Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 177: 116967, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38908206

ABSTRACT

Snakebite envenomation often induces acute kidney injury (AKI) and acute liver injury (ALI), leading to augmented injuries and poor rehabilitation. Phospholipase A2 (PLA2) and metalloproteinase (SVMP) present in venom are responsible for the envenomation-associated events. In this study, mice envenomed with Deinagkistrodon acutus, Naja atra, or Agkistrodon halys pallas venom exhibited typical AKI and ALI symptoms, including significantly increased plasma levels of myoglobin, free hemoglobin, uric acid, aspartate aminotransferase, and alanine aminotransferase and upregulated expression of kidney NGAL and KIM-1. These effects were significantly inhibited when the mice were pretreated with natural inhibitors of PLA2 and SVMP isolated from Sinonatrix annularis (SaPLIγ and SaMPI). The inhibitors protected the physiological structural integrity of the renal tubules and glomeruli, alleviating inflammatory infiltration and diffuse hemorrhage in the liver. Furthermore, the dual therapy alleviated oxidative stress and apoptosis in the kidneys and liver by mitigating mitochondrial damage, thereby effectively reducing the lethal effect of snake venom in the inhibitor-treated mouse model. This study showed that dual therapy with inhibitors of metalloproteinase and phospholipase can effectively prevent ALI and AKI caused by snake bites. Our findings suggest that intrinsic inhibitors present in snakes are prospective therapeutic agents for multi-organ injuries caused by snake envenoming.

2.
Int J Mol Sci ; 25(2)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38255948

ABSTRACT

Heat shock proteins (Hsps) are a group of stress-induced proteins involved in protein folding and maturation. Based on their molecular weight, Hsps can be divided into six families: small Hsps, Hsp40, Hsp60, Hsp70, Hsp90, and large Hsps. In the process of breast cancer tumorigenesis, Hsps play a central role in regulating cell reactions and functions including proliferation, metastasis, and apoptosis. Moreover, some of the critical Hsps also regulate the fine balance between the protective and destructive immunological responses within the tumor microenvironment. In this review, we systematically summarize the roles of major Hsps in breast cancer biology and point out the potential uses of these proteins in breast cancer diagnosis and therapy. Understanding the roles of different families of Hsps in breast cancer pathogenesis will help in the development of more effective prevention and treatment measures for breast cancer.


Subject(s)
Heat-Shock Proteins, Small , Neoplasms , Heat-Shock Proteins , HSP70 Heat-Shock Proteins , HSP90 Heat-Shock Proteins , Apoptosis
3.
Cell Signal ; 112: 110913, 2023 12.
Article in English | MEDLINE | ID: mdl-37797796

ABSTRACT

Chemotherapy resistance of breast cancer cells is one of the major factors affecting patient survival rate. Heat shock protein 27 (Hsp27) is a member of the small heat shock protein family that has been reported to be associated with chemotherapy resistance in tumor cells, but the exact mechanism is not fully understood. Here, we explored the regulation of Hsp27 in adriamycin-resistant pathological conditions of breast cancer in vitro and in vivo. We found that overexpression of Hsp27 in MCF-7 breast cancer cells reversed DNA damage induced by adriamycin, and thereby reduced subsequent cell apoptosis. Non-phosphorylated Hsp27 accelerated ubiquitin-mediated degradation of c-Myc under normal physiological conditions. After stimulation with adriamycin, Hsp27 was phosphorylated and translocated from the cytoplasm into the nucleus, where phosphorylated Hsp27 upregulated c-Myc and Nijmegen breakage syndrome 1 (NBS1) protein levels thus leading to ATM activation. We further showed that phosphorylated Hsp27 promoted c-Myc nuclear import and stabilization by regulating T58/S62 phosphorylation of c-Myc through a protein phosphatase 2A (PP2A)-dependent mechanism. Collectively, the data presented in this study demonstrate that Hsp27, in its phosphorylation state, plays a critical role in adriamycin-resistant pathological conditions of breast cancer cells.


Subject(s)
Breast Neoplasms , Doxorubicin , Female , Humans , Apoptosis , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , HSP27 Heat-Shock Proteins/metabolism , Phosphorylation
4.
Toxicology ; 490: 153511, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37059347

ABSTRACT

Sodium p-perfluorous nonenoxybenzene sulfonate (OBS), an economical alternative to perfluorooctane sulfonate (PFOS) in multiple industrial fields, is widely detected in the environment. The toxicity of OBS has received increasing attention. Pituitary cells are components of the endocrine system and act as vital regulators of homeostatic endocrine balance. However, the effects of OBS on pituitary cells remain unknown. The present study explores the effects of OBS (0.5, 5, and 50 µM) on GH3 rat pituitary cells after treatment for 24, 48, and 72 h. We found that OBS significantly inhibited cell proliferation in GH3 cells with remarkable senescent phenotypes, including enhanced SA-ß-gal activity and expression of senescence-associated secretory phenotype (SASP)-related genes, cell cycle arrest, and upregulation of the senescence-related proteins γ-H2A.X and Bcl-2. OBS caused significant cell cycle arrest of GH3 cells at the G1-phase and concomitantly downregulated the expression of some key proteins for the G1/S transition, including cyclin D1 and cyclin E1. Consistently, the phosphorylation of retinoblastoma (RB), which plays a central role in regulating the cell cycle, was prominently reduced after OBS exposure. Furthermore, OBS notably activated the p53-p21 signalling pathway in GH3 cells, as evidenced by increased p53 and p21 expressions, enhanced p53 phosphorylation, and augmented p53 nuclear import. To our knowledge, this study is the first to reveal that OBS triggers senescence in pituitary cells via the p53-p21-RB signalling pathway. Our study demonstrates a novel toxic effect of OBS in vitro, and provides new perspectives for understanding the potential toxicity of OBS.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Animals , Rats , Cell Cycle , Cell Cycle Checkpoints , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Retinoblastoma Protein , Tumor Suppressor Protein p53/metabolism
5.
Inflamm Res ; 72(3): 603-621, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36690783

ABSTRACT

OBJECTIVE: To explore the molecular mechanism of γ-glutamylcysteine (γ-GC) in response to inflammation in vivo and in vitro on regulating the polarization of macrophages. METHODS: The expressions of gene or protein were assessed by qPCR and Western blot assays, respectively. Cell viability was investigated by CCK-8 assay. Eight-week-old male BALB/c mice were established to examine the therapeutic effects of γ-GC in vivo. The release of TNF-α and IL-4 was determined by ELISA assay. Macrophages polarization was identified by flow cytometry assay. RESULTS: Our data showed that γ-GC treatment significantly improved the survival, weight loss, and colon tissue damage of IBD mice. Furthermore, we established M1- and M2-polarized macrophages, respectively, and our findings provided evidence that γ-GC switched M1/M2-polarized macrophages through activating AMPK/SIRT1 axis and inhibiting inflammation-related signaling pathway. CONCLUSION: Collectively, both in vivo and in vitro experiments suggested that γ-GC has the potential to become a promising novel therapeutic dipeptide for the treatment of IBD, which provide new ideas for the treatment of inflammatory diseases in the future.


Subject(s)
Inflammatory Bowel Diseases , Male , Animals , Mice , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Macrophages/metabolism , Inflammation/metabolism , Dipeptides/pharmacology , Dipeptides/therapeutic use , Dipeptides/metabolism
6.
Sensors (Basel) ; 22(17)2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36080860

ABSTRACT

A radar is an important part of an air defense and combat system. It is of great significance to military defense to improve the effectiveness of radar state monitoring and the accuracy of fault diagnosis during operation. However, the complexity of radar equipment's structure and the uncertainty of the operating environment greatly increase the difficulty of fault diagnosis in real life situations. Therefore, a Bayesian network diagnosis method based on multi-source information fusion technology is proposed to solve the fault diagnosis problems caused by uncertain factors such as the high integration and complexity of the system during the process of fault diagnosis. Taking a fault of a radar receiver as an example, we study 2 typical fault phenomena and 21 fault points. After acquiring and processing multi-source information, establishing a Bayesian network model, determining conditional probability tables (CPTs), and finally outputting the diagnosis results. The results are convincing and consistent with reality, which verifies the effectiveness of this method for fault diagnosis in radar receivers. It realizes device-level fault diagnosis, which shortens the maintenance time for radars and improves the reliability and maintainability of radars. Our results have significance as a guide for judging the fault location of radars and predicting the vulnerable components of radars.

7.
Nutr Metab (Lond) ; 19(1): 29, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35428314

ABSTRACT

BACKGROUND: L-theanine, a non-protein amino acid was found principally in the green tea, has been previously shown to exhibit potent anti-obesity property and hepatoprotective effect. Herein, we investigated the effects of L-theanine on alleviating nonalcoholic hepatic steatosis in vitro and in vivo, and explored the underlying molecular mechanism. METHODS: In vitro, HepG2 and AML12 cells were treated with 500 µM oleic acid (OA) or treated with OA accompanied by L-theanine. In vivo, C57BL/6J mice were fed with normal control diet (NCD), high-fat diet (HFD), or HFD along with L-theanine for 16 weeks. The levels of triglycerides (TG), accumulation of lipid droplets and the expression of genes related to hepatocyte lipid metabolic pathways were detected in vitro and in vivo. RESULTS: Our data indicated that, in vivo, L-theanine significantly reduced body weight, hepatic steatosis, serum levels of alanine transaminase (ALT), aspartate transaminase (AST), TG and LDL cholesterol (LDL-C) in HFD-induced nonalcoholic fatty liver disease (NAFLD) mice. In vitro, L-theanine also significantly alleviated OA induced hepatocytes steatosis. Mechanic studies showed that L-theanine significantly inhibited the nucleus translocation of sterol regulatory element binding protein 1c (SREBP-1c) through AMPK-mTOR signaling pathway, thereby contributing to the reduction of fatty acid synthesis. We also identified that L-theanine enhanced fatty acid ß-oxidation by increasing the expression of peroxisome proliferator-activated receptor α (PPARα) and carnitine palmitoyltransferase-1 A (CPT1A) through AMP-activated protein kinase (AMPK). Furthermore, our study indicated that L-theanine can active AMPK through its upstream kinase Calmodulin-dependent protein kinase kinase-ß (CaMKKß). CONCLUSIONS: Taken together, our findings suggested that L-theanine alleviates nonalcoholic hepatic steatosis by regulating hepatocyte lipid metabolic pathways via the CaMKKß-AMPK signaling pathway.

8.
Cell Mol Life Sci ; 79(1): 54, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34936032

ABSTRACT

Glutathione S-transferase pi (GSTpi) is an important phase II detoxifying enzyme that participates in various physiological processes, such as antioxidant, detoxification, and signal transduction. The high expression level of GSTpi has been reported to be related to drug-resistant and anti-inflammatory and it functioned via its non-catalytic ligandin. However, the previous protection mechanism of GSTpi in DNA damage has not been addressed so far. Nijmegen breakage syndrome 1 (NBS1) is one of the most important sensor proteins to detect damaged DNA. Here, we investigated the interaction between GSTpi and NBS1 in HEK-293 T cells and human breast adenocarcinoma cells during DNA damage. Our results showed that overexpression of GSTpi in cells by transfecting DNA vector decreased the DNA damage level after methyl methanesulfonate (MMS) or adriamycin (ADR) treatment. We found that cytosolic GSTpi could increase NBS1 ubiquitin-mediated degradation in unstimulated cells, which suggested that GSTpi could maintain the basal level of NBS1 during normal conditions. In response to DNA damage, GSTpi can be phosphorylated in Ser184 and inhibit the ubiquitination degradation of NBS1 mediated by Skp2 to recover NBS1 protein level. Phosphorylated GSTpi can further enhance NBS1 nuclear translocation to activate the ATM-Chk2-p53 signaling pathway. Finally, GSTpi blocked the cell cycle in the G2/M phase to allow more time for DNA damage repair. Thus, our finding revealed the novel mechanism of GSTpi via its Ser184 phosphorylation to protect cells from cell death during DNA damage and it enriches the function of GSTpi in drug resistance.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , Glutathione S-Transferase pi/physiology , Nijmegen Breakage Syndrome/metabolism , Nuclear Proteins/metabolism , HEK293 Cells , Humans , MCF-7 Cells , Phosphorylation , Ubiquitination
9.
Toxicol Appl Pharmacol ; 433: 115778, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34755645

ABSTRACT

Indoprofen is a non-steroidal anti-inflammatory drug, and has provided insights into treatment of spinal muscular atrophies; however, the treatment effect of indoprofen on sepsis and the precise underlying mechanism remain to be elucidated. This study was carried out to examine the inhibitory effect of indoprofen on high mobility group box 1 (HMGB1)-mediated inflammatory responses in vivo and in vitro. Intraperitoneal injection of indoprofen (20 or 40 mg/kg) at 8 h post-sepsis markedly improved the survival of BALB/c mice and ameliorated multiple-organ injury by blocking the inflammatory responses. In addition, indoprofen partially reduced the HMGB1 level in the serum and in the lung, as well as ameliorated pulmonary edema. Mechanistically, indoprofen potently inhibited the release of HMGB1 following stimulation by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly I:C), and suppressed recombinant human HMGB1(rhHMGB1)-induced inflammatory responses. It was also found that indoprofen has both cyclooxygenase 2-dependent and -independent inhibitory effects on the proinflammatory effect of HMGB1 in THP-1 cells. Further, the drug reduced rhHMGB1-induced cell surface levels of toll-like receptor 2, toll-like receptor 4, and receptor of advanced glycation end-products in a concentration-dependent manner. Collectively, these data demonstrated that the anti-inflammatory effect of indoprofen in sepsis was associated with HMGB1-mediated inflammatory responses, thus offering a favorable mechanistic basis to support the therapeutic potential of indoprofen for the treatment of lethal sepsis or other inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , HMGB1 Protein/metabolism , Indoprofen/pharmacology , Inflammation Mediators/metabolism , Lung/drug effects , Sepsis/prevention & control , Animals , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Humans , Lung/immunology , Lung/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , RAW 264.7 Cells , Receptor for Advanced Glycation End Products/metabolism , Sepsis/immunology , Sepsis/metabolism , Signal Transduction , THP-1 Cells , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
10.
Life Sci ; 275: 119355, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-33744326

ABSTRACT

AIM: The aim of this study was to explore the antitumor effect of citrate on prostate cancer and its underlying mechanism. MAIN METHODS: CCK-8 and Colony formation assay were performed to detect the anti-proliferative effect of citrate on prostate cancer. Flow cytometry analysis was conducted to investigate the pro-apoptosis effect of citrate on prostate cancer. Immunofluorescence assay was taken to detect whether citrate induced autophagy in prostate cancer. Western blot and Immunohistochemical assay were performed to explore the underlying mechanism by which citrate activates autophagic death in prostate cancer cells. Xenograft tumorigenicity assay was conducted to explore whether citrate suppressed the growth of xenograft prostate tumors in vivo. KEY FINDINGS: We found citrate could significantly induce apoptosis and autophagy of prostate cancer cells in vitro and in vivo. Furthermore, treatment with autophagy inhibitor (chloroquine) drastically suppresses the apoptosis rate of prostate cancer induced by citrate. Based on the Ca2+-chelating property of citrate, the further study suggested that citrate activates autophagic cell death in prostate cancer cells via downregulation CaMKII/AKT/mTOR pathway. Finally, citrate suppresses the growth of xenograft prostate tumors without remarkable toxicity in mice. SIGNIFICANCE: Our study elucidated a novel molecular mechanism about the anti-cancer activities of citrate. That citrate activates autophagic cell death of prostate cancer via downregulation CaMKII/AKT/mTOR pathway and without remarkable toxicity in mice. This study suggests that citrate might be a promising therapeutic agent for the treatment of prostate cancer.


Subject(s)
Autophagic Cell Death/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Citric Acid/pharmacokinetics , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Down-Regulation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , PC-3 Cells , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects
11.
J Nutr Biochem ; 89: 108556, 2021 03.
Article in English | MEDLINE | ID: mdl-33249185

ABSTRACT

Prostate cancer (PCa) is a very prevalent male-specific malignancy; most PCa patients eventually die as a result of metastasis. L-theanine (C7H14N2O3), a nonprotein amino acid derivative from green tea leaves, has been demonstrated to act as an anticarcinogen through proapoptotic and antiproliferative effects. However, the antimetastatic effect of L-theanine in tumor cells and its underlying mechanism are still unclear. Here, we found that L-theanine could suppress invasion, migration, and increase cell-cell adhesion of prostate cancer cells in vitro and in vivo. We also found that L-theanine could inhibit the epithelial-mesenchymal transition process in PCa. Our study revealed that L-theanine could downregulate MMP9, N-cadherin, Vimentin, Snail, and upregulate E-cadherin. Furthermore, L-theanine suppressed the transcription of MMP9 and Snail by significantly inhibiting the ERK/NF-κB signaling pathway and the binding activity of p65 to the promoter regions of MMP9 and Snail. All of these findings suggest that L-theanine has therapeutic potential for metastatic PCa and may be considered a promising candidate for antimetastatic therapy of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Glutamates/pharmacology , Matrix Metalloproteinase 9/metabolism , Neoplasm Metastasis/pathology , Prostatic Neoplasms/pathology , Snail Family Transcription Factors/metabolism , Animals , Antineoplastic Agents/metabolism , Cadherins/metabolism , Cell Movement/drug effects , Down-Regulation , Epithelial-Mesenchymal Transition/drug effects , Glutamates/metabolism , Humans , Male , Mice , NF-kappa B/metabolism , PC-3 Cells , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Signal Transduction/drug effects , Tea/chemistry , Vimentin/metabolism
12.
J Nutr Biochem ; 82: 108398, 2020 08.
Article in English | MEDLINE | ID: mdl-32402912

ABSTRACT

Neointimal hyperplasia is a prominent pathological phenomenon in the process of stent restenosis. Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) play major pathological processes involved in the development of restenosis. l-Theanine, one of the major amino acid components in green tea, has been reported to improve vascular function. Here we display the effects of l-theanine on neointima formation and the underlying mechanism. In the rat carotid-artery balloon-injury model, l-theanine greatly inhibited neointima formation and prevented VSMCs from a contractile phenotype switching to a synthetic phenotype. In vitro study showed that l-theanine significantly inhibited PDGF-BB-induced VSMC proliferation and migration, which was comparable with the effect of l-theanine on AngII-induced VSMC proliferation and migration. Western blot analysis demonstrated that l-theanine suppressed PDGF-BB and AngII-induced reduction of SMA and SM22α and increment of OPN, suggesting that l-theanine inhibited the transformation of VSMCs from contractile to the synthetic phenotype. Further experiments showed that l-theanine exhibits potential preventive effects on neointimal hyperplasia and related vascular remodeling via inhibition of phosphorylation of Elk-1 and activation of MAPK1. The present study provides the new experimental evidence that l-theanine has potential clinical application as an anti-restenosis agent for the prevention of restenosis.


Subject(s)
Carotid Artery Injuries/pathology , Glutamates/pharmacology , Muscle, Smooth, Vascular/drug effects , Neointima/prevention & control , Animals , Becaplermin/pharmacology , Carotid Artery Injuries/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coronary Restenosis/prevention & control , Disease Models, Animal , Hyperplasia/drug therapy , Male , Mitogen-Activated Protein Kinase 1/metabolism , Myocytes, Smooth Muscle/drug effects , Neointima/pathology , Phenotype , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tea/chemistry , ets-Domain Protein Elk-1/metabolism
13.
Inflammation ; 43(3): 1157-1169, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32128658

ABSTRACT

Glutathione S-transferase Pi (GSTP1) was originally identified as one of the cytosolic phase II detoxification enzymes and was also considered to function via its non-catalytic, ligand-binding activity. Autophagy is a self-protective mechanism of the cell to remove unnecessary or dysfunctional components, which plays a crucial role in balancing the beneficial and detrimental effects of immunity and inflammation. However, little is known about whether and how GSTP1 mediates autophagy via inhibiting LPS-induced inflammatory response. Here, we show that LPS-induced autophagy and autophagic flux blockade in THP-1 cells in a concentration- and time-dependent manner. Further, we found that the autophagy activation inhibited the activation of inflammatory signaling pathway and the release of inflammatory factors. However, inhibition of autophagy by 3-methyladenine or chloroquine significantly reduced the anti-inflammatory effect of GSTP1. In addition, our findings provide evidence that GSTP1 regulates autophagy through PI3K-Akt-mTOR pathway and inhibits LPS-induced inflammation. Overall, the current study provides an important reference for future applications of GSTP1 in the treatment of inflammatory diseases.


Subject(s)
Autophagy/physiology , Glutathione S-Transferase pi/biosynthesis , Lipopolysaccharides/toxicity , THP-1 Cells/metabolism , Autophagy/drug effects , Dose-Response Relationship, Drug , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , THP-1 Cells/drug effects
14.
Front Immunol ; 11: 625542, 2020.
Article in English | MEDLINE | ID: mdl-33603756

ABSTRACT

CREB binding protein (CBP), a transcriptional coactivator and acetyltransferase, is involved in the pathogenesis of inflammation-related diseases. High mobility group box-1 protein (HMGB1) is a critical mediator of lethal sepsis, which has prompted investigation for the development of new treatment for inflammation. Here, we report that the potent and selective inhibition of CBP bromodomain by SGC-CBP30 blocks HMGB1-mediated inflammatory responses in vitro and in vivo. Our data suggest that CBP bromodomain inhibition suppresses LPS-induced expression and release of HMGB1, when the inhibitor was given 8 h post LPS stimulation; moreover, CBP bromodomain inhibition attenuated pro-inflammatory activity of HMGB1. Furthermore, our findings provide evidence that SGC-CBP30 down-regulated rhHMGB1-induced activation of MAPKs and NF-κB signaling by triggering the reactivation of protein phosphatase 2A (PP2A) and the stabilization of MAPK phosphatase 1 (MKP-1). Collectively, these results suggest that CBP bromodomain could serve as a candidate therapeutic target for the treatment of lethal sepsis via inhibiting LPS-induced expression and release of HMGB1 and suppressing the pro-inflammatory activity of HMGB1.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CREB-Binding Protein/antagonists & inhibitors , Down-Regulation/drug effects , HMGB1 Protein/immunology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Sepsis , Animals , CREB-Binding Protein/immunology , Down-Regulation/immunology , Humans , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Inbred BALB C , Sepsis/chemically induced , Sepsis/drug therapy , Sepsis/immunology , Sepsis/pathology , THP-1 Cells
15.
Cell Biol Int ; 44(1): 253-267, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31483551

ABSTRACT

Heat shock protein 27 (Hsp27) is a member of the small heat shock protein family expressed at high levels to protect cells against heat shock and other conditions of stress. Hsp27 has been indicated in the regulation of inflammation signaling pathway, and Hsp27 phosphorylation is vital for efficient control of host-defense response in early stages of lipopolysaccharide (LPS)-stimulated inflammation. The notion that CREB-binding protein (CBP) is involved in the regulation of two major transcription factors, nuclear factor-κB (NF-κB) and AP-1, suggests that CBP, as a coactivator protein, may also play an important role in the cellular response to inflammation. Here, we explored the mechanism underlying the regulatory relationships between Hsp27 and CBP in THP-1 cells, and found that phosphorylated Hsp27 was critical to the protein level of CBP. Furthermore, in exploring the signaling mechanisms underlying its action, we found that p38MAPK-MK2-Hsp27 regulated NF-κB via CBP, which acted as a multi-protein complex assembly scaffold. Finally, we demonstrated that phosphorylated Hsp27 reduced reactive oxygen species accumulation thereby significantly repressed LPS-induced excessive increase of CBP. Taken together, our data demonstrated that Hsp27, in its phosphorylation state, plays a critical role in controlling LPS-induced inflammatory response by modulating CBP.

16.
Mol Immunol ; 108: 45-55, 2019 04.
Article in English | MEDLINE | ID: mdl-30780033

ABSTRACT

Heat-shock protein 27 (Hsp27) is a member of the small heat shock protein family that has been reported to protect cells against pro-inflammatory stresses. High mobility group box 1 (HMGB1) is a proinflammatory cytokine associated with death from sepsis and other inflammatory diseases. After being acetylated by CREB-binding protein (CBP), the transcriptional adaptor and acetyltransferase, HMGB1 translocates from the nucleus to the cytoplasm. In the present study, we investigated the effects of Hsp27 on HMGB1 translocation from the nucleus to the cytoplasm in THP-1 cells. We found that Hsp27 phosphorylation decreased LPS-induced HMGB1 acetylation and translocation from the nucleus to the cytoplasm, as well as its release from THP-1 cells. The study further showed that cytosolic non-phosphorylated Hsp27 enhanced CBP ubiquitination and degradation in LPS-unstimulated cells, which suggested that Hsp27 maintained suitable CBP levels under normal physiological conditions. After LPS stimulation, Hsp27 was phosphorylated at serine residues 15/78 and translocated from the cytoplasm into the nucleus. Consequently, LPS stimulation increased CBP levels and promoted its translocation into the nucleus. In the nucleus, Hsp27 bound to CBP and suppressed CBP acetyltransferase activity and the subsequent CBP-dependent acetylation of HMGB1. Taken together, our data demonstrated that cytosolic non-phosphorylated Hsp27 enhanced the ubiquitin-mediated degradation of CBP, while phosphorylated Hsp27 inhibited CBP acetyltransferase activity in the nucleus. By regulating CBP, Hsp27 maintained cell homeostasis and inhibited excessive inflammatory response.


Subject(s)
CREB-Binding Protein/metabolism , HMGB1 Protein/metabolism , HSP27 Heat-Shock Proteins/metabolism , Ubiquitination , Acetylation , Cell Nucleus/metabolism , Histones/metabolism , Humans , Lipopolysaccharides , Models, Biological , Phosphorylation , Protein Transport , Proteolysis , THP-1 Cells , Ubiquitin/metabolism
17.
Cell Death Differ ; 26(10): 2086-2099, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30683915

ABSTRACT

Glutathione S-transferases P1 (GSTP1) is a phase II detoxifying enzyme and increased expression of GSTP1 has been linked with acquired resistance to anti-cancer drugs. However, most anticancer drugs are not good substrates for GSTP1, suggesting that the contribution of GSTP1 to drug resistances might not be dependent on its capacity to detoxify chemicals or drugs. In the current study, we found a novel mechanism by which GSTP1 protects human breast cancer cells from adriamycin (ADR)-induced cell death and contributes to the drug resistance. GSTP1 protein level is very low in human breast cancer cell line MCF-7 but is high in ADR-resistant MCF-7/ADR cells. Under ADR treatment, MCF-7/ADR cells showed a higher autophagy level than MCF-7 cells. Overexpression of GSTP1 in MCF-7 cells by using the DNA transfection vector enhanced autophagy and down-regulation of GSTP1 through RNA interference in MCF-7/ADR cells decreased autophagy. When autophagy was prevented, GSTP1-induced ADR resistance reduced. We found that GSTP1 enhanced autophagy level in MCF-7 cells through interacting with p110α subunit of phosphatidylinositol-3-kinase (PI3K) and then inhibited PI3K/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) activity. Proline123, leucine160, and glutamine163, which located in C terminal of GSTP1, are essential for GSTP1 to interact with p110α, and the following autophagy and drug resistance regulation. Taken together, our findings demonstrate that high level of GSTP1 maintains resistance of breast cancer cells to ADR through promoting autophagy. These new molecular insights provide an important contribution to our better understanding the effect of GSTP1 on the resistance of tumors to chemotherapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Doxorubicin/pharmacology , Glutathione S-Transferase pi/metabolism , Antibiotics, Antineoplastic/pharmacology , Autophagy/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Humans , MCF-7 Cells , Mass Spectrometry/methods , Microscopy, Electron, Transmission/methods , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transfection
18.
Cancer Lett ; 424: 84-96, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29524558

ABSTRACT

Non-small cell lung cancer (NSCLC) patients harboring EGFR-activating mutations initially respond to EGFR tyrosine kinase inhibitors (EGFR-TKIs) and have shown favorable outcomes. However, acquired drug resistance to EGFR-TKIs develops in almost all patients mainly due to the EGFR T790 M mutation. Here, we show that treatment with low-dose EGFR-TKI results in the emergence of the EGFR T790 M mutation and in the reduction of HSP70 protein levels in HCC827 cells. Erlotinib treatment inhibits HSP70 phosphorylation at tyrosine 41 and increases HSP70 ubiquitination, resulting in HSP70 degradation. We show that EGFR-TKI treatment causes increased DNA damage and enhanced gene mutation rates, which are secondary to the EGFR-TKI-induced reduction of HSP70 protein. Importantly, HSP70 overexpression delays the occurrence of Erlotinib-induced EGFR T790 M mutation. We further demonstrate that HSP70 interacts with multiple enzymes in the base excision repair (BER) pathway and promotes not only the efficiency but also the fidelity of BER. Collectively, our findings show that EGFR-TKI treatment facilitates gene mutation and the emergence of EGFR T790 M secondary mutation by the attenuation of BER via induction of HSP70 protein degradation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm , HSP72 Heat-Shock Proteins/metabolism , Lung Neoplasms/genetics , Protein Kinase Inhibitors/adverse effects , A549 Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , DNA Repair , ErbB Receptors/genetics , Erlotinib Hydrochloride/adverse effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mutation , Phosphorylation/drug effects , Proteolysis , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...