. Meanwhile, the serum mRNA expression of omentin-1 was positively correlated with Barthel index or high-sensitivity C-reactive protein (hs-CRP) in patients undergoing intravenous thrombolysis for ACI. As observed from the in vitro model, Omentin-1 reduced inflammation, promoted cell growth, alleviated ROS-induced oxidative stress, and enhanced AMPK activity through activating NLRP3 ubiquitination. Omentin-1 presented ACI in the mouse model of ACI. Regulating AMPK activity contributed to controlling the effects of Omentin-1 on the in vitro model. CONCLUSIONS: Omentin-1 reduced neuroinflammation and ROS-induced oxidative stress in the mouse model of ACI, which was achieved by inhibiting NLRP3 ubiquitination through regulating AMPK activity. Therefore, omentin-1 may serve as a treatment factor for the intravenous thrombolysis of ACI in further clinical application.


Cytokines , GPI-Linked Proteins , Lectins , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Ubiquitination , Animals , Cytokines/metabolism , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , GPI-Linked Proteins/metabolism , Humans , Ubiquitination/drug effects , Disease Models, Animal , Cerebral Infarction/drug therapy , AMP-Activated Protein Kinases/metabolism , Thrombolytic Therapy/methods , Middle Aged , Aged
6.
CNS Neurosci Ther ; 30(4): e14711, 2024 04.
Article En | MEDLINE | ID: mdl-38644551

OBJECTIVE: To elucidate the relationship between USP19 and O(6)-methylguanine-DNA methyltransferase (MGMT) after temozolomide treatment in glioblastoma (GBM) patients with chemotherapy resistance. METHODS: Screening the deubiquitinase pannel and identifying the deubiquitinase directly interacts with and deubiquitination MGMT. Deubiquitination assay to confirm USP19 deubiquitinates MGMT. The colony formation and tumor growth study in xenograft assess USP19 affects the GBM sensitive to TMZ was performed by T98G, LN18, U251, and U87 cell lines. Immunohistochemistry staining and survival analysis were performed to explore how USP19 is correlated to MGMT in GBM clinical management. RESULTS: USP19 removes the ubiquitination of MGMT to facilitate the DNA methylation damage repair. Depletion of USP19 results in the glioblastoma cell sensitivity to temozolomide, which can be rescued by overexpressing MGMT. USP19 is overexpressed in glioblastoma patient samples, which positively correlates with the level of MGMT protein and poor prognosis in these patients. CONCLUSION: The regulation of MGMT ubiquitination by USP19 plays a critical role in DNA methylation damage repair and GBM patients' temozolomide chemotherapy response.


Antineoplastic Agents, Alkylating , DNA Methylation , DNA Modification Methylases , DNA Repair Enzymes , Drug Resistance, Neoplasm , Temozolomide , Tumor Suppressor Proteins , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , DNA Modification Methylases/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , DNA Methylation/drug effects , Mice, Nude , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Mice , Male , Female , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , DNA Repair/drug effects , Endopeptidases/metabolism , Endopeptidases/genetics , Xenograft Model Antitumor Assays , Ubiquitination/drug effects
7.
Bioorg Med Chem Lett ; 105: 129759, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38636717

Histone H2A mono-ubiquitination plays important roles in epigenetic gene expression and is also involved in tumorigenesis. Small molecules controlling H2A ubiquitination are of interest as potential chemical tools and anticancer drugs. To identify novel small molecule inhibitors of H2A ubiquitination, we synthesized and evaluated several compounds designed based on PRT4165 (1), which is a reported histone ubiquitin ligase RING1A inhibitor. We found that compound 11b strongly inhibited the viability and reduced histone H2A mono-ubiquitination in human osteosarcoma U2OS cells. Therefore, compound 11b is a promising lead compound for the development of H2A histone ubiquitination-inhibiting small molecules.


Histones , Small Molecule Libraries , Ubiquitination , Humans , Histones/metabolism , Ubiquitination/drug effects , Cell Line, Tumor , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Structure , Cell Survival/drug effects , Dose-Response Relationship, Drug
8.
Cells ; 13(8)2024 Apr 13.
Article En | MEDLINE | ID: mdl-38667290

Excessive secretion of pro-inflammatory cytokines leads to the disruption of intestinal barrier in inflammatory bowel disease (IBD). The inflammatory cytokine tumor necrosis factor alpha (TNFα) induces the assembly of the NLRP3 inflammasome, resulting in the augmented secretion of inflammatory cytokines implicated in the pathogenesis of inflammatory bowel disease (IBD). TNFα has also been known to induce the formation of immunoproteasome (IP), which incorporates immunosubunits LMP2, LMP7, and MECL-1. Inhibition of IP activity using the IP subunit LMP2-specific inhibitor YU102, a peptide epoxyketone, decreased the protein levels of NLRP3 and increased the K48-linked polyubiquitination levels of NLRP3 in TNFα-stimulated intestinal epithelial cells. We observed that inhibition of IP activity caused an increase in the protein level of the ubiquitin E3 ligase, tripartite motif-containing protein 31 (TRIM31). TRIM31 facilitated K48-linked polyubiquitination and proteasomal degradation of NLRP3 with an enhanced interaction between NLRP3 and TRIM31 in intestinal epithelial cells. In addition, IP inhibition using YU102 ameliorated the symptoms of colitis in the model mice inflicted with dextran sodium sulfate (DSS). Administration of YU102 in the DSS-treated colitis model mice caused suppression of the NLRP3 protein levels and accompanied inflammatory cytokine release in the intestinal epithelium. Taken together, we demonstrated that inhibiting IP under inflammatory conditions induces E3 ligase TRIM31-mediated NLRP3 degradation, leading to attenuation of the NLRP3 inflammatory response that triggers disruption of intestinal barrier.


Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Proteasome Endopeptidase Complex , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Ubiquitination , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Ubiquitin-Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Inflammasomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Mice , Humans , Ubiquitination/drug effects , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/pathology , Colitis/metabolism , Colitis/immunology , Tumor Necrosis Factor-alpha/metabolism , Dextran Sulfate , Disease Models, Animal
9.
Nat Commun ; 15(1): 3410, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38649684

Estrogen receptor α (ERα) plays a crucial role in regulating glucose and energy homeostasis during type 2 diabetes mellitus (T2DM). However, the underlying mechanisms remain incompletely understood. Here we find a ligand-independent effect of ERα on the regulation of glucose homeostasis. Deficiency of ERα in the liver impairs glucose homeostasis in male, female, and ovariectomized (OVX) female mice. Mechanistic studies reveal that ERα promotes hepatic insulin sensitivity by suppressing ubiquitination-induced IRS1 degradation. The ERα 1-280 domain mediates the ligand-independent effect of ERα on insulin sensitivity. Furthermore, we identify a peptide based on ERα 1-280 domain and find that ERα-derived peptide increases IRS1 stability and enhances insulin sensitivity. Importantly, administration of ERα-derived peptide into obese mice significantly improves glucose homeostasis and serum lipid profiles. These findings pave the way for the therapeutic intervention of T2DM by targeting the ligand-independent effect of ERα and indicate that ERα-derived peptide is a potential insulin sensitizer for the treatment of T2DM.


Diabetes Mellitus, Type 2 , Estrogen Receptor alpha , Glucose , Homeostasis , Insulin Resistance , Liver , Obesity , Animals , Female , Humans , Male , Mice , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Estrogen Receptor alpha/metabolism , Glucose/metabolism , Homeostasis/drug effects , Insulin Receptor Substrate Proteins/metabolism , Liver/metabolism , Liver/drug effects , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/drug therapy , Ovariectomy , Peptides/pharmacology , Ubiquitination/drug effects
10.
Int J Biochem Cell Biol ; 171: 106582, 2024 Jun.
Article En | MEDLINE | ID: mdl-38649007

DNA methylation is one of the most important epigenetic mark involved in many physiologic cellular processes and pathologies. During mitosis, the transmission of DNA methylation patterns from a mother to the daughter cells is ensured through the action of the Ubiquitin-like, containing PHD and RING domains, 1/DNA methyltransferase 1 (UHRF1/DNMT1) tandem. UHRF1 is involved in the silencing of many tumor suppressor genes (TSGs) via mechanisms that remain largely to be deciphered. The present study investigated the role and the regulation of UHRF1 poly-ubiquitination induced by thymoquinone, a natural anti-cancer drug, known to enhance or re-activate the expression of TSGs. We found that the auto-ubiquitination of UHRF1, induced by TQ, is mediated by reactive oxygen species, and occurs following DNA damage. We demonstrated that the poly-ubiquitinated form of UHRF1 is K63-linked and can still silence the tumor suppressor gene p16INK4A/CDKN2A. We further showed that TQ-induced auto-ubiquitination is mediated via the activity of Tip60. Since this latter is known as a nuclear receptor co-factor, we investigated if the glucocorticoid receptor (GR) might be involved in the regulation of UHRF1 ubiquitination. Activation of the GR, with dexamethasone, did not influence auto-ubiquitination of UHRF1. However, we could observe that TQ induced a K48-linked poly-ubiquitination of GR, probably involved in the proteosomal degradation pathway. Mass-spectrometry analysis of FLAG-HA-tagged UHRF1 identified UHRF1 partners involved in DNA repair and showed that TQ increased their association with UHRF1, suggesting that poly-ubiquitination of UHRF1 is involved in the DNA repair process. We propose that poly-ubiquitination of UHRF1 serves as a scaffold to recruit the DNA repair machinery at DNA damage sites.


Benzoquinones , CCAAT-Enhancer-Binding Proteins , DNA Repair , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Ubiquitination/drug effects , Benzoquinones/pharmacology , DNA Repair/drug effects , Antineoplastic Agents/pharmacology , DNA Damage/drug effects
11.
Environ Toxicol ; 39(6): 3548-3562, 2024 Jun.
Article En | MEDLINE | ID: mdl-38477013

Tumor cell metastasis is the key cause of death in patients with nasopharyngeal carcinoma (NPC). MiR-2110 was cloned and identified in Epstein-Barr virus (EBV)-positive NPC, but its role is unclear in NPC. In this study, we investigated the effect of miR-2110 on NPC metastasis and its related molecular basis. In addition, we also explored whether miR-2110 can be regulated by cinobufotalin (CB) and participate in the inhibition of CB on NPC metastasis. Bioinformatics, RT-PCR, and in situ hybridization were used to observe the expression of miR-2110 in NPC tissues and cells. Scratch, Boyden, and tail vein metastasis model of nude mouse were used to detect the effect of miR-2110 on NPC metastasis. Western blot, Co-IP, luciferase activity, colocalization of micro confocal and ubiquitination assays were used to identify the molecular mechanism of miR-2110 affecting NPC metastasis. Finally, miR-2110 induced by CB participates in CB-stimulated inhibition of NPC metastasis was explored. The data showed that increased miR-2110 significantly suppresses NPC cell migration, invasion, and metastasis. Suppressing miR-2110 markedly restored NPC cell migration and invasion. Mechanistically, miR-2110 directly targeted FGFR1 and reduced its protein expression. Decreased FGFR1 attenuated its recruitment of NEDD4, which downregulated NEDD4-induced phosphatase and tensin homolog (PTEN) ubiquitination and degradation and further increased PTEN protein stability, thereby inactivating PI3K/AKT-stimulated epithelial-mesenchymal transition signaling and ultimately suppressing NPC metastasis. Interestingly, CB, a potential new inhibitory drug for NPC metastasis, significantly induced miR-2110 expression by suppressing PI3K/AKT/c-Jun-mediated transcription inhibition. Suppression of miR-2110 significantly restored cell migration and invasion in CB-treated NPC cells. Finally, a clinical sample assay indicated that reduced miR-2110 was negatively correlated with NPC lymph node metastasis and positively related to NPC patient survival prognosis. In summary, miR-2110 is a metastatic suppressor involving in CB-induced suppression of NPC metastasis.


Bufanolides , Cell Movement , Mice, Nude , MicroRNAs , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , PTEN Phosphohydrolase , Receptor, Fibroblast Growth Factor, Type 1 , Ubiquitination , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Animals , Cell Line, Tumor , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/metabolism , Ubiquitination/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Bufanolides/pharmacology , Cell Movement/drug effects , Mice , Mice, Inbred BALB C , Male , Neoplasm Metastasis , Female , Gene Expression Regulation, Neoplastic/drug effects
12.
Eur J Neurosci ; 59(10): 2732-2747, 2024 May.
Article En | MEDLINE | ID: mdl-38501537

Elevated serum homocysteine (Hcy) level is a risk factor for Alzheimer's disease (AD) and accelerates cell aging. However, the mechanism by which Hcy induces neuronal senescence remains largely unknown. In this study, we observed that Hcy significantly promoted senescence in neuroblastoma 2a (N2a) cells with elevated ß-catenin and Kelch-like ECH-associated protein 1 (KEAP1) levels. Intriguingly, Hcy promoted the interaction between KEAP1 and the Wilms tumor gene on the X chromosome (WTX) while hampering the ß-catenin-WTX interaction. Mechanistically, Hcy attenuated the methylation level of the KEAP1 promoter CpG island and activated KEAP1 transcription. However, a slow degradation rate rather than transcriptional activation contributed to the high level of ß-catenin. Hcy-upregulated KEAP1 competed with ß-catenin to bind to WTX. Knockdown of both ß-catenin and KEAP1 attenuated Hcy-induced senescence in N2a cells. Our data highlight a crucial role of the KEAP1-ß-catenin pathway in Hcy-induced neuronal-like senescence and uncover a promising target for AD treatment.


Cellular Senescence , Homocysteine , Kelch-Like ECH-Associated Protein 1 , Neuroblastoma , Ubiquitination , beta Catenin , beta Catenin/metabolism , Cellular Senescence/drug effects , Cellular Senescence/physiology , Animals , Homocysteine/pharmacology , Homocysteine/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Cell Line, Tumor , Ubiquitination/drug effects , Neuroblastoma/metabolism , Humans , Neurons/metabolism , Neurons/drug effects
13.
Am J Physiol Renal Physiol ; 326(5): F814-F826, 2024 May 01.
Article En | MEDLINE | ID: mdl-38545647

Aquaporin 2 (AQP2) is a vasopressin (VP)-regulated water channel in the renal collecting duct. Phosphorylation and ubiquitylation of AQP2 play an essential role in controlling the cellular abundance of AQP2 and its accumulation on the plasma membrane in response to VP. Cullin-RING ubiquitin ligases (CRLs) are multisubunit E3 ligases involved in ubiquitylation and degradation of their target proteins, eight of which are expressed in the collecting duct. Here, we used an established cell model of the collecting duct (mpkCCD14 cells) to study the role of cullins in modulating AQP2. Western blotting identified Cul-1 to Cul-5 in mpkCCD14 cells. Treatment of cells for 4 h with a pan-cullin inhibitor (MLN4924) decreased AQP2 abundance, prevented a VP-induced reduction in AQP2 Ser261 phosphorylation, and attenuated VP-induced plasma membrane accumulation of AQP2 relative to the vehicle. AQP2 ubiquitylation levels were significantly higher after MLN4924 treatment compared with controls, and they remained higher despite VP treatment. Cullin inhibition increased ERK1/2 activity, a kinase that regulates AQP2 Ser261 phosphorylation, and VP-induced reductions in ERK1/2 phosphorylation were absent during MLN4924 treatment. Furthermore, the greater Ser261 phosphorylation and reduction in AQP2 abundance during MLN4924 treatment were attenuated during ERK1/2 inhibition. MLN4924 increased intracellular calcium levels via calcium release-activated calcium channels, inhibition of which abolished MLN4924 effects on Ser261 phosphorylation and AQP2 abundance. In conclusion, CRLs play a vital role in mediating some of the effects of VP to increase AQP2 plasma membrane accumulation and AQP2 abundance. Whether modulation of cullin activity can contribute to body water homeostasis requires further studies.NEW & NOTEWORTHY Aquaporin 2 (AQP2) is essential for body water homeostasis and is regulated by the antidiuretic hormone vasopressin. The posttranslational modification ubiquitylation is a key regulator of AQP2 abundance and plasma membrane localization. Here we demonstrate that cullin-RING E3 ligases play a vital role in mediating some of the effects of vasopressin to increase AQP2 abundance and plasma membrane accumulation. The results suggest that manipulating cullin activity could be a novel strategy to alter kidney water handling.


Aquaporin 2 , Cullin Proteins , Cyclopentanes , Kidney Tubules, Collecting , Pyrimidines , Ubiquitination , Aquaporin 2/metabolism , Cullin Proteins/metabolism , Animals , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/enzymology , Ubiquitination/drug effects , Phosphorylation , Mice , Vasopressins/metabolism , Vasopressins/pharmacology , Cell Line , Cell Membrane/metabolism , Cell Membrane/drug effects , Ubiquitin-Protein Ligases/metabolism , Calcium/metabolism
14.
Biomed Pharmacother ; 174: 116447, 2024 May.
Article En | MEDLINE | ID: mdl-38518606

Sepsis-induced acute respiratory distress syndrome (ARDS) causes significant fatalities worldwide and lacks pharmacological intervention. Alveolar fluid clearance (AFC) plays a pivotal role in the remission of ARDS and is markedly impaired in the pathogenesis of ARDS. Here, we demonstrated that erythropoietin could effectively ameliorate lung injury manifestations and lethality, restore lung function and promote AFC in a rat model of lipopolysaccharide (LPS)-induced ARDS. Moreover, it was proven that EPO-induced restoration of AFC occurs through triggering the total protein expression of ENaC and Na,K-ATPase channels, enhancing their protein abundance in the membrane, and suppressing their ubiquitination for degeneration. Mechanistically, the data indicated the possible involvement of EPOR/JAK2/STAT3/SGK1/Nedd4-2 signaling in this process, and the pharmacological inhibition of the pathway markedly eliminated the stimulating effects of EPO on ENaC and Na,K-ATPase, and subsequently reversed the augmentation of AFC by EPO. Consistently, in vitro studies of alveolar epithelial cells paralleled with that EPO upregulated the expression of ENaC and Na,K-ATPase, and patch-clamp studies further demonstrated that EPO substantially strengthened sodium ion currents. Collectively, EPO could effectively promote AFC by improving ENaC and Na,K-ATPase protein expression and abundance in the membrane, dependent on inhibition of ENaC and Na,K-ATPase ubiquitination, and resulting in diminishing LPS-associated lung injuries.


Epithelial Sodium Channels , Erythropoietin , Rats, Sprague-Dawley , Respiratory Distress Syndrome , Sepsis , Sodium-Potassium-Exchanging ATPase , Ubiquitination , Animals , Epithelial Sodium Channels/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Erythropoietin/pharmacology , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , Ubiquitination/drug effects , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Male , Rats , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Lipopolysaccharides , Signal Transduction/drug effects , Disease Models, Animal
15.
Int Immunopharmacol ; 130: 111749, 2024 Mar 30.
Article En | MEDLINE | ID: mdl-38430804

AIMS: Saikosaponin F (SsF) is one of the major active ingredients of Radix Bupleuri, an herb widely used in the treatment of depression. Studies have shown that dry eye disease often occurs together with depression. The aim of this study is to investigate whether SsF can improve depression-associated dry eye disease and explore the underlying mechanism. METHODS: Behavioral test was used to verify the effect of SsF on CUMS-induced depression-like behaviors in mice. Corneal fluorescein staining, phenol red cotton thread test and periodic acid-Schiff (PAS) staining were used to observe the effect of SsF on depression-associated dry eye disease. Western blot (WB) was performed to observe the expression of TAK1 protein and key proteins of NF-κB and MAPK (P38) inflammatory pathways in the hippocampus and cornea. Immunohistochemical staining was used to observe the expression of microglia, and immunoprecipitation was used to observe K63-linked TAK1 ubiquitination. Subsequently, we constructed a viral vector sh-TAK1 to silence TAK1 protein to verify whether SsF exerted its therapeutic effect based on TAK1. The expression of inflammatory factors such as IL-1ß, TNF-α and IL-18 in hippocampus and cornea were detected by ELISA. Overexpression of TRIM8 (OE-TRIM8) by viral vector was used to verify whether SsF improved depression-associated dry eye disease based on TRIM8. RESULTS: SsF treatment significantly improved the depression-like behavior, increased tear production and restored corneal injury in depression-related dry eye model mice. SsF treatment downregulated TAK1 expression and TRIM8-induced K63-linked TAK1 polyubiquitination, while inhibiting the activation of NF-κB and MAPK (P38) inflammatory pathways and microglial expression. In addition, selective inhibition of TAK1 expression ameliorated depression-associated dry eye disease, while overexpression of TRIM8 attenuated the therapeutic effect of SsF on depression-associated dry eye disease. CONCLUSION: SsF inhibited the polyubiquitination of TAK1 by acting on TRIM8, resulting in the downregulation of TAK1 expression, inhibition of inflammatory response, and improvement of CUMS-induced depression-associated dry eye disease.


Antidepressive Agents , Depression , Dry Eye Syndromes , MAP Kinase Kinase Kinases , NF-kappa B , Oleanolic Acid , Saponins , Ubiquitin-Protein Ligases , Animals , Male , Mice , Depression/complications , Depression/drug therapy , Dry Eye Syndromes/drug therapy , Dry Eye Syndromes/etiology , Inflammation/drug therapy , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins , NF-kappa B/metabolism , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Saponins/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
16.
J Biol Chem ; 300(3): 105752, 2024 Mar.
Article En | MEDLINE | ID: mdl-38354780

Cullin (CUL)-RING (Really Interesting New Gene) E3 ubiquitin (Ub) ligases (CRLs) are the largest E3 family. The E3 CRL core ligase is a subcomplex formed by the CUL C-terminal domain bound with the ROC1/RBX1 RING finger protein, which acts as a hub that mediates and organizes multiple interactions with E2, Ub, Nedd8, and the ARIH family protein, thereby resulting in Ub transfer to the E3-bound substrate. This report describes the modulation of CRL-dependent ubiquitination by small molecule compounds including KH-4-43, #33, and suramin, which target the CRL core ligases. We show that both KH-4-43 and #33 inhibit the ubiquitination of CK1α by CRL4CRBN. However, either compound's inhibitory effect on this reaction is significantly reduced when a neddylated form of CRL4CRBN is used. On the other hand, both #33 and KH-4-43 inhibit the ubiquitination of ß-catenin by CRL1ß-TrCP and Nedd8-CRL1ß-TrCP almost equally. Thus, neddylation of CRL1ß-TrCP does not negatively impact the sensitivity to inhibition by #33 and KH-4-43. These findings suggest that the effects of neddylation to alter the sensitivity of CRL inhibition by KH-4-43/#33 is dependent upon the specific CRL type. Suramin, a compound that targets CUL's basic canyon, can effectively inhibit CRL1/4-dependent ubiquitination regardless of neddylation status, in contrast to the results observed with KH-4-43/#33. This observed differential drug sensitivity of KH-4-43/#33 appears to echo CUL-specific Nedd8 effects on CRLs as revealed by recent high-resolution structural biology efforts. The highly diversified CRL core ligase structures may provide opportunities for specific targeting by small molecule modulators.


Ligands , Ubiquitin-Protein Ligases , Ubiquitination , Animals , Humans , Mice , beta Catenin/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Cullin Proteins/metabolism , Suramin/pharmacology , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , NEDD8 Protein/metabolism
17.
Am J Physiol Gastrointest Liver Physiol ; 326(4): G426-G437, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38290991

This study aims to investigate the role and molecular mechanism of anthocyanin in improving liver fibrosis through ferroptosis, providing a basis for drug development and targeted therapy. In this study, a mouse model of liver fibrosis was established using CCl4, and the anthocyanin treatment groups were administered 100 mg/kg anthocyanin daily via gavage. Furthermore, real-time fluorescent quantitative PCR (qRT-PCR), Western blotting (WB), and enzyme-linked immunosorbent assay were used to assess liver fibrosis indicators and liver injury markers. Histopathological methods were used to confirm the morphology of liver injury in different treatment groups. The effects of anthocyanins on ferroptosis markers, NCOA4 and FTH1 expression, were examined through qRT-PCR, WB, and Co-IP. Confocal microscopy was used to validate the colocalization of ferritin and lysosomes. A differential expression model of TRIM7 was constructed to verify its impact on the progression of liver fibrosis. The present study demonstrates the hepatoprotective effects of anthocyanins in liver fibrosis, highlighting their ability to enhance hepatic stellate cell (HSC) ferroptosis and regulate ferritin autophagy. Moreover, TRIM7 is identified as a key mediator of anthocyanin-induced regulation of hepatic stellate cells activation for liver fibrosis treatment through modulation of ferroautophagy. Mechanistic investigations further reveal that TRIM7 exerts its influence on the process of ferroautophagy by controlling NCOA4 ubiquitination. Our study discovered that anthocyanins could improve liver fibrosis by regulating NCOA4 ubiquitination through TRIM7, thereby affecting hepatic stellate cells' ferroptosis levels.NEW & NOTEWORTHY This was the first study to demonstrate that anthocyanins can improve the progression of liver fibrosis by promoting hepatic stellate cell (HSC) ferroptosis. Anthocyanins could affect the content of Fe2+ by promoting ferroautophagy in HSCs, thereby promoting the level of ferroptosis. This study demonstrates for the first time that anthocyanins can inhibit the expression of TRIM7 and then affect the ubiquitination of NCOA4 to regulate the level of ferritin autophagy and ferroptosis.


Anthocyanins , Blueberry Plants , Ferroptosis , Liver Cirrhosis , Animals , Mice , Anthocyanins/pharmacology , Anthocyanins/metabolism , Anthocyanins/therapeutic use , Blueberry Plants/chemistry , Ferritins , Ferroptosis/drug effects , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Ubiquitination/drug effects , Nuclear Receptor Coactivators/drug effects , Nuclear Receptor Coactivators/metabolism , Tripartite Motif Proteins/drug effects , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/metabolism
18.
Nature ; 626(8000): 874-880, 2024 Feb.
Article En | MEDLINE | ID: mdl-38297121

Stress response pathways detect and alleviate adverse conditions to safeguard cell and tissue homeostasis, yet their prolonged activation induces apoptosis and disrupts organismal health1-3. How stress responses are turned off at the right time and place remains poorly understood. Here we report a ubiquitin-dependent mechanism that silences the cellular response to mitochondrial protein import stress. Crucial to this process is the silencing factor of the integrated stress response (SIFI), a large E3 ligase complex mutated in ataxia and in early-onset dementia that degrades both unimported mitochondrial precursors and stress response components. By recognizing bifunctional substrate motifs that equally encode protein localization and stability, the SIFI complex turns off a general stress response after a specific stress event has been resolved. Pharmacological stress response silencing sustains cell survival even if stress resolution failed, which underscores the importance of signal termination and provides a roadmap for treating neurodegenerative diseases caused by mitochondrial import defects.


Mitochondria , Mitochondrial Proteins , Mutation , Neurodegenerative Diseases , Stress, Physiological , Ubiquitin-Protein Ligases , Apoptosis/drug effects , Ataxia/genetics , Cell Survival/drug effects , Dementia/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Stability/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Stress, Physiological/drug effects , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
19.
J Mol Biol ; 435(23): 168316, 2023 12 01.
Article En | MEDLINE | ID: mdl-37858708

Deubiquitinases (DUBs) are proteolytic enzymes that catalyze the removal of ubiquitin from protein substrates. The critical role of DUBs in regulating protein ubiquitination makes them attractive drug targets in oncology, neurodegenerative disease, and antiviral development. Biochemical assays for quantifying DUB activity have enabled characterization of substrate preferences and discovery of small molecule inhibitors. However, assessing the efficacy of these inhibitors in cellular contexts to support clinical drug development has been limited by a lack of tractable cell-based assays. To address this gap, we developed a two-color flow cytometry-based assay that allows for sensitive quantification of DUB activity and inhibition in living cells. The utility of this system was demonstrated by quantifying the potency of GRL0617 against the viral DUB SARS-CoV-2 PLpro, identifying potential GRL0617 resistance mutations, and performing structure-function analysis of the vOTU domain from the recently emerged Yezo virus. In addition, the system was optimized for cellular DUBs by modifying a GFP-targeting nanobody to recruit USP7 and USP28 to benchmark a panel of reported inhibitors and assess inhibition kinetics. Together, these results demonstrate the utility of these assays for studying DUB biology in a cellular context with potential to aid in inhibitor discovery and development.


Deubiquitinating Enzymes , Flow Cytometry , Protease Inhibitors , Humans , Aniline Compounds/pharmacology , Benzamides/pharmacology , Deubiquitinating Enzymes/analysis , Deubiquitinating Enzymes/antagonists & inhibitors , Neurodegenerative Diseases/enzymology , Ubiquitin/metabolism , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitination/drug effects , Flow Cytometry/methods , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Coronavirus Papain-Like Proteases/analysis , Coronavirus Papain-Like Proteases/antagonists & inhibitors , Single-Domain Antibodies
20.
Acta Pharmacol Sin ; 44(5): 940-953, 2023 May.
Article En | MEDLINE | ID: mdl-36357669

Dopaminergic neuron degeneration is a hallmark of Parkinson's disease (PD). We previously reported that the inactivation of von Hippel‒Lindau (VHL) alleviated dopaminergic neuron degeneration in a C. elegans model. In this study, we investigated the specific effects of VHL loss and the underlying mechanisms in mammalian PD models. For in vivo genetic inhibition of VHL, AAV-Vhl-shRNA was injected into mouse lateral ventricles. Thirty days later, the mice received MPTP for 5 days to induce PD. Behavioral experiments were conducted on D1, D3, D7, D14 and D21 after the last injection, and the mice were sacrificed on D22. We showed that knockdown of VHL in mice significantly alleviated PD-like syndromes detected in behavioral and biochemical assays. Inhibiting VHL exerted similar protective effects in MPP+-treated differentiated SH-SY5Y cells and the MPP+-induced C. elegans PD model. We further demonstrated that VHL loss-induced protection against experimental parkinsonism was independent of hypoxia-inducible factor and identified the Dishevelled-2 (DVL-2)/ß-catenin axis as the target of VHL, which was evolutionarily conserved in both C. elegans and mammals. Inhibiting the function of VHL promoted the stability of ß-catenin by reducing the ubiquitination and degradation of DVL-2. Thus, in vivo overexpression of DVL-2, mimicking VHL inactivation, protected against PD. We designed a competing peptide, Tat-DDF-2, to inhibit the interaction between VHL and DVL-2, which exhibited pharmacological potential for protection against PD in vitro and in vivo. We propose the therapeutic potential of targeting the interaction between VHL and DVL-2, which may represent a strategy to alleviate neurodegeneration associated with PD.


Dishevelled Proteins , Parkinson Disease , Von Hippel-Lindau Tumor Suppressor Protein , Animals , Humans , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , beta Catenin/metabolism , Caenorhabditis elegans/metabolism , Disease Models, Animal , Dishevelled Proteins/drug effects , Dishevelled Proteins/metabolism , Dopamine/pharmacology , Dopaminergic Neurons/metabolism , Mammals , Mice, Inbred C57BL , Neuroblastoma/metabolism , Parkinson Disease/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Ubiquitination/drug effects , Ubiquitination/genetics , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
...