Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Vet Res ; 55(1): 42, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575961

ABSTRACT

African Swine Fever virus (ASFV), the causative agent of African swine fever, is a highly lethal hemorrhagic virus affecting domestic pigs and wild boars. The primary target cells for ASFV infection are porcine alveolar macrophages (PAMs), which are difficult to obtain and maintain in vitro, and less subjective to genetic editing. To overcome these issues and facilitate ASFV research, we obtained a subclonal cell line PK1-C5 by subcloning LLC-PK1 cells that support stable ASFV proliferation. This consequential cell line exhibited high ASFV infection levels and similar viral growth characteristics to PAMs, while also allowing high-efficiency genomic editing through transfection or lentivirus transduction of Cas9. Taken together, our study provided a valuable tool for research aspects including ASFV-host interactions, pathogenicity, and vaccine development.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine Diseases , Swine , Animals , African Swine Fever Virus/genetics , Sus scrofa , Cell Line , Kidney
2.
Appl Opt ; 63(10): 2621-2629, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38568545

ABSTRACT

When a binocular vision sensor (BVS) is installed in a narrow space, traditional calibration methods are limited as all targets should be placed in more than three different positions. To solve this problem, an on-site calibration method based on the phase-shift algorithm is proposed in our paper. Intrinsic parameters of these two cameras should be first calibrated offline. Series of phase-shift patterns are projected onto any one target with known three-dimensional information to determine the relationship between two cameras. The target utilized in our proposed method can be selected arbitrarily, which is suitable to achieve the on-site calibration of BVS, especially in industrial vibration environments. Experiments are conducted to validate the effectiveness and robustness of our proposed method.

3.
Int Orthop ; 48(4): 1011-1016, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37819386

ABSTRACT

PURPOSE: We explored the risk factors for avascular necrosis (AVN) after surgery using open reduction, pelvic osteotomy, and femoral osteotomy for Tönnis grade IV developmental dysplasia of the hip (DDH). METHODS: In this retrospective study, we collected data of patients with Tönnis grade IV DDH treated with open reduction and pelvic osteotomy combined with femoral osteotomy from January 2012 to May 2020. The patients were divided into the AVN group and non-AVN group using the Kalamchi-MacEwen classification system. The clinical and imaging data of the two groups were collected, and the possible risk factors were included in the analysis. Univariate and multivariate logistic regression analyses were used to identify the independent risk factors and odds ratios of AVN. RESULTS: In all, 254 patients (mean age; 2.6±0.9 years, 278 hips) were included. The mean follow-up time was 3.8±1.5 years. A total of 89 hips (32%) were finally classified as AVN (Kalamchi-MacEwen II-IV). Univariate analysis showed significant associations with AVN for age (p=0.006), preoperative femoral neck anteversion (FAV) (p<0.001), femoral osteotomy length to dislocation height ratio (FDR) <1 (p<0.001), and the epiphyseal ossific nucleus diameter to the neck diameter ratio (ENR) <50% (p=0.009). Multivariate logistic regression analysis showed that only excessive preoperative FAV (OR: 1.04; 95% CI: 1.02-1.05; p<0.001) and FDR<1 (OR: 3.58; 95% CI: 2.03-6.31; p<0.001) were independent risk factors for femoral head necrosis. CONCLUSION: Excessive preoperative FAV and FDR<1 are important risk factors for femoral AVN after open reduction, pelvic osteotomy, and femoral osteotomy for Tönnis grade IV DDH. For children with DDH with high dislocation and excessive FAV, clinicians should fully evaluate their condition and design more personalized treatment programs to prevent AVN.


Subject(s)
Developmental Dysplasia of the Hip , Femur Head Necrosis , Hip Dislocation, Congenital , Joint Dislocations , Osteonecrosis , Child , Humans , Infant , Retrospective Studies , Hip Dislocation, Congenital/diagnostic imaging , Developmental Dysplasia of the Hip/complications , Developmental Dysplasia of the Hip/surgery , Radiography , Osteonecrosis/complications , Osteotomy/adverse effects , Risk Factors , Joint Dislocations/etiology , Necrosis/complications , Femur Head Necrosis/epidemiology , Femur Head Necrosis/etiology , Femur Head Necrosis/prevention & control , Treatment Outcome
4.
Int J Mol Sci ; 24(23)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38069424

ABSTRACT

Swine acute diarrhea syndrome coronavirus (SADS-CoV), a member of the family Coronaviridae and the genus Alphacoronavirus, primarily affects piglets under 7 days old, causing symptoms such as diarrhea, vomiting, and dehydration. It has the potential to infect human primary and passaged cells in vitro, indicating a potential risk of zoonotic transmission. In this study, we successfully generated and purified six monoclonal antibodies (mAbs) specifically targeting the spike protein of SADS-CoV, whose epitope were demonstrated specificity to the S1A or S1B region by immunofluorescence assay and enzyme-linked immunosorbent assay. Three of these mAbs were capable of neutralizing SADS-CoV infection on HeLa-R19 and A549. Furthermore, we observed that SADS-CoV induced the agglutination of erythrocytes from both humans and rats, and the hemagglutination inhibition capacity and antigen-antibody binding capacity of the antibodies were assessed. Our study reveals that mAbs specifically targeting the S1A domain demonstrated notable efficacy in suppressing the hemagglutination phenomenon induced by SADS-CoV. This finding represents the first instance of narrowing down the protein region responsible for SADS-CoV-mediated hemagglutination to the S1A domain, and reveals that the cell attachment domains S1A and S1B are the main targets of neutralizing antibodies.


Subject(s)
Alphacoronavirus , Swine Diseases , Rats , Animals , Humans , Swine , Spike Glycoprotein, Coronavirus/chemistry , Antibodies, Monoclonal , Antibodies, Neutralizing/metabolism
5.
Molecules ; 28(14)2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37513469

ABSTRACT

Lithium-oxygen batteries have attracted considerable attention in the past several years due to their ultra-high theoretical energy density. However, there are still many serious issues that must be addressed before considering practical applications, including the sluggish oxygen redox kinetics, the limited capacity far from the theoretical value, and the poor cycle stability. This study proposes a surface modification strategy that can enhance the catalytic activity by loading Fe3C particles on carbon fibers, and the microstructure of Fe3C particle-modified carbon fibers is studied by multiple materials characterization methods. Experiments and density functional theory (DFT) calculations show that the discharge products on the Fe3C carbon fiber (Fe3C-CF) cathode are mainly Li2-xO2. Fe3C-CF exhibits high catalytic ability based on its promotion of the formation/decomposition processes of Li2-xO2. Consequently, the well-designed electrode catalyst exhibits a large specific capacity of 17,653.1 mAh g-1 and an excellent cyclability of 263 cycles at a current of 200 mA g-1.

6.
Mol Cancer ; 21(1): 97, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35395767

ABSTRACT

BACKGROUND: N6-methyladenosine (m6A) RNA modification plays a critical role in various physiological and pathological conditions. However, the role of m6A modification in head and neck squamous cell carcinoma (HNSCC) remains elusive. METHODS: In this study, the expression of m6A demethylases was detected by HNSCC tissue microarray. m6A-RNA immunoprecipitation (MeRIP) sequencing and RNA sequencing were used to identify downstream targets of ALKBH5. Comprehensive identification of RNA-binding proteins by mass spectrometry (ChIRP-MS) was used to explore the m6A "readers". Tumor-infiltrating lymphocytes were analyzed in SCC7-bearing xenografts in C3H mice. RESULTS: Here, we demonstrate the downregulation of m6A status and upregulation of two demethylases in HNSCC. Silencing the m6A demethylase alkB homolog 5, RNA demethylase (ALKBH5) suppresses tumor progression in vitro and in vivo. m6A-RNA immunoprecipitation sequencing reveals that ALKBH5 downregulates the m6A modification of DDX58 mRNA. Moreover, RIG-I, encoded by the DDX58 mRNA, reverses the protumorigenic characteristics of ALKBH5. ChIRP-MS demonstrates that HNRNPC binds to the m6A sites of DDX58 mRNA to promote its maturation. ALKBH5 overexpression inhibits RIG-I-mediated IFNα secretion through the IKKε/TBK1/IRF3 pathway. The number of tumor-infiltrating lymphocytes in C3H immunocompetent mice is reduced by ALKBH5 overexpression and restored by IFNα administration. Upregulation of AKLBH5 negatively correlates with RIG-I and IFNα expression in HNSCC patients. CONCLUSIONS: These findings unveil a novel mechanism of immune microenvironment regulation mediated by m6A modification through the ALKBH5/RIG-I/IFNα axis, providing a rationale for therapeutically targeting epitranscriptomic modulators in HNSCC.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Head and Neck Neoplasms , I-kappa B Kinase , Squamous Cell Carcinoma of Head and Neck , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Animals , DEAD Box Protein 58 , Head and Neck Neoplasms/genetics , Humans , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-alpha , Mice , Mice, Inbred C3H , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Tumor Microenvironment
7.
BMC Med ; 20(1): 231, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35773668

ABSTRACT

BACKGROUND: Cisplatin resistance is one of the main causes of treatment failure and death in head and neck squamous cell carcinoma (HNSCC). A more comprehensive understanding of the cisplatin resistance mechanism and the development of effective treatment strategies are urgent. METHODS: RNA sequencing, RT-PCR, and immunoblotting were used to identify differentially expressed genes associated with cisplatin resistance. Gain- and loss-of-function experiments were performed to detect the effect of CREB5 on cisplatin resistance and mitochondrial apoptosis in HNSCC. Chromatin immunoprecipitation (ChIP) assay, dual-luciferase reporter assay, and immunoblotting experiments were performed to explore the underlying mechanisms of CREB5. RESULTS: CREB5 was significantly upregulated in cisplatin-resistant HNSCC (CR-HNSCC) patients, which was correlated with poor prognosis. CREB5 overexpression strikingly facilitated the cisplatin resistance of HNSCC cells in vitro and in vivo, while CREB5 knockdown enhanced cisplatin sensitivity in CR-HNSCC cells. Interestingly, the activation of AKT signaling induced by cisplatin promoted nucleus translocation of CREB5 in CR-HNSCC cells. Furthermore, CREB5 transcriptionally activated TOP1MT expression depending on the canonical motif. Moreover, CREB5 silencing could trigger mitochondrial apoptosis and overcome cisplatin resistance in CR-HNSCC cells, which could be reversed by TOP1MT overexpression. Additionally, double-targeting of CREB5 and TOP1MT could combat cisplatin resistance of HNSCC in vivo. CONCLUSIONS: Our findings reveal a novel CREB5/TOP1MT axis conferring cisplatin resistance in HNSCC, which provides a new basis to develop effective strategies for overcoming cisplatin resistance.


Subject(s)
Antineoplastic Agents , Cisplatin , Cyclic AMP Response Element-Binding Protein A , DNA Topoisomerases, Type I , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Cyclic AMP Response Element-Binding Protein A/metabolism , DNA Topoisomerases, Type I/genetics , Drug Resistance, Neoplasm , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics
8.
Cell Mol Biol (Noisy-le-grand) ; 68(6): 167-173, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-36227659

ABSTRACT

To investigate the changes in CT4+ and CT8+ lymphocyte subpopulations of patients with non-small cell lung carcinoma (NSCLC) by monomethoxy polyethylene glycol-hyaluronic acid-platinum (MPEG-HA-Pt) and the correlation between efficacy evaluation and the changes in T lymphocyte subpopulation, 76 NSCLC patients treated at oncology department of Chengdu First People's Hospital were selected and randomly divided into the treatment group and the control group (38 cases in each). mPEG-HA-Pt was used for the treatment of the included patients in the research. The patients in the control group were performed with traditional chemotherapy for 2 treatment courses. The changes in the T-lymphocyte subpopulation before and after the treatment were detected and the therapeutic effects on the patients in the two groups were compared. The particle size of mPEG-HA-Pt ranged between 78nm and 100nm with an average of 84.6±7.5nm. After that, a transmission electron microscope (TEM) was used to observe the spheres with uniform size. The drug loading capacity and entrapped efficiency of mPEG-HA-Pt were 18.7% and 87.4%, respectively. After the treatment for NSCLC patients by nanomicelle, cellular immune functions were all improved. In particular, cellular immune functions of the patients with good efficacy evaluation were improved more apparently.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Hyaluronic Acid/therapeutic use , Immunotherapy , Lung Neoplasms/drug therapy , Platinum/therapeutic use , Polyethylene Glycols/therapeutic use
9.
Oral Dis ; 2022 Aug 23.
Article in English | MEDLINE | ID: mdl-35997137

ABSTRACT

OBJECTIVE: This study aimed to determine whether the RNA, 5-methylcytosine (m5C), is involved in the progression of head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS: We used least absolute shrinkage and selection operator to establish a prognostic score (PS) model based on the m5C regulator expression. Immune scores were calculated using the estimation of stromal and immune cells in malignant tumor tissues using expression data. The biological functions of the m5C regulator, NOP2/Sun RNA methyltransferase 3 (NSUN3), were thoroughly investigated in vitro and in vivo. RESULTS: The PS model acted as efficient prognostic factors in HNSCC. The expression of NSUN3, with the maximum weight, was found to be upregulated and indicated a poor prognosis. Meanwhile, NSUN3 knockdown inhibited the tumor proliferation and growth both in vitro and in vivo. High PS status was negatively correlated with CD8+ T, γδ+ T, and M1 macrophage percentages. NSUN3 knockdown increased the infiltration of M1 macrophages but decreased the percentage of M2 macrophages. CONCLUSIONS: The PS index is a novel and promising biomarker for predicting the prognosis and immune infiltration microenvironment in HNSCC. Moreover, NSUN3 plays a key role in this process and may serve as a potential therapeutic target for HNSCC.

10.
J Org Chem ; 86(2): 1549-1559, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33372795

ABSTRACT

The follow-up reaction pathways of the diradical species formed from cycloaromatization of enediynes or enyne-allenes determine their ability of H-abstraction from DNA, significantly affecting their biological activity performance. To gain a deeper understanding of subsequent reaction pathways of the diradical intermediates formed from acyclic enediynes based on maleimide-assisted rearrangement and cycloaromatization (MARACA), a maleimide-based enediyne featuring methylene groups adjacent to the propargyl sites of the terminal alkynes was synthesized through the Sonogashira coupling reaction. Three thermal cyclization products after intramolecular hydrogen atom transfer (HAT) were obtained from the thermolysis experiment and their structures were confirmed by 1D and 2D nuclear magnetic resonance spectroscopic analysis. Density functional theory was employed to analyze the important elementary steps including rearrangement, cycloaromatization, and intramolecular HAT processes toward the formation of the cyclized products, where the low-energy barriers of HAT pathways relative to the formation of diradicals from cycloaromatization were successfully identified. Overall, the HAT processes to consume diradicals intramolecularly have become competitive with that of intermolecular H-abstraction, implying that the DNA-cleavage ability of enediynes can be further boosted once the HAT processes are halted. This study offers a promising direction for designing novel and potent acyclic enediynes for antitumor applications.

11.
Arch Virol ; 166(5): 1355-1370, 2021 May.
Article in English | MEDLINE | ID: mdl-33709216

ABSTRACT

Porcine teschovirus (PTV) is a causative agent of reproductive disorders, encephalomyelitis, respiratory diseases, and diarrhea in swine, with a worldwide distribution. In this work, we identified PTV-associated nonsuppurative encephalitis as a potential cause of posterior paralysis in neonatal pigs in northeast China. Using indirect immunofluorescence assay, western blot, electron microscopy, and genome sequencing, we identified a neurotropic PTV strain, named CHN-NP1-2016, in the supernatants of pooled cerebrum and cerebellum samples from an affected piglet. Nucleotide sequence alignment revealed that the whole genome of CHN-NP1-2016 shared the highest sequence similarity (86.76% identity) with PTV 1 strain Talfan. A combination of phylogenetic and genetic divergence analysis was applied based on the deduced amino acid sequence of the P1 gene with a cutoff value of the genetic distance (0.102 ± 0.008) for defining PTV genotypes, and this showed that CHN-NP1-2016 is a variant of genotype 1. In total, 16 unique mutations and five mutant clusters were detected in the capsid proteins VP1 and VP2 of CHN-NP1-2016 when compared to other PTV1 isolates. Importantly, we detected three mutant clusters located in the exposed surface loops of the capsid protein, potentially indicating significant differences in major neutralization epitopes. Moreover, a potential recombination event in the P1 region of PTV CHN-NP1-2016 was detected. These findings provide valuable insights into the role of recombination in the evolution of teschoviruses. To our knowledge, this is the first case report of PTV-1-associated encephalitis in northeast China. Future investigations will narrow on the serology and pathogenicity of this novel isolate.


Subject(s)
Encephalitis, Viral/veterinary , Picornaviridae Infections/veterinary , Swine Diseases/virology , Teschovirus/genetics , Teschovirus/isolation & purification , Animals , Brain/virology , China/epidemiology , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Genome, Viral/genetics , Genotype , Mutation , Phylogeny , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , RNA, Viral/genetics , Recombination, Genetic , Swine , Teschovirus/classification , Viral Proteins/genetics
12.
Nanotechnology ; 33(7)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34171849

ABSTRACT

Transition metal carbides and nitrides (MXenes), as a large family of emerging two-dimensional materials, have demonstrated extraordinary performance in many fields such as electronics, optics and energy storage. However, their susceptibility to oxidation during preparation and storage in ambient air environment is undesirable for long-term and stable applications. Here, we have demonstrated that the spontaneous oxidation of Ti3C2Txcan be harnessed ingeniously to prepare Ti3C2Tx/TiO2in situformed heterojunctions. Furthermore, a self-powered ultraviolet photodetector was constructed based on the photoelectrochemical performance of Ti3C2Tx/TiO2heterojunctions. Since the highly conductive Ti3C2Txcan promote the separation and transfer of photogenerated carriers in TiO2, the prepared photodetector exhibits high responsivity (2.06 mA W-1), short rise and decay times (45 and 69 ms) and long-term stability. This work demonstrates the controllable synthesis of Ti3C2Tx/TiO2heterojunctions and provides a new promising potential of MXenes for photodetection applications.

13.
Mol Cancer ; 19(1): 4, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31907020

ABSTRACT

BACKGROUND: Interferon alpha (IFNα) is a well-established regulator of immunosuppression in head and neck squamous cell carcinoma (HNSCC), while the role of long noncoding RNAs (lncRNAs) in immunosuppression remains largely unknown. METHODS: Differentially expressed lncRNAs were screened under IFNα stimulation using lncRNA sequencing. The role and mechanism of lncRNA in immunosuppression were investigated in HNSCC in vitro and in vivo. RESULTS: We identified a novel IFNα-induced upregulated lncRNA, lncMX1-215, in HNSCC. LncMX1-215 was primarily located in the cell nucleus. Ectopic expression of lncMX1-215 markedly inhibited expression of the IFNα-induced, immunosuppression-related molecules programmed cell death 1 ligand 1 (PD-L1) and galectin-9, and vice versa. Subsequently, histone deacetylase (HDAC) inhibitors promoted the expression of PD-L1 and galectin-9. Binding sites for H3K27 acetylation were found on PD-L1 and galectin-9 promoters. Mechanistically, we found that lncMX1-215 directly interacted with GCN5, a known H3K27 acetylase, to interrupt its binding to H3K27 acetylation. Clinically, negative correlations between lncMX1-215 and PD-L1 and galectin-9 expression were observed. Finally, overexpression of lncMX1-215 suppressed HNSCC proliferation and metastasis capacity in vitro and in vivo. CONCLUSIONS: Our results suggest that lncMX1-215 negatively regulates immunosuppression by interrupting GCN5/H3K27ac binding in HNSCC, thus providing novel insights into immune checkpoint blockade treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Histones/metabolism , Interferon-alpha/pharmacology , RNA, Long Noncoding/genetics , Squamous Cell Carcinoma of Head and Neck/secondary , Acetylation , Animals , Antiviral Agents/pharmacology , Apoptosis , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Galectins/genetics , Galectins/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Histones/chemistry , Histones/genetics , Humans , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Nude , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Transcriptional Activation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
14.
Cancer Sci ; 111(9): 3222-3235, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32589311

ABSTRACT

Receptor tyrosine kinases (RTKs) and Yes-associated protein (YAP) are critical driving factors in tumors. Currently, the regulation of RTKs in the Hippo-YAP pathway has been recognized as an important issue. However, the relationship between AXL, one of the RTKs, and YAP in head and neck squamous cell carcinoma (HNSCC) remains unknown. In this study, the crosstalk between AXL and YAP was thoroughly investigated in vitro and in vivo. We determined that there was a positive correlation between AXL and YAP in the HNSCC tissue samples and the Cancer Genome Atlas (TCGA) dataset, and high co-expression was associated with poor prognosis. Inhibiting YAP decreased AXL expression in HNSCC cells, while YAP overexpression increased AXL. Moreover, ectopic expression of AXL reversed tumor suppressor phenotypes mediated by YAP silencing. This reversal effect was also confirmed in vivo. In addition, AXL overexpression and Gas6, a ligand of AXL, stimulated YAP dephosphorylation, nuclear translocation, and target gene transcription. AXL inhibition decreased YAP dephosphorylation and nuclear translocation. Mechanistically, Gas6 induced a competitive binding to phosphorylated signal transducers and activators of transcription 3 (STAT3) with large tumor suppressor kinase 1 (LATS1) and inhibited the Hippo pathway. This study revealed a novel non-transcriptional effect of STAT3 in Gas6/AXL-induced YAP activity, suggesting that STAT3 acted as a critical "molecular switch" during the mutual promotion between AXL and YAP, which might be a promising therapeutic target in HNSCC.


Subject(s)
Cell Cycle Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Silencing , Heterografts , Hippo Signaling Pathway , Humans , Immunohistochemistry , Mice , Phenotype , Phosphorylation , Prognosis , Proportional Hazards Models , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/mortality , Axl Receptor Tyrosine Kinase
15.
Cancer Sci ; 111(4): 1303-1313, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31994271

ABSTRACT

The survival benefit from docetaxel, cisplatin and 5-fluorouracil (TPF) induction chemotherapy in oral squamous cell carcinoma (OSCC) patients is not satisfactory. Previously, we identified that stathmin, a microtubule-destabilizing protein, is overexpressed in OSCC. Here, we further investigated its role as a biomarker that impacts on OSCC chemosensitivity. We analyzed the predictive value of stathmin on TPF induction chemotherapy and its impact on OSCC cell chemosensitivity. Then, we further investigated the therapeutic effects of the combination therapy of TPF chemotherapy and PI3K-AKT-mTOR inhibitors in vitro and in vivo. We found that OSCC patients with low stathmin expression benefited from TPF induction chemotherapy, while OSCC patients with high stathmin expression could not benefit from TPF induction chemotherapy. Stathmin overexpression promoted cellular proliferation and decreased OSCC cell sensitivity to TPF treatment. In addition, inhibition of the PI3K-AKT-mTOR signaling pathway decreased stathmin expression and phosphorylation. The combination therapy of TPF chemotherapy and PI3K-AKT-mTOR inhibitors exhibited a potent antitumor effect both in vitro and in vivo. Therefore, stathmin can be used as a predictive biomarker for TPF induction chemotherapy and a combination therapy regimen based on stathmin expression might improve the survival of OSCC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , Stathmin/genetics , Aged , Animals , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Disease-Free Survival , Docetaxel/administration & dosage , Female , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Male , Mice , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Precision Medicine , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , Taxoids/administration & dosage
16.
J Org Chem ; 85(15): 9808-9819, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32657121

ABSTRACT

Acyclic enediynes are generally inactive under physiological conditions to be used as antitumor agents like their natural enediyne counterparts. A new mechanism named as maleimide-assisted rearrangement and cycloaromatization (MARACA) is uncovered to trigger the reactivity of acyclic enediynes. Through this mechanism, cascade 1,3-proton transfer processes are accelerated with the maleimide moiety at the ene position to enable the acyclic enediynes to undergo cycloaromatization and generate reactive radicals under physiological conditions. Computational studies suggest that the highest energy barrier for MARACA is 26.0 kcal/mol, much lower than that of Bergman cyclization pathway (39.6 kcal/mol). Experimental results show that maleimide-based enediynes exhibit low onset temperature, fast generation of radical species at 37 °C, and much faster reaction in aqueous solution than in nonpolar solvent, which is beneficial to achieve both high reactivity in physiological environment and high stability for storage and delivery in nonpolar media. The generated radical species are capable of causing high percentage of double-strand (ds) DNA cleavage, leading to significant cytotoxicity toward a panel of cancer cell lines with half inhibition concentration down to submicromolar level. Overall, the discovery of the MARACA mechanism provides a platform for designing novel acyclic enediynes with high potency for antitumor applications.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Cyclization , Enediynes/pharmacology , Humans , Maleimides/pharmacology
17.
Mol Cancer ; 18(1): 38, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30857539

ABSTRACT

BACKGROUND: Long intergenic noncoding RNA p21 (lincRNA-p21) is considered a target of wild-type p53, but little is known about its regulation by mutant p53 and its functions during the progression of head and neck squamous cell carcinoma (HNSCC). METHODS: RNAscope was used to detect the expression and distribution of lincRNA-p21. Chromatin immunoprecipitation and electrophoretic mobility shift assays were performed to analyze the transcriptional regulation of lincRNA-p21 in HNSCC cells. The biological functions of lincRNA-p21 were investigated in vitro and in vivo. RNA immunoprecipitation and pull-down assays were used to detect the direct binding of lincRNA-p21. RESULTS: Lower lincRNA-p21 expression was observed in HNSCC tissues and indicated worse prognosis. Both wild and mutant type p53 transcriptionally regulated lincRNA-p21, but nuclear transcription factor Y subunit alpha (NF-YA) was essential for mutant p53 in the regulation of lincRNA-p21. Ectopic expression of lincRNA-p21 significantly inhibited cell proliferation capacity in vitro and in vivo and vice versa. Moreover, the overexpression of lincRNA-p21 induced G1 arrest and apoptosis. Knockdown NF-YA expression reversed tumor suppressor activation of lincRNA-p21 in mutant p53 cells, not wild-type p53 cells. A negative correlation was observed between lincRNA-p21 and the phosphorylation of signal transducer and activator of transcription 3 (p-STAT3) in HNSCC tissues. High lincRNA-p21 expression inhibited Janus kinase 2 (JAK2)/STAT3 signal activation and vice versa. Further, we observed direct binding to STAT3 by lincRNA-p21 in HNSCC cells, which suppressed STAT3-induced oncogenic potential. CONCLUSIONS: Our results revealed the transcriptional regulation of lincRNA-p21 by the mutant p53/NF-YA complex in HNSCC. LincRNA-p21 acted as a tumor suppressor in HNSCC progression, which was attributed to direct binding to STAT3 and blocking of JAK2/STAT3 signaling.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Janus Kinase 2/metabolism , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Follow-Up Studies , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Janus Kinase 2/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Prognosis , STAT3 Transcription Factor/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
18.
Br J Cancer ; 120(3): 317-330, 2019 02.
Article in English | MEDLINE | ID: mdl-30555157

ABSTRACT

BACKGROUND: An immunosuppressive microenvironment is critical for cancer initiation and progression. Whether interferon alpha (IFNα) can suppress immune and cancer cells and its involved mechanism still remain largely elusive. METHODS: We examine the expression of interferon alpha/beta receptor-1 (IFNAR1), CD8, CD56 and programmed death ligand 1 (PDL1) in head and neck squamous cell carcinomas (HNSCC). The effect of IFNα on PDL1 and programmed cell death protein 1 (PD1) expression in tumour cells and immune cells was detected in vitro and in vivo. RESULTS: Overexpression of IFNAR1, MX1 and signal transducer and activator of transcription 1 (Stat1) indicated the endogenous IFNα activation in tumour microenvironment, which correlated with immunosuppression status in HNSCC patients. Moreover, IFNα transcriptionally activated the expression of PDL1 through p-Stat1 (Tyr701) and promoted PD1 expression in immune cells through IFNAR1. The inhibition of IFNα signalling enhanced the cytotoxic activity of nature killer cells. At lastastly, we confirmed the upregulation of PDL1 and PD1 in response to IFNα treatment in both xenograft tumour models and patient-derived xenograft models. CONCLUSIONS: Our findings demonstrate that IFNα-induced PDL1 and PD1 expression is a new mechanism of immunosuppression in HNSCC, suggesting that blocking IFNα signalling may enhance the efficacy of immune checkpoint blockade.


Subject(s)
Interferon-alpha/genetics , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Animals , Apoptosis/genetics , B7-H1 Antigen/genetics , CD56 Antigen/genetics , CD8 Antigens/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Immunosuppression Therapy , Male , Mice , Myxovirus Resistance Proteins/genetics , Programmed Cell Death 1 Receptor/genetics , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Microenvironment/genetics
19.
Cell Mol Biol Lett ; 24: 21, 2019.
Article in English | MEDLINE | ID: mdl-30915128

ABSTRACT

Melatonin receptors MT1 and MT2 (genes officially named MTNR1A and MTNR1B, respectively) play crucial roles in melatonin-mediated regulation of circadian rhythms, the immune system, and control of reproduction in seasonally breeding animals. In this study, immunolocalization assay showed that MT1 and MT2 are highly expressed in Leydig cell membrane. To understand the biological function of melatonin receptors in hCG-induced testosterone synthesis, we generated melatonin receptor knockdown cells using specific siRNA and performed testosterone detection after hCG treatment. We found that knockdown of melatonin receptors, especially MTNR1A, led to an obvious decrease (> 60%) of testosterone level. Our further study revealed that knockdown of melatonin receptors repressed expression, at both the mRNA level and the protein level, of key steroidogenic genes, such as p450scc, p450c17 and StAR, which are essential for testosterone synthesis. hCG triggered endoplasmic reticulum (ER) stress to regulate steroidogenic genes' expression and apoptosis. To further investigate the potential roles of melatonin receptors in hCG-induced regulation of ER stress and apoptosis, we examined expression of some crucial ER stress markers, including Grp78, Chop, ATF4, Xbp1, and IRE1. We found that inhibition of melatonin receptors increased hCG-induced expression of Grp78, Chop and ATF4, but not Xbp1 and IRE1, suggesting that hCG may modulate IRE1 signaling pathways in a melatonin receptor-dependent manner. In addition, our further data showed that knockdown of MTNR1A and MTNR1B promoted hCG-induced expression of apoptosis markers, including p53, caspase-3 and Bcl-2. These results suggested that the melatonin receptors MTNR1A and MTNR1B are essential to repress hCG-induced ER stress and cell apoptosis. Our studies demonstrated that the mammalian melatonin receptors MT1 and MT2 are involved in testosterone synthesis via mediating multiple cell pathways.


Subject(s)
Chorionic Gonadotropin/pharmacology , Gene Deletion , Leydig Cells/metabolism , Receptor, Melatonin, MT1/metabolism , Testosterone/metabolism , Animals , Apoptosis/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Gene Knockdown Techniques , Male , Mice , RNA, Small Interfering/metabolism , Steroids/metabolism , Testis/drug effects , Testis/metabolism , Testosterone/biosynthesis
20.
Br J Cancer ; 118(4): 509-521, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29348488

ABSTRACT

BACKGROUND: The epidermal growth factor receptor (EGFR)-targeted therapies have been tested in the clinic as treatments for head and neck squamous cell carcinoma (HNSCC). Owing to intrinsic or acquired resistance, EGFR-targeted therapies often lead to a low response rate and treatment failure. Interferon-alpha (IFNα) is a chemosensitising agent and multi-functional cytokine with a tumour inhibitory effect. However, the synergic effect of IFNα and EGFR-targeted therapies (erlotinib and nimotuzumab) and their mechanisms in HNSCC remain unclear. METHODS: The interactions between IFNα, erlotinib, and nimotuzumab were evaluated in vitro in HNSCC cells. The synergistic effect of IFNα (20 000 IU per day, s.c.), erlotinib (50 mg kg-1 per day, i.g.) and nimotuzumab (10 mg kg-1 per day, i.p.) was further confirmed in vivo using HNSCC xenografts in nude mice. The upregulation of retinoic-acid inducible gene I (RIG-I) induced by IFNα and EGFR-targeted therapies and its mechanism were detected in vitro and in vivo. RESULTS: IFNα enhances the antitumour effects of erlotinib and nimotuzumab on HNSCC cells both in vitro and in vivo. Importantly, both IFNα and EGFR-targeted therapies promote the expression of RIG-I by activating signal transducers and activators of transcription 1 (STAT1) in HNSCC cells. RIG-I knockdown reduced the sensitivity of HN4 and HN30 cells to IFNα, erlotinib, and nimotuzumab. Moreover, IFNα transcriptionally induced RIG-I expression in HNSCC cells through STAT1. CONCLUSIONS: IFNα enhances the effect of EGFR-targeted therapies by upregulating RIG-I, and its expression may represent a predictor of the effectiveness of a combination treatment including IFNα in HNSCC.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Erlotinib Hydrochloride/administration & dosage , Head and Neck Neoplasms/drug therapy , Interferon-alpha/administration & dosage , Receptors, Retinoic Acid/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line, Tumor , Drug Administration Schedule , Drug Synergism , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Interferon-alpha/pharmacology , Mice , Receptors, Retinoic Acid/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL