Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Adv Res ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38897272

ABSTRACT

INTRODUCTION: Chemoimmunotherapy, which benefits from the combination of chemotherapy and immunotherapy, has emerged as a promising strategy in cancer treatment. However, effectively inducing a robust immune response remains challenging due to the limited responsiveness across patients. Endoplasmic reticulum (ER) stress is essential for activating intracellular signaling pathways associated with immunogenic cell death (ICD), targeting drugs to ER might enhance ER stress and improve ICD-related immunotherapy. OBJECTIVES: To improve the immune response of Chemoimmunotherapy. METHODS: ER targeting nanoparticles TSE-CEL/NP were constructed to enhance immunogenic cancer cell death. Flow cytometry, confocal microscope, TEM and immunofluorescence were used to evaluate the ER targeting effect and immunogenic tumor cell death in vitro on B16F10 tumor cells. Unilateral and bilateral tumor models were constructed to investigate the efficacy of anti-tumor and immunotherapy in vivo. Lung metastasis B16F10 melanoma tumor-bearing mice were used to assess the anti-metastasis efficacy. RESULTS: TSE-CEL/NP could specially accumulate in ER, thereby induce ER stress. High ER stress trigger the exposure of CRT, the extracellular release of HMGB1 and ATP. These danger signals subsequently promote the recruitment and maturation of dendritic cells (DCs), which in turn increase the proliferation of cytotoxic T lymphocytes (CD8+ T cells), ultimately resulted in an improved immunotherapy efficacy against melanoma. Invivo experiments showed that TSE-CEL/NP exhibits excellent antitumor efficacy and triggers a strong immune response. CONCLUSION: Our findings demonstrated that celastrol ER targeting delivery could amplify immunogenic cell death in melanoma, which provide experimental basis for melanoma immunotherapy.

2.
Immunol Res ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691319

ABSTRACT

Recent advances in cancer immunotherapy, especially immune checkpoint inhibitors (ICIs), have revolutionized the clinical outcome of many cancer patients. Despite the fact that impressive progress has been made in recent decades, the response rate remains unsatisfactory, and many patients do not benefit from ICIs. Herein, we summarized advanced studies and the latest insights on immune inhibitory factors in the tumor microenvironment. Our in-depth discussion and updated landscape of tumor immunosuppressive microenvironment may provide new strategies for reversing tumor immune evasion, enhancing the efficacy of ICIs therapy, and ultimately achieving a better clinical outcome.

3.
J Transl Med ; 22(1): 96, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38263194

ABSTRACT

BACKGROUND: Periodontitis is a chronic oral inflammatory disease that seriously affects people's quality of life. The purpose of our study was to investigate the correlation between the systemic immune inflammation index (SII) and periodontitis by utilizing a large national survey. This will establish a reference for the early identification and management of periodontitis. METHODS: This study comprised the adult US population who participated in a national periodontitis surveillance project during the six years from 2009 to 2014. Through the utilization of univariate and multivariate weighted logistic regression, we investigated the correlation between the systemic immune inflammation index and periodontitis. Additionally, we employed sensitivity analyses to evaluate the robustness of our findings. RESULTS: The study involved 10,366 participants with an average age of 51.00 years, of whom 49.45% were male (N = 5126) and 50.55% were female (N = 5240). The prevalence of periodontitis is estimated to be about 38.43% in the US adults aged 30 or older population. Our logistic regression models indicated a positive association between a SII higher than 978 × 109/L and periodontitis. The elder group (aged 50 or older) with SII higher than 978 × 109/L demonstrated a significant correlation with periodontitis in the fully adjusted model (Odds Ratio [OR] = 1.409, 95% Confidence Interval [CI] 1.037, 1.915, P = 0.022). However, there is no statistical difference among adults aged 30 to 50. The robustness of our findings was confirmed through sensitivity analyses. CONCLUSIONS: Our study highlights that SII is associated with periodontitis in a nationally representative sample of US adults. And the SII is significantly associated with a high risk of periodontitis in individuals aged 50 or older.


Subject(s)
Periodontitis , Quality of Life , Adult , Female , Male , Humans , Middle Aged , Cross-Sectional Studies , Inflammation , Logistic Models
4.
Neoplasia ; 48: 100966, 2024 02.
Article in English | MEDLINE | ID: mdl-38237304

ABSTRACT

Although immune checkpoint therapy has significantly improved the prognosis of patients with melanoma, urgent attention still needs to be paid to the low patient response rates and the challenges of precisely identifying patients before treatment. Therefore, it is crucial to investigate novel immunosuppressive mechanisms and targets in the tumor microenvironment in order to reverse tumor immune escape. In this study, we found that the cell cycle checkpoint Aurora kinase B (AURKB) suppressed the anti-tumor immune response, and its inhibitor, Tozasertib, effectively activated T lymphocyte cytokine release in vitro and anti-tumor immunity in vivo. Tozasertib significantly inhibited melanoma xenograft tumor growth by decreasing the number of inhibitory CD4+ Treg cells in the tumors, which, in turn, activated CD8+ T cells. Single-cell analysis revealed that AURKB suppressed anti-tumor immunity by increasing MIF-CD74/CXCR4 signaling between tumor cells and lymphocytes. Our study suggests that AURKB is a newly identified anti-tumor immunity suppressor, whose inhibitors may be developed as novel anti-tumor immunity drugs and may have synergistic anti-melanoma effects with immune checkpoint therapies.


Subject(s)
Antineoplastic Agents , Melanoma , Humans , Melanoma/pathology , T-Lymphocytes, Regulatory , CD8-Positive T-Lymphocytes , Antineoplastic Agents/therapeutic use , Tumor Microenvironment
5.
FEBS J ; 290(24): 5720-5743, 2023 12.
Article in English | MEDLINE | ID: mdl-37581853

ABSTRACT

Src homolog and collagen homolog binding protein 1 (SHCBP1) binds to the SH2 domain of SHC-transforming protein 1 (SHC1) and is involved in midbody organization and cytokinesis completion. SHCBP1 has been reported to be a cancer driver gene, promoting cancer progression. However, the functional role and underlying mechanism of SHCBP1 in regulating lung adenocarcinoma (LUAD) cell proliferation and migration are incompletely understood. Here, we discovered that SHCBP1 is overexpressed in LUAD tissues and is associated with a poor prognosis. SHCBP1 knockdown inhibited LUAD cell proliferation and migration by arresting the cell cycle and preventing epithelial-mesenchymal transition (EMT) via decreasing cyclin-dependent kinase 1 (CDK1) expression. Mechanistically, CDK1 overexpression reversed SHCBP1 knockdown-induced inhibition of proliferation and migration, confirming CDK1 as a key downstream target of SHCBP1. In addition, we proposed that rucaparib may be a small-molecule inhibitor of SHCBP1 and validated both in vitro and in vivo that rucaparib inhibits cell proliferation and migration via suppression of the SHCBP1/CDK1 pathway in LUAD. Our study elucidates a newly identified role of SHCBP1 in promoting cell proliferation and migration in LUAD, and suggests rucaparib as a potential inhibitor for LUAD treatment.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , CDC2 Protein Kinase/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Cell Proliferation , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Cell Movement , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Shc Signaling Adaptor Proteins
6.
EBioMedicine ; 94: 104724, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37480625

ABSTRACT

BACKGROUND: Cancer is the world's leading cause of death and a key hindrance to extending life expectancy. Celastrol, a bioactive compound derived from Tripterygium wilfordii, has been shown to have excellent antitumor activity, but its poor solubility and severe organ toxicity side effects have hampered its clinical application. METHODS: In this study, a self-assembled nanodrug (PLC-NP) was designed to deliver celastrol to tumor sites while efficiently reducing its side effects by conjugating celastrol with the bioactive material LMWH and P-selectin targeting peptide (PSN). Extensive in vitro and in vivo experiments were performed to investigate both therapeutic efficacy and adverse effects. Furthermore, the specific mechanism of the antitumor activity has also been explored. FINDING: The PLC-NP nanodrugs were spherical in shape, with a mean particle size of 115.83 ± 6.93 nm. PLC-NP was sufficiently stable during blood circulation, with a selective target to P-selectin-highly expressed tumor cells, followed by releasing the containing celastrol under acidic environment and high levels of esterase in tumor cells. Both in vitro and in vivo results confirmed that celastrol's antitumor and anti-metastatic abilities were not attenuated and were actually strengthened after being formed into nanodrugs. More importantly, the organ toxicities of the modified celastrol nanodrug were dramatically reduced. Mechanistic study indicated that the inactivation of PI3K/Akt/mTOR signaling pathway and ROS-mediated mitochondrial dysfunction play critical roles in celastrol-mediated autophagy and apoptosis. INTERPRETATION: Our findings could offer a potential strategy for the translation of toxic compounds into clinical therapeutic nanomedicine. FUNDING: See a detailed list of funding bodies in the Acknowledgements section at the end of the manuscript.


Subject(s)
Chemical and Drug Induced Liver Injury , Nanoparticles , Neoplasms , Triterpenes , Humans , Triterpenes/pharmacology , P-Selectin , Phosphatidylinositol 3-Kinases/metabolism , Heparin, Low-Molecular-Weight/pharmacology , Neoplasms/drug therapy , Nanoparticles/therapeutic use , Apoptosis
7.
Biomed Mater ; 18(3)2023 04 19.
Article in English | MEDLINE | ID: mdl-36917859

ABSTRACT

Cu2-xSe nanoparticles (Cu2-xSe NPs) as a new therapeutic drug platform is widely used in disease treatment due to their strong near-infrared optical absorption. In recent years, with their continuous expansion of applications in different fields, their own biological effects have received increasing attention. However, little is known about the effect of Cu2-xSe NPs on cancer cell. In this research, we found that Cu2-xSe NPs inhibited proliferation of HepG2 cells (IC50: 15.91µM) and SMMC-7721 cells (IC50: 43.15µM) and they mainly induced cell cycle arrest at the G2/M phase. Moreover, Cu2-xSe NPs inhibited HepG2 and SMMC-7721 cell migration and lamellopodia formation. Further studies indicated that Cu2-xSe NPs impaired mitochondrial respiration by inhibiting electron transport chain complex activity, thus reducing adenosine triphosphate levels. The insufficient energy supply subsequently impaired actin cytoskeleton assembly, ultimately inhibiting HepG2 and SMMC-7721 cell proliferation and migration. These findings suggest that Cu2-xSe NPs may have potentially antitumor activity, which might provide new insights of NPs into specific cancer treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Humans , Liver Neoplasms/drug therapy , Nanoparticles/therapeutic use , Hep G2 Cells , Cell Proliferation
8.
Carbohydr Polym ; 294: 119805, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35868765

ABSTRACT

Uncontrolled hemorrhage poses a severe life-threatening situation. However, traditional hemostats still have various limitations. It is urgent to develop a material with excellent biocompatibility and hemostatic ability. Evidence has shown that carboxymethyl chitosan (CMCS) has hemostatic properties and good compatibility. Herein, we develop an expandable hemostatic sponge by modifying CMCS with cellulose nanofibrils (CNFs) through the CO-NH cross-linking method. We verified its potential as a hemostatic agent both in vivo and in vitro. The results demonstrated that the prepared carboxymethyl chitosan/cellulose nanofiber composite (CNF-CMCS) sponges could absorb blood, quickly expand to exert pressure in the wound, and exhibit an excellent coagulation ability. The CNF-CMCS sponges significantly decreased the bleeding time and blood loss in several hemorrhage models and possessed a significant advantage in treating the deep penetrating injury hemorrhage. Therefore, the sponges provide a unique application prospect and potential as a penetrating trauma hemostatic agent.


Subject(s)
Chitosan , Hemostatics , Nanofibers , Carboxymethylcellulose Sodium/pharmacology , Cellulose/pharmacology , Chitosan/pharmacology , Chitosan/therapeutic use , Hemorrhage/drug therapy , Hemostasis , Hemostatics/pharmacology , Hemostatics/therapeutic use , Humans
9.
J Nanobiotechnology ; 20(1): 300, 2022 Jun 25.
Article in English | MEDLINE | ID: mdl-35752856

ABSTRACT

Deep tumor cells (cells in the center of solid tumors) play a crucial role in drug tolerance, metastasis, recurrence and microenvironment immune suppression. However, their deep location endows them with an untouched abdomen and makes them refractory to current treatments. Herein, we exploited the characteristic of higher autophagy in deep tumor cells than in superficial tumor cells and designed autophagy-responsive multifunctional nanoparticles (PGN) to enhance drug accumulation in deep tumor cells. PGNs were prepared by densely coating poly (lactic-co-glycolic acid) (PLGA) with cationic autophagy-responsive cell-penetrating peptide (GR9) and anionic 2,3-dimethylmaleic anhydride (DMA)-modified DSPE-PEG. The suitable nanoparticle size (122.4 nm) and charge-neutral surface (0.21 mV) of the NPs enabled long blood circulation. The hydrolysis of surface-anchored anionic DMA in the acidic microenvironment led to the exposure of the GR9 peptide and enhance tumor penetration. Once the PGN arrived in deep tumor cells with strong autophagy, GR9 was cut off by an autophagy shear enzyme, and the nanoparticles remained in the cells to undergo degradation. Furthermore, we prepared docetaxel (DTX) and chloroquine (CQ) loaded d-PGN. CQ inhibits autophagosome fusion with lysosomes, resulting in autophagosome accumulation, which further enhances the sensitivity of d-PGN to autophagy and their deep tumor retention. In vivo experiments showed that drug-loaded d-PGN achieved excellent antitumor efficacy with a peak inhibition rate of 82.1%. In conclusion, autophagy-responsive multifunctional nanoparticles provide a novel potential strategy for solid tumor treatment.


Subject(s)
Nanoparticles , Neoplasms , Autophagy , Cell Line, Tumor , Chloroquine/pharmacology , Docetaxel/pharmacology , Drug Delivery Systems , Humans , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Tumor Microenvironment
10.
Front Chem ; 10: 855843, 2022.
Article in English | MEDLINE | ID: mdl-35402373

ABSTRACT

In the aerobic oxidation of aldehydes to acids, how the solvent affect the reaction remains unclear. Herein, the solvent effect in the oxidation of 2-ethylhexanal (2-ETH) to 2-ethylhexanoic acid (2-ETA) was systematically investigated. The vastly different product distributions were observed which could be ascribed to the dominant intermolecular forces. Though strong intermolecular forces in protic solvents limit the oxidation, the optimal 2-ETA yield (96%) was obtained in ipropanol via gradually evaporating the solvent to remove the interactions. Theoretical calculations further revealed that the hydrogen bonds between reactant and protic solvent increase the C-H bond energy (-CHO in 2-ETH). Meanwhile, the hydrogen bonds may improve 2-ETA selectivity by promoting H transfer in the oxidation rearrangement step. Our work discloses the critical role of polarity in determining the reactivity and selectivity of 2-ETH oxidation, and could guide the rational design of more desirable reaction processes with solvent effect.

11.
ACS Appl Mater Interfaces ; 13(22): 25635-25648, 2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34038097

ABSTRACT

Metastasis is the main cause of death in cancer patients. The efficacy of pharmacological therapy for cancer is limited by the heterogeneous nature of cancer cells and the lack of knowledge of microenvironments in metastasis. Evidence has shown that activated platelets possess both tumor-homing and metastasis-targeting properties via intrinsic cell adhesion molecules on platelets, and malaria protein VAR2CSA is able to specifically bind to oncofetal chondroitin sulfate, which is overexpressed on cancer cells with both epithelial and mesenchymal phenotypes. Inspired by these mechanisms, we developed a recombinant VAR2CSA peptide (rVAR2)-modified activated platelet-mimicking nanoparticles (rVAR2-PM/PLGA-ss-HA) by coating the surface of disulfide-containing biodegradable PLGA conjugate nanoparticles (PLGA-ss-HA) with an activated platelet membrane. The results demonstrated that the engineered 122 nm rVAR2-PM/PLGA-ss-HA inherited the innate properties of the activated platelet membrane and achieved enhanced homing to both primary and metastatic foci. The nanoparticles were endocytosed and responded to a high intracellular concentration of reduced glutathione, resulting in nanoparticle disintegration and the release of chemotherapeutic drugs to kill tumor cells. Thus, rVAR2-decorated activated platelet-targeting nanoparticles with controlled drug release provide a promising drug delivery strategy for efficient treatment of primary and metastatic cancer.


Subject(s)
Antigens, Protozoan/metabolism , Blood Platelets/chemistry , Docetaxel/pharmacology , Drug Delivery Systems , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Nanoparticles/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Membrane/chemistry , Chondroitin Sulfates/metabolism , Delayed-Action Preparations , Docetaxel/chemistry , Hyaluronic Acid/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Rats , Rats, Sprague-Dawley
12.
Nanotechnology ; 32(34)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34010828

ABSTRACT

Novel core-shell nanostructure electrodes benefit from the excellent properties of their constituent materials, as well as the synergy between them. However, it is challenging to fabricate such structures efficiently. In this study, NiSe nanorods were fabricated using Ni foam as the conductive substrate and reactant via a one-step hydrothermal process, and Ni(OH)2nanosheets were coated on the surface of the nanorods via one-step electrodeposition. The effect of the structure and morphology on the properties of the material was explored using scanning electron microscopy, x-ray diffraction, and electrochemical technology. The obtained core-shell NiSe/Ni(OH)2exhibited an areal capacity of 1.89 mAh cm-2at a current density of 5 mA cm-2. The assembled NiSe/Ni(OH)2//AC hybrid supercapacitor exhibited excellent energy and power densities, indicating that NiSe/Ni(OH)2has great potential for use as a battery-type electrode in energy storage systems.

13.
Oncogene ; 40(20): 3492-3509, 2021 05.
Article in English | MEDLINE | ID: mdl-33911205

ABSTRACT

Lung cancer is one of the most intractable diseases with high incidence and mortality worldwide. Adenylate cyclase-associated protein 1 (CAP1), a well-known actin depolymerization factor, is recently reported to be an oncogene accelerating cancer cell proliferation. However, the physiological significance of CAP1 in lung cancer is incompletely understood and the novel functions of CAP1 in transcriptional regulation remain unknown. Here we found that CAP1 was highly expressed in lung cancer tissues and cells, which was also negatively associated with prognosis in lung cancer patients. Moreover, CAP1 promoted A549 cells proliferation by promoting protein synthesis to accelerate cell cycle progression. Mechanistically, we revealed that CAP1 facilitated cyclin-dependent kinase 9 (CDK9)-mediated RNA polymerases (Pol) II-Ser2 phosphorylation and subsequent transcription elongation, and CAP1 performed its function in this progress depending on its actin-depolymerization activity in nucleoplasm. Furthermore, our in vivo findings confirmed that CAP1-promoted A549 xenograft tumor growth was associated with CDK9-mediated Pol II-Ser2 phosphorylation. Our study elucidates a novel role of CAP1 in modulating transcription by promoting polymerase II phosphorylation and suggests that CAP1 is a newly identified biomarker for lung cancer treatment and prognosis prediction.


Subject(s)
Actins/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 9/metabolism , Cytoskeletal Proteins/metabolism , Lung Neoplasms/metabolism , RNA Polymerase II/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase 9/genetics , Cytoskeletal Proteins/genetics , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Phosphorylation , Polymerization , RNA Polymerase II/genetics , Survival Rate
14.
Article in English | MEDLINE | ID: mdl-33312453

ABSTRACT

The novel coronavirus SARS-CoV-2 is damaging the world's social and economic fabrics seriously. Effective drugs are urgently needed to decrease the high mortality rate of COVID-19 patients. Unfortunately, effective antiviral drugs or vaccines are currently unavailable. Herein, we systematically evaluated the effect of SARS-CoV-2 on gene expression of both lung tissue and blood from COVID-19 patients using transcriptome profiling. Differential gene expression analysis revealed potential core mechanism of COVID-19-induced pneumonia in which IFN-α, IFN-ß, IFN-γ, TNF and IL6 triggered cytokine storm mediated by neutrophil, macrophage, B and DC cells. Weighted gene correlation network analysis identified two gene modules that are highly correlated with clinical traits of COVID-19 patients, and confirmed that over-activation of immune system-mediated cytokine release syndrome is the underlying pathogenic mechanism for acute phase of COVID-19 infection. It suggested that anti-inflammatory therapies may be promising regimens for COVID-19 patients. Furthermore, drug repurposing analysis of thousands of drugs revealed that TNFα inhibitor etanercept and γ-aminobutyric acid-B receptor (GABABR) agonist baclofen showed most significant reversal power to COVID-19 gene signature, so we are highly optimistic about their clinical use for COVID-19 treatment. In addition, our results suggested that adalimumab, tocilizumab, rituximab and glucocorticoids may also have beneficial effects in restoring normal transcriptome, but not chloroquine, hydroxychloroquine or interferons. Controlled clinical trials of these candidate drugs are needed in search of effective COVID-19 treatment in current crisis.

15.
J Biol Chem ; 296: 100235, 2021.
Article in English | MEDLINE | ID: mdl-33376138

ABSTRACT

Epigenetic mechanisms that alter heritable gene expression and chromatin structure play an essential role in many biological processes, including liver function. Human MOF (males absent on the first) is a histone acetyltransferase that is globally downregulated in human steatohepatitis. However, the function of MOF in the liver remains unclear. Here, we report that MOF plays an essential role in adult liver. Genetic deletion of Mof by Mx1-Cre in the liver leads to acute liver injury, with increase of lipid deposition and fibrosis akin to human steatohepatitis. Surprisingly, hepatocyte-specific Mof deletion had no overt liver abnormality. Using the in vitro coculturing experiment, we show that Mof deletion-induced liver injury requires coordinated changes and reciprocal signaling between hepatocytes and Kupffer cells, which enables feedforward regulation to augment inflammation and apoptotic responses. At the molecular level, Mof deletion induced characteristic changes in metabolic gene programs, which bore noticeable similarity to the molecular signature of human steatohepatitis. Simultaneous deletion of Mof in both hepatocytes and macrophages results in enhanced expression of inflammatory genes and NO signaling in vitro. These changes, in turn, lead to apoptosis of hepatocytes and lipotoxicity. Our work highlights the importance of histone acetyltransferase MOF in maintaining metabolic liver homeostasis and sheds light on the epigenetic dysregulation in liver pathogenesis.


Subject(s)
Histone Acetyltransferases/genetics , Inflammation/metabolism , Liver Diseases/genetics , Liver/injuries , Nitric Oxide/genetics , Apoptosis/genetics , Chromatin/genetics , Epigenesis, Genetic/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Deletion , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Histone Acetyltransferases/chemistry , Humans , Inflammation/genetics , Inflammation/pathology , Lipids/adverse effects , Lipids/genetics , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Macrophages/metabolism , Macrophages/pathology , Nitric Oxide/metabolism , Signal Transduction/genetics
16.
FASEB J ; 34(11): 14863-14877, 2020 11.
Article in English | MEDLINE | ID: mdl-32918517

ABSTRACT

Appropriate control of hepatic gluconeogenesis is essential for the organismal survival upon prolonged fasting and maintaining systemic homeostasis under metabolic stress. Here, we show protein arginine methyltransferase 1 (PRMT1), a key enzyme that catalyzes the protein arginine methylation process, particularly the isoform encoded by Prmt1 variant 2 (PRMT1V2), is critical in regulating gluconeogenesis in the liver. Liver-specific deletion of Prmt1 reduced gluconeogenic capacity in cultured hepatocytes and in the liver. Prmt1v2 was expressed at a higher level compared to Prmt1v1 in hepatic tissue and cells. Gain-of-function of PRMT1V2 clearly activated the gluconeogenic program in hepatocytes via interactions with PGC1α, a key transcriptional coactivator regulating gluconeogenesis, enhancing its activity via arginine methylation, while no effects of PRMT1V1 were observed. Similar stimulatory effects of PRMT1V2 in controlling gluconeogenesis were observed in human HepG2 cells. PRMT1, specifically PRMT1V2, was stabilized in fasted liver and hepatocytes treated with glucagon, in a PGC1α-dependent manner. PRMT1, particularly Prmt1v2, was significantly induced in the liver of streptozocin-induced type 1 diabetes and high fat diet-induced type 2 diabetes mouse models and liver-specific Prmt1 deficiency drastically ameliorated diabetic hyperglycemia. These findings reveal that PRMT1 modulates gluconeogenesis and mediates glucose homeostasis under physiological and pathological conditions, suggesting that deeper understanding how PRMT1 contributes to the coordinated efforts in glycemic control may ultimately present novel therapeutic strategies that counteracts hyperglycemia in disease settings.


Subject(s)
Gluconeogenesis , Hepatocytes/metabolism , Hyperglycemia/genetics , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cells, Cultured , Gain of Function Mutation , Glucagon/metabolism , Glucose/metabolism , Hep G2 Cells , Humans , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein-Arginine N-Methyltransferases/genetics
17.
J Cell Mol Med ; 24(10): 5695-5706, 2020 05.
Article in English | MEDLINE | ID: mdl-32297701

ABSTRACT

Diabetic retinopathy (DR) is one of the most prominent microvascular complications of diabetes, which remains the leading cause of legal blindness in the world. Arctiin, a bioactive compound from Arctium lappa L., has been reported to have antidiabetic activity. In this study, we investigated the effect of arctiin on a human retinal capillary endothelial cell (HRCEC) line and how arctiin inhibits cell proliferation in high glucose (HG)-induced HRCECs. Results showed that arctiin decreased HG-induced HRCECs proliferation in a dose-dependent manner by inducing cell cycle arrest at the G0/G1 phase. Tube formation assay and immunofluorescence staining indicated that arctiin abrogated tube formation induced by HG-induced HRCECs in a dose-dependent manner via down-regulation of VEGF expression. Mechanistic study indicated that perturbation of the ROCK1/PTEN/PI3K/Akt signalling pathway plays a vital role in the arctiin-mediated anti-proliferative effect. Furthermore, pre-incubation of HRCECs with Y-27632 attenuated arctiin-induced cell cycle arrest, cell proliferation and tube formation inhibition. Y-27632 also reversed the activation of PTEN, the inactivation/dephosphorylation of PI3K/Akt and down-regulation of VEGF. Taken together, the results demonstrated that arctiin inhibits the proliferation of HG-induced HRCECs through the activation of ROCK1 and PTEN and inactivation of PI3K and Akt, resulting in down-regulation of VEGF, which inhibits endothelial cell proliferation.


Subject(s)
Endothelial Cells/metabolism , Glucose/metabolism , Glucosides/genetics , Retinal Vessels/cytology , Retinal Vessels/metabolism , Signal Transduction , Biomarkers , Cell Cycle Checkpoints , Cell Proliferation , Diabetic Retinopathy/etiology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endothelial Cells/drug effects , Fluorescent Antibody Technique , Furans/metabolism , Glucose/pharmacology , Glucosides/metabolism , Humans , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
18.
J Cell Mol Med ; 24(1): 618-631, 2020 01.
Article in English | MEDLINE | ID: mdl-31724308

ABSTRACT

Proliferation and metastasis are significantly malignant characteristics of human lung cancer, but the underlying molecular mechanisms are poorly understood. Chromobox 4 (CBX4), a member of the Polycomb group (PcG) family of epigenetic regulatory factors, enhances cellular proliferation and promotes cancer cell migration. However, the effect of CBX4 in the progression of lung cancer is not fully understood. We found that CBX4 is highly expressed in lung tumours compared with adjacent normal tissues. Overexpression of CBX4 significantly promotes cell proliferation and migration in human lung cancer cell lines. The knockdown of CBX4 obviously suppresses the cell growth and migration of human lung cancer cells in vitro. Also, the proliferation and metastasis in vivo are blocked by CBX4 knockdown. Furthermore, CBX4 knockdown effectively arrests cell cycle at the G0/G1 phase through suppressing the expression of CDK2 and Cyclin E and decreases the formation of filopodia through suppressing MMP2, MMP9 and CXCR4. Additionally, CBX4 promotes proliferation and metastasis via regulating the expression of BMI-1 which is a significant regulator of proliferation and migration in lung cancer cells. Taken together, these data suggest that CBX4 is not only a novel prognostic marker but also may be a potential therapeutic target in lung cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Ligases/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/pathology , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Female , Humans , Ligases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/genetics , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Oncol Rep ; 42(6): 2856, 2019 12.
Article in English | MEDLINE | ID: mdl-31638240

ABSTRACT

After having carefully checked the original data of Fig. 3, the authors noted that the student in their research group had inadvertently selected incorrect images for the 10 and 20 µM lycorine experiments to show the effect of lycorine on the migration of HepG2 cells during the figure compilation process. The corrected version of Fig. 3 is shown. The authors confirm that this error did not influence the statistical analysis shown for the migration of the cells, and neither were the overall results and conclusions of this article affected. The authors appreciate this opportunity to correct the scientific record, and all authors agree with this correction. Furthermore, the authors apologize for not noticing this error prior to publication, and for any inconvenience caused. [the original article was published in Oncology Reports 40: 2298­2306, 2018; DOI: 10.3892/or.2018.6609].

20.
Exp Mol Med ; 51(10): 1-13, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578315

ABSTRACT

Dopamine deficiency is mainly caused by apoptosis of dopaminergic nerve cells in the substantia nigra of the midbrain and the striatum and is an important pathologic basis of Parkinson's disease (PD). Recent research has shown that dynamin-related protein 1 (Drp1)-mediated aberrant mitochondrial fission plays a crucial role in dopaminergic nerve cell apoptosis. However, the upstream regulatory mechanism remains unclear. Our study showed that Drp1 knockdown inhibited aberrant mitochondrial fission and apoptosis. Importantly, we found that ROCK1 was activated in an MPP+-induced PD cell model and that ROCK1 knockdown and the specific ROCK1 activation inhibitor Y-27632 blocked Drp1-mediated aberrant mitochondrial fission and apoptosis of dopaminergic nerve cells by suppressing Drp1 dephosphorylation/activation. Our in vivo study confirmed that Y-27632 significantly improved symptoms in a PD mouse model by inhibiting Drp1-mediated aberrant mitochondrial fission and apoptosis. Collectively, our findings suggest an important molecular mechanism of PD pathogenesis involving ROCK1-regulated dopaminergic nerve cell apoptosis via the activation of Drp1-induced aberrant mitochondrial fission.


Subject(s)
Dopamine/deficiency , Dopaminergic Neurons/metabolism , Dynamins/genetics , Parkinson Disease, Secondary/genetics , rho-Associated Kinases/genetics , 1-Methyl-4-phenylpyridinium/administration & dosage , Amides/pharmacology , Animals , Apoptosis/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dynamins/antagonists & inhibitors , Dynamins/metabolism , Gene Expression Regulation , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Oxidative Stress , PC12 Cells , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/pathology , Pyridines/pharmacology , Rats , Signal Transduction , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , rho-Associated Kinases/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...