Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Biochem Genet ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649557

ABSTRACT

Breast cancer (BRCA) is currently the most commonly diagnosed malignancy in women worldwide. Previous studies have demonstrated that mitophagy is important for the prevention and treatment of BRCA. However, few studies have focused on the individual mitochondrial autophagy-related genes (MARG) in human cancers. Based on bioinformatics analyses, TOMM40 was identified as a prognostic DEMARG (PDEMARGs); Kaplan-Meier (KM) survival analysis also indicates that TOMM40 can be useful as a prognostic indicator in BRCAs, with patients in the high expression group having a poorer prognosis. For 20 distinct cancer kinds, there were appreciable differences in the expression of TOMM40 between tumor and normal tissues; in addition, in 21 different cancer types, there were associations between the expression profile of TOMM40 and patient prognosis. Gene Set Enrichment Analysis (GSEA), functional enrichment analysis, and immunological and drug sensitivity analyses of TOMM40 have indicated its biological significance in pan-cancers. Knockdown of TOMM40 in MDA-MB-231 cells inhibited their proliferation, migration, and invasiveness. In conclusion, we found that TOMM40 has prognostic value in 21 cancers, including breast cancer, by bioinformatics analysis. Based on immune correlation analysis, TOMM40 may also be a potential immunotherapeutic target for the treatment of BRCA. Therefore, our results may provide researchers to further explore the role of MARGs, especially TOMM40, in the developmental process of breast cancer, which may provide new directions and targets for the improvement of prognosis of breast cancer patients and their treatment.

2.
Breast Cancer ; 31(1): 135-147, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37981615

ABSTRACT

BACKGROUND: Triple-negative breast cancers (TNBC) are highly aggressive malignancies with poor prognosis. As an essential enzyme in the tryptophan-kynurenine metabolic pathway, indoleamine 2,3 dioxygenase-1 (IDO-1) has been reported to facilitate immune escape of various tumors. However, the mechanism underlying the immunosuppressive role of IDO-1 in TNBC remains largely uncharacterized. METHODS: We examined the IDO-1 expression in 93 clinical TNBC tissues and paired adjacent normal tissues, and analyzed the regulation role of environmental cytokines like IFN-γ in IDO-1 expression. The effect of IDO-1 expression in TNBC cells on the function of NK cells were then evaluated and the underlying mechanisms were exploited. RESULTS: IDO-1 expressed in 50 of 93 (54.1%) TNBC patients. TNBC patients with high IDO-1 expression tended to have more infiltrated immune cells including NK cells, which are less active than patients with low IDO-1 expression. NK cells could produce IFN-γ, which induced IDO-1 expression in TNBC cells, whereas IDO-1 impaired the cytotoxicity of co-cultured NK cells by upregulation of HLA-G. Blockade of HLA-G improved the antitumor activity of NK cells to TNBC in vivo. CONCLUSION: TNBC cells induce dysfunction of NK cells through an IFN-γ/IDO-1/HLA-G pathway, which provide novel insights into the mechanisms of TNBC progression and demonstrate the applicability of IDO-1 and HLA-G targeting in the treatment of TNBC.


Subject(s)
HLA-G Antigens , Triple Negative Breast Neoplasms , Humans , HLA-G Antigens/metabolism , HLA-G Antigens/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Killer Cells, Natural/metabolism , Triple Negative Breast Neoplasms/metabolism , Up-Regulation
3.
Heliyon ; 9(12): e23267, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38144329

ABSTRACT

Breast cancer (BRCA) is a common neoplasm characterized by high levels of molecular heterogeneity. Previous studies have noted the importance of mitophagy for the progression and prognosis of BRCA. However, little was found in the similarity and difference of mitophagy-related gene expression patterns of BRCA. This study intended to investigate the differences in functional activation, somatic mutation, and immune-related characteristics among different subtypes of BRCA associated with mitophagy. Based on bioinformatics analysis, we systematically examined the heterogeneity of breast cancer concerning mitophagy and observed two distinct subtypes with different tumor microenvironments and prognoses. BRCA samples from TCGA database were divided into two subtypes based on the expression of 29 mitophagy-related genes by ConsensusClusterPlus algorithm. Two mitophagy-related subtypes with marked prognostic discrepancies were significantly correlated with race, intrinsic subtype grouped based on PAM50 subtype purity and BRCA Pathology. The results of GSVA and immune microenvironment analysis showed significant differences in cancer-related and immune-related features between the two subtypes. METABRIC datasets were extracted to validate the immune characteristics scoring and the expression of immune checkpoints between different subtypes based on the medium value of TCGA-Mitophagy score. It is noteworthy that the present study is the first to demonstrate a new classification based on the mitophagy of breast cancer, which comes up with a new perspective for the assessment and prognoses of BRCA.

4.
Genes (Basel) ; 14(6)2023 05 26.
Article in English | MEDLINE | ID: mdl-37372337

ABSTRACT

Triple-negative breast cancer (TNBC) has a very poor prognosis due to the disease's lack of established targeted treatment options. Glia maturation factor γ (GMFG), a novel ADF/cofilin superfamily protein, has been reported to be differentially expressed in tumors, but its expression level in TNBC remains unknown. The question of whether GMFG correlates with the TNBC prognosis is also unclear. In this study, data from the Cancer Genome Atlas (TCGA), Clinical Proteomic Tumor Analysis Consortium (CPTAC), Human Protein Atlas (HPA), and Genotype-Tissue Expression (GTEx) databases were used to analyze the expression of GMFG in pan-cancer and the correlation between clinical factors. Gene Set Cancer Analysis (GSCA) and Gene Set Enrichment Analysis (GSEA) were also used to analyze the functional differences between the different expression levels and predict the downstream pathways. GMFG expression in breast cancer tissues, and its related biological functions, were further analyzed by immunohistochemistry (IHC), immunoblotting, RNAi, and function assay; we found that TNBC has a high expression of GMFG, and this higher expression was correlated with a poorer prognosis in TCGA and collected specimens of the TNBC. GMFG was also related to TNBC patients' clinicopathological data, especially those with histological grade and axillary lymph node metastasis. In vitro, GMFG siRNA inhibited cell migration and invasion through the EMT pathway. The above data indicate that high expression of GMFG in TNBC is related to malignancy and that GMFG could be a biomarker for the detection of TNBC metastasis.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Cell Movement/genetics , Prognosis , Proteomics , RNA Interference , Triple Negative Breast Neoplasms/pathology
6.
Pain Res Manag ; 2023: 9058774, 2023.
Article in English | MEDLINE | ID: mdl-36819745

ABSTRACT

Objective: Paclitaxel-induced peripheral neuropathy (PIPN) is a debilitating and difficult-to-treat side effect of paclitaxel. Soluble epoxide hydrolase (sEH) can rapidly metabolize the endogenous anti-inflammatory mediators' epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids. This study aimed to assess whether the sEH inhibitor N-(1-(1-oxopropy)-4-piperidinyl]-N'-(trifluoromethoxy) phenyl)-urea (TPPU) plays a critical role in PIPN of rats and provides a new target for treatment. Methods: A Sprague-Dawley male rat model of PIPN induced by nab-paclitaxel was established. Rats were randomly divided into a control group, nab-paclitaxel group, and nab-paclitaxel + TPPU (sEH inhibitor) group, with 36 rats in each group. The effects of the sEH inhibitor TPPU on behavioural assays, apoptosis, glial activation, axonal injury, microstructure, and permeability of the blood-spinal cord barrier were detected, and the underlying mechanisms were explored by examining the expression of NF-κB signalling pathways, inflammatory cytokines, and oxidative stress. Results: The results showed that the mechanical and thermal pain thresholds of rats were decreased after nab-paclitaxel treatment, accompanied by an increased expression of axonal injury-related proteins, enhanced cell apoptosis, aggravated destruction of vascular permeability, intense glial responses, and elevated inflammatory cytokines and oxidative stress in the L4-L6 spinal cord. TPPU restored the mechanical and thermal thresholds, decreased cell apoptosis, alleviated axonal injury and glial responses, and protected vascular permeability by increasing the expression of tight junction proteins. TPPU relieved PIPN by inhibiting the activation of the sEH and NF-κB signalling pathways by decreasing the levels of inflammatory cytokines and oxidative stress. Conclusion: These findings support a role for sEH in PIPN and suggest that the inhibition of sEH represents a potential new therapeutic target for PIPN.


Subject(s)
NF-kappa B , Neuralgia , Animals , Male , Rats , Cytokines/metabolism , Epoxide Hydrolases/metabolism , Paclitaxel , Rats, Sprague-Dawley , Spinal Cord/metabolism
7.
Neurosci Lett ; 793: 136975, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36427814

ABSTRACT

Chemotherapy results in long-term effects on cognitive dysfunction called chemotherapy-induced cognitive impairment (CICI) in cancer survivors. However, little is known about the potential molecular mechanisms of CICI. This study aimed to determine the role and potential underlying mechanisms of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in cognitive impairments induced by chemotherapeutic agents commonly used in breast cancer. The cognitive effects of chemotherapy were investigated in a rat model using the cocktail of doxorubicin and cyclophosphamide. The NLRP3 pathway was found to be differentially expressed after chemotherapy by iTRAQ-based proteomic analysis of normal and chemotherapeutic hippocampi. Treatment with the NLRP3 inhibitor MCC950 following chemotherapy significantly reduced cognitive impairment and decreased the expression of NLRP3, caspase-1 and ASC. Chemotherapy led to increased expression of the glial response markers Iba-1 and GFAP and the axonal injury markers NF-L and NF-M, an elevated number of apoptotic cells and enhanced microstructural damage to axons and mitochondria, while MCC950 treatment alleviated the glial response, cell death and axonal injury. The protective effect of MCC950 was related to the NLRP3 pathway and levels of inflammatory cytokines (TNF-α, IL-1ß, IL-18, IL-6, IL-4, and IL-10) and oxidative stress-responsive markers (SOD, MDA, CAT and GSH). The results indicate that CICI is associated with NLRP3 pathway-induced oxidative damage and the inflammatory response and provide a potential therapeutic target to treat cognitive impairment after chemotherapy (doxorubicin and cyclophosphamide).


Subject(s)
Chemotherapy-Related Cognitive Impairment , Cognitive Dysfunction , Indenes , Animals , Rats , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proteomics , Chemotherapy-Related Cognitive Impairment/drug therapy , Inflammasomes/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Furans/pharmacology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Doxorubicin/toxicity , Cyclophosphamide
8.
Int J Gen Med ; 15: 2607-2620, 2022.
Article in English | MEDLINE | ID: mdl-35282644

ABSTRACT

Introduction: Hepatocellular carcinoma (HCC) is one of several tumors with poor prognosis and causes a significant social burden. A growing number of studies have shown that RRM1 plays a crucial role in the development and progression of multiple human cancers. However, the specific role and mechanism of RRM1 have not been fully defined in HCC. Methods: TCGA and GTEx data were used for the first time to conduct a pan-cancer analysis of RRM1 expression and prognosis, and identified RRM1 as a possible potential oncogene in HCC. At the same time, a combination of analyses (including expression analysis, correlation analysis or survival analysis) identified non-coding RNAs (ncRNAs) that contribute to RRM1 overexpression. Results: MIR4435-2HG/miR-22-3p and SNHG6/miR-101-3p were identified as the most promising RRM1 upstream ncRNA-related pathways in HCC. In addition, RRM1 levels were significantly and positively correlated with tumor immune cell infiltration, immune cell biomarker or immune checkpoint expression. Conclusion: These results suggest that high expression of RRM1 mediated by ncRNAs is associated with poor prognosis and tumor immune infiltration in HCC.

9.
Signal Transduct Target Ther ; 6(1): 236, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34158475

ABSTRACT

Despite the successful use of the humanized monoclonal antibody trastuzumab (Herceptin) in the clinical treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer, the frequently occurring drug resistance remains to be overcome. The regulatory mechanisms of trastuzumab-elicited immune response in the tumor microenvironment remain largely uncharacterized. Here, we found that the nonclassical histocompatibility antigen HLA-G desensitizes breast cancer cells to trastuzumab by binding to the natural killer (NK) cell receptor KIR2DL4. Unless engaged by HLA-G, KIR2DL4 promotes antibody-dependent cell-mediated cytotoxicity and forms a regulatory circuit with the interferon-γ (IFN-γ) production pathway, in which IFN-γ upregulates KIR2DL4 via JAK2/STAT1 signaling, and then KIR2DL4 synergizes with the Fcγ receptor to increase IFN-γ secretion by NK cells. Trastuzumab treatment of neoplastic and NK cells leads to aberrant cytokine production characterized by excessive tumor growth factor-ß (TGF-ß) and IFN-γ, which subsequently reinforce HLA-G/KIR2DL4 signaling. In addition, TGF-ß and IFN-γ impair the cytotoxicity of NK cells by upregulating PD-L1 on tumor cells and PD-1 on NK cells. Blockade of HLA-G/KIR2DL4 signaling improved the vulnerability of HER2-positive breast cancer to trastuzumab treatment in vivo. These findings provide novel insights into the mechanisms underlying trastuzumab resistance and demonstrate the applicability of combined HLA-G and PD-L1/PD-1 targeting in the treatment of trastuzumab-resistant breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , HLA-G Antigens/genetics , Receptor, ErbB-2/genetics , Receptors, KIR2DL4/genetics , Trastuzumab/pharmacology , Adult , Aged , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Humans , Interferon-gamma/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Middle Aged , Receptors, Natural Killer Cell/genetics , Receptors, Natural Killer Cell/immunology , Trastuzumab/adverse effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
J Thorac Dis ; 13(2): 918-926, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33717564

ABSTRACT

BACKGROUND: The trachea is the uppermost respiratory airway element connecting the larynx to the bronchi Airway reconstructions in humans are often developed from animal models but there is limited knowledge comparing tracheal biomechanics between species. We aimed to assess the structure and biomechanics of porcine, canine, caprine and human airways. METHODS: Tracheas from pigs (n=15), goats (n=9) and canines (n=9) were divided into three groups (4, 6 and 8-ringswhile human left principal brochi (n=12) were divided into two groups (3and-rings). Airway structures were compared using histology and scanning electron microscopy. Biomechanical properties were measured subjecting samples to uniaxial tension and compression, recording the elastic modulus and (tensile and compressive) strengths. RESULTS: The structures of animal tracheal and human bronchia appeared similar. Biomechanical testing revealed that the elastic modulus of 8-ring tracheas was 1,190.48±363.68, 2,572.00±608.19 and 1,771.27±145.54 kPa, for porcine, canine and caprine samples, respectively, while corresponding tensile strengths were 437.63±191.41, 808.38±223.48 and 445.76±44.00 kPa. Comparable measures of anterior-posterior (A-P) compression strengths were 7.94±0.82, 7.54±0.07 and 8.10±1.87 N, respectively, whereas lateral compression strengths were 8.75±0.82, 14.55±2.29 and 11.12±0.40 N. Compression testing of human samples showed significant differences (P<0.05) between the 3-ring (A-P, 1.06±0.02 N; lateral, 0.55±0.06 N) and 5-ring groups (A-P, 1.08±0.64 N; lateral, 2.32±1.95 N). CONCLUSIONS: The tensile and compressive strengths of mammalian airways show positive correlations with the cartilage ring number (length). On the basis of structural and biomechanical comparisons, porcine, canine and caprine species appear suitable models for the study of airway reconstruction in human.

11.
Thorac Cancer ; 12(1): 21-29, 2021 01.
Article in English | MEDLINE | ID: mdl-33205914

ABSTRACT

BACKGROUND: The majority of previous studies of the clinical outcome of video-assisted thoracoscopic surgery (VATS) versus open lobectomy for pathological N2 non-small cell lung cancer (pN2 NSCLC) have been single-center experiences with small patient numbers. The aim of this study was therefore to investigate these procedures but in a large cohort of Chinese patients with pathological N2 NSCLC in real-world conditions. METHODS: Patients who underwent lobectomy for pN2 NSCLC by either VATS or thoracotomy were retrospectively reviewed from 10 tertiary hospitals between January 2014 and September 2017. Perioperative outcomes and overall survival of the patients were analyzed. Cox regression analysis was performed to identify potential prognostic factors. Propensity-score analysis was performed to reduce cofounding biases and compare the clinical outcomes between both groups. RESULTS: Among 2144 pN2 NSCLC, 1244 patients were managed by VATS and 900 by open procedure. A total of 305 (24.5%) and 344 patients died during VATS and the thoracotomy group during a median follow-up of 16.7 and 15.6 months, respectively. VATS lobectomy patients had better overall survival when compared with those undergoing the open procedure (P < 0.0001). Multivariate COX regression analysis showed VATS lobectomy independently favored overall survival (HR = 0.75, 95% CI: 0.621-0.896, P = 0.0017). Better perioperative outcomes, including less blood loss, shorter drainage time and hospital stay, were also observed in patients undergoing VATS lobectomy (P < 0.05). After propensity-score matching, 169 patients in each group were analyzed, and no survival difference were found between the two groups. Less blood loss was observed in the VATS group, but there was a longer operation time. CONCLUSIONS: VATS lobectomy might be a feasible alternative to conventional open surgery for resectable pN2 NSCLC. KEY POINTS: Significant findings of the study: VATS lobectomy has comparative OS in pN2 NSCLC versus open procedure in resectable patients. WHAT THIS STUDY ADDS: VATS lobectomy might be feasible for pN2 NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/surgery , Lung Neoplasms/surgery , Pneumonectomy/methods , Thoracic Surgery, Video-Assisted/methods , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Young Adult
12.
Biomed Res Int ; 2020: 5946205, 2020.
Article in English | MEDLINE | ID: mdl-32964038

ABSTRACT

OBJECTIVE: We have previously demonstrated that inflammation induced by toll-like receptors (TLRs) 2/4 exert cerebral deleterious effects after diffuse axonal injury (DAI); however, the underlying mechanisms are not fully understood. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine involved in inflammatory responses. The purpose of this study was to investigate the role of MIF in inflammation induced by TLRs in the cortices of DAI rats. METHODS: The rat DAI model was established by head rotational acceleration and confirmed by ß-APP, HE, and silver staining. MIF protein expression at 3 h, 6 h, 12 h, 1 d, and 3 d after DAI was measured by western blot. The localization of MIF was measured by immunofluorescence. MIF antagonist ISO-1 was intracerebroventricularly injected to inhibit MIF. Neuronal and axonal injury and glial responses were assessed by TUNEL, immunohistochemistry, and TEM. Expression of TLR2, TLR4, ERK, phospho-ERK, NF-κB, and phospho-NF-κB was examined by western blot. The level of IL-1ß, IL-6, and TNF-α was measured by ELISA. RESULTS: MIF expression was significantly increased, peaking at 1 day after DAI, and MIF was mainly localized in microglial cells and neurons. ISO-1 suppressed neuronal apoptosis, axonal injury, and glial responses and decreased the expression of downstream signaling molecules related to TLR2/4, including ERK, phospho-ERK, NF-κB, phospho-NF-κB, IL-1ß, IL-6, and TNF-α. CONCLUSION: MIF was involved in the neuronal and axonal damage through a TLR-related pathway following DAI.


Subject(s)
Diffuse Axonal Injury/metabolism , Inflammation/metabolism , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/metabolism , Toll-Like Receptors/metabolism , Animals , Apoptosis/physiology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Microglia/metabolism , NF-kappa B/metabolism , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
13.
J Pharm Biomed Anal ; 190: 113522, 2020 Oct 25.
Article in English | MEDLINE | ID: mdl-32777732

ABSTRACT

The evaluation of EGFR mutation genes in circulating tumor DNA (ctDNA) in blood sample is key for patients with lung cancer. Surface-enhanced Raman scattering (SERS) has potential for trace detection of DNA or RNA. The detection rate offered by current methods can not meet clinical demand. By combining asymmetric polymerase chain reaction (PCR) and SERS, a highly-selective detection for EGFR mutation genes in lung cancer was developed. Sea-urchin like Au nanoclusters (AuNCs) were synthesized via Ag seed-mediated growth. AuNCs with a diameter of 120 nm were covered with 79 nanopricks (20 nm). Then, EGFR mutation specific molecular beacons (MBs) labeled with Cy3 were coated on the surface of AuNCs. The loading amount of MBs was calculated as 5720 ± 740 on one AuNCs. These AuNCs probes had good efficiency (equilibrium time: 20 minutes) with high sensitivity (detection limit: 5.8 nM), high specificity (capable of single-base mismatch recognition) and good stability against nucleases. Following this, asymmetric PCR was performed to obtain large numbers of single-stranded DNA (ssDNA, E746-A750del). The ssDNA was incubated with the AuNCs probes and tested quantitatively based on the SERS signals of the AuNCs probes. This combined asymmetric PCR-SERS method had a very high detection threshold (4.24 fM). The asymmetric PCR-SERS method was shown to have an overall sensitivity of 75% and specificity of 100% in a further 15 clinical blood samples. This method is proved to be promising for non-invasive and sensitive detection of EGFR mutations in ctDNA.


Subject(s)
Lung Neoplasms , Spectrum Analysis, Raman , ErbB Receptors/genetics , Humans , Lung Neoplasms/genetics , Mutation , Polymerase Chain Reaction
14.
Neurosci Lett ; 736: 135234, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32721428

ABSTRACT

Blood-brain barrier (BBB) disruption exacerbates diffuse axonal injury (DAI), but the underlying mechanisms are not fully understood. Inactivation or deletion of erythropoietin-producing hepatoma (EPH) receptor A2 (EphA2) attenuated BBB damage and promoted tight junction formation. In this study, we aimed to investigate the role of EphA2 in the protection of BBB integrity and the relevant mechanisms involved in a rat model of DAI. Blocking activation of the EphA receptor by EphA2-Fc ameliorated axonal injury, cell apoptosis, and glial activation, protected BBB integrity and increased expression of the tight junction-associated proteins ZO-1, claudin-5 and occludin-1. In vitro BBB models established by human brain microvascular endothelial cells (HBMECs) were subjected to oxygen deprivation (OGD). Treatment with EphrinA1, which activates EphA2, exacerbated the OGD-induced destruction of permeability and integrity of the BBB models by reducing the expression of tight junction-associated proteins. However, inhibition of Rho-associated coiled coil-containing protein kinases 1 and 2 (ROCK1 and 2) abrogated all of the effects of EphrinA1 on the BBB models in vitro. In conclusion, we provide evidence that EphA2 plays an important role in the destruction of BBB integrity by decreasing the expression of tight junction proteins through the ROCK pathway.


Subject(s)
Blood-Brain Barrier/pathology , Diffuse Axonal Injury/pathology , Receptor, EphA2/metabolism , rho-Associated Kinases/metabolism , Animals , Blood-Brain Barrier/metabolism , Diffuse Axonal Injury/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Male , Rats , Rats, Sprague-Dawley
15.
Oxid Med Cell Longev ; 2020: 6841581, 2020.
Article in English | MEDLINE | ID: mdl-32566095

ABSTRACT

Persistent senescence seems to exert detrimental effects fostering ageing and age-related disorders, such as cancer. Chemotherapy is one of the most valuable treatments for cancer, but its clinical application is limited due to adverse side effects. Melatonin is a potent antioxidant and antiageing molecule, is nontoxic, and enhances the efficacy and reduces the side effects of chemotherapy. In this review, we first summarize the mitochondrial protective role of melatonin in the context of chemotherapeutic drug-induced toxicity. Thereafter, we tabulate the protective actions of melatonin against ageing and the harmful roles induced by chemotherapy and chemotherapeutic agents, including anthracyclines, alkylating agents, platinum, antimetabolites, mitotic inhibitors, and molecular-targeted agents. Finally, we discuss several novel directions for future research in this area. The information compiled in this review will provide a comprehensive reference for the protective activities of melatonin in the context of chemotherapy drug-induced toxicity and will contribute to the design of future studies and increase the potential of melatonin as a therapeutic agent.


Subject(s)
Aging/pathology , Antineoplastic Agents/adverse effects , Melatonin/therapeutic use , Neoplasms/drug therapy , Animals , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Protective Agents/pharmacology , Protective Agents/therapeutic use
16.
Article in English | MEDLINE | ID: mdl-32373063

ABSTRACT

Aim: Melatonin is an indolamine secreted by the pineal gland, as well as most of the organs and tissues. In addition to regulating circadian biology, studies have confirmed the multiple pharmacological effects of melatonin. Melatonin provides a strong defense against septic myocardial injury. However, the underlying mechanism has not been fully described. In this study, we investigated the protective effects of melatonin against lipopolysaccharide (LPS)-induced myocardial injury as well as the mechanisms involved. Methods: Mice were intraperitoneally injected with LPS to induce a septic myocardial injury model or an LPS shock model, depending on the dose of LPS. Melatonin was given (20 mg/kg/day, via intraperitoneal injection) for a week prior to LPS insult. 6 h after LPS injection, echocardiographic analysis, TUNEL staining, transmission electron microscopy (TEM), western blot, quantitative real-time PCR and ELISA were used to investigate the protective effects of melatonin against LPS induced myocardial injury. AMPK inhibitor, autophagy activator and inhibitor, siRNAs were used for further validation. Results: Survival test showed that melatonin significantly increased the survival rate after LPS-induced shock. In the sepsis model, melatonin markedly ameliorated myocardial dysfunction, decreased the release of inflammatory cytokines, activated AMP-activated protein kinase (AMPK), improved mitochondrial function, and activated autophagy. To confirm whether the protection of melatonin was mediated by AMPK and autophagy, Compound C, an AMPK inhibitor; 3-MA, an autophagy inhibitor; and Rapamycin (Rapa), an autophagy activator, were used in this study. AMPK inhibition down-regulated autophagy, abolished protection of melatonin, as indicated by significantly decreased cardiac function, increased inflammation and damaged mitochondrial function. Furthermore, autophagy inhibition by 3-MA significantly impaired the protective effects of melatonin, whereas autophagy activation by Rapa reversed LPS + Compound C induced myocardial injury. In addition, in vitro studies further confirmed the protection of melatonin against LPS-induced myocardial injury and the mechanisms involving AMPK-mediated autophagy signaling. Conclusions: In summary, our results demonstrated that melatonin protects against LPS-induced septic myocardial injury by activating AMPK mediated autophagy pathway.


Subject(s)
Cardiomyopathies/prevention & control , Cardiotonic Agents/pharmacology , Lipopolysaccharides/toxicity , Melatonin/pharmacology , Sepsis/chemically induced , Sepsis/complications , Animals , Cardiomyopathies/etiology , Cells, Cultured , Heart/drug effects , Heart/physiology , Male , Melatonin/therapeutic use , Mice , Mice, Inbred C57BL , Myocardium/pathology , Rats , Shock, Septic/mortality , Shock, Septic/pathology , Shock, Septic/prevention & control
17.
Onco Targets Ther ; 12: 10299-10309, 2019.
Article in English | MEDLINE | ID: mdl-31819514

ABSTRACT

PURPOSE: High metastasis is a leading risk factor for the survival of non-small cell lung cancer (NSCLC) and epithelial-mesenchymal transition (EMT) is a vital step of metastasis. The expression of novel oncogene with kinase domain (NOK) has been observed in some human malignancies, including non-small cell lung cancer (NSCLC); however, the biological function of NOK in NSCLC remains unclear. In the study, we explored the function of NOK in NSCLC, with an aim to elucidate the relevant underlying mechanisms. PATIENTS AND METHODS: We investigate the expression of NOK, p-Akt, p-GSK-3ß, E-cadherin and N-cadherin expression by immunohistochemical analysis using tissue microarrays of 72 paired NSCLC samples of cancerous and adjacent normal tissues. The associations between NOK expression and clinicopathological factors, overall survival, other proteins were assessed. Immunofluorescence analysis of NSCLC tissues was performed to study the location of NOK, Akt and GSK-3ß. Up or down-regulated of NOK were conducted in two NSCLC cell lines to analyze its impact on AKT/GSK3ß pathway. RESULTS: Statistical analysis revealed NOK expression increased in NSCLC tissues compared with normal tissues (P<0.05). It also showed that low NOK expression were associated with a higher possibility of non-lymphatic metastasis, an early pN stage and clinical stage (P<0.05). Moreover, NOK expression was positively correlated with the expression of oncogene p-Akt (Thr308), p-GSK-3ß (Ser9) and N-cadherin (P<0.05). Immunofluorescence analysis of NSCLC tissues revealed that NOK is co-located with Akt and GSK-3ß. Further study in NSCLC cell lines revealed that NOK overexpression can activate the AKT/GSK3ß pathway. Conversely, knockdown of NOK can suppress the AKT/GSK3ß pathway. CONCLUSION: Our results suggest that NOK overexpression correlated significantly with lymphatic metastasis, advanced pN and clinical stage in NSCLC. And NOK may promote EMT by activating the AKT/GSK3ß/N-cadherin pathway in NSCLC.

18.
Biochem Biophys Res Commun ; 519(4): 659-666, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31542233

ABSTRACT

Serine Threonine Tyrosine Kinase 1 (STYK1) presents oncogenic properties in many studies, and emerging evidence suggests that ferroptosis serve as a novel tumor suppressor. However, the interplay between STYK1 and ferroptosis in NSCLC remains unclear. Our aim is to illustrate the expression of ferroptotic regulator Glutathione peroxidase 4 (GPX4) in NSCLC and the relationship between STYK1 and ferroptosis. Herein, results based on ONCOMINE database, clinical specimens, and cellular manipulation revealed GPX4 was upregulated in NSCLC tissues and cell lines, and high GPX4 expression predicted worse prognosis. High STYK1 expression predicted worse OS and was related to high GPX4 in NSCLC tissues; overexpression of STYK1 in lung cancer cell line SW900 upregulated the expression of GPX4, promoted proliferation, and attenuated diverse mitochondrial abnormalities specific to ferroptosis, whereas knockdown of GPX4 exacerbated such attenuations without affecting cell proliferation. Taken together, ferroptosis as an anti-tumor factor is inhibited in NSCLC, and targeting ferroptosis could be a novel therapeutic strategy for the management of NSCLC; furthermore, regulating ferroptosis could be another cancerous mechanism of STYK1.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Ferroptosis/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , A549 Cells , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Survival Analysis , Up-Regulation
19.
Cell Death Dis ; 10(6): 435, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31164631

ABSTRACT

Non-small cell lung cancer (NSCLC) is the leading cause of cancer deaths worldwide. However, the molecular mechanisms underlying NSCLC progression remains not fully understood. In this study, 347 patients with complete clinicopathologic characteristics who underwent NSCLC surgery were recruited for the investigation. We verified that elevated serine threonine tyrosine kinase 1 (STYK1) or decreased serine peptidase inhibitor Kunitz type 2 (SPINT2/HAI-2) expression significantly correlated with poor prognosis, tumor invasion, and metastasis of NSCLC patients. STYK1 overexpression promoted NSCLC cells proliferation, migration, and invasion. STYK1 also induced epithelial-mesenchymal transition by E-cadherin downregulation and Snail upregulation. Moreover, RNA-seq, quantitative polymerase chain reaction (qRT-PCR), and western blot analyses confirmed that STYK1 overexpression significantly decreased the SPINT2 level in NSCLC cells, and SPINT2 overexpression obviously reversed STYK1-mediated NSCLC progression both in vitro and in vivo. Further survival analyses showed that NSCLC patients with high STYK1 level and low SPINT2 level had the worst prognosis and survival. These results indicated that STYK1 facilitated NSCLC progression via reducing SPINT2 expression. Therefore, targeting STYK1 and SPINT2 may be a novel therapeutic strategy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Middle Aged , Prognosis , RNA-Seq , Receptor Protein-Tyrosine Kinases/genetics , Snail Family Transcription Factors/metabolism , Tissue Array Analysis , Transplantation, Heterologous
20.
Pharmacol Res ; 145: 104265, 2019 07.
Article in English | MEDLINE | ID: mdl-31078746

ABSTRACT

Pterostilbene, a natural dimethylated analog of resveratrol, exerts pleiotropic anticancer effects against a variety of cancer types. Due to the better lipophilic and oral absorption, higher cellular uptake and a longer half-life than resveratrol, pterostilbene may have a good prospect in the future clinic application. In this review, we summarize the previous in vitro and in vivo studies about the anticancer actions of pterostilbene on malignances, and we also evaluate the evidence related to the effects of pterostilbene on blocking normal cell carcinogenesis. Special focus is placed on the oncostatic effects of pterostilbene, including inhibition of tumor growth, metastasis, angiogenesis and cancer stem cells, activation of apoptosis, and enhancement of immunotherapy. We then clarify the emerging investigations about pterostilbene and chemotherapy and radiotherapy. Taken together, the information complied herein may serve as a comprehensive reference for the anticancer mechanisms of pterostilbene and may advance it as a future adjuvant therapeutic agent for cancer.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Stilbenes/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Combined Modality Therapy , Humans , Immunotherapy , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/radiotherapy , Neoplastic Stem Cells/drug effects , Neovascularization, Pathologic/drug therapy , Stilbenes/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...