Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Hematol Oncol Clin North Am ; 38(2): 305-319, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38302306

ABSTRACT

Cereblon-targeting degraders, including immunomodulatory imide drugs lenalidomide and pomalidomide alongside cereblon E3 ligase modulators like iberdomide and mezigdomide, have demonstrated significant anti-myeloma effects. These drugs play a crucial role in diverse therapeutic approaches for multiple myeloma (MM), emphasizing their therapeutic importance across various disease stages. Despite their evident efficacy, approximately 5% to 10% of MM patients exhibit primary resistance to lenalidomide, and resistance commonly develops over time. Understanding the intricate mechanisms of action and resistance to this drug class becomes imperative for refining and advancing novel therapeutic combinations.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Lenalidomide/pharmacology , Lenalidomide/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/therapeutic use , Ubiquitin-Protein Ligases/therapeutic use
2.
J Natl Compr Canc Netw ; 22(2)2024 02 15.
Article in English | MEDLINE | ID: mdl-38364363

ABSTRACT

More than 90 distinct fusion partners of ALK rearrangement have been identified. Different ALK fusions may exhibit different sensitivities to ALK tyrosine kinase inhibitors. The emergence of rare fusions poses significant challenges to targeted therapies. This study aimed to investigate the response of KANK1::ALK fusion to alectinib in an advanced lung adenocarcinoma. A novel KANK1::ALK fusion was identified by next-generation sequencing (NGS) and Ventana immunohistochemistry assessments. A 73-year-old woman who had never smoked was admitted with hemoptysis in May 2020. PET/CT revealed a nodule in the left upper lobe, with bilateral pulmonary and multiple lymph node metastases. The upper lobe nodule of the left lung was diagnosed as adenocarcinoma through bronchofiberscopy biopsy, resulting in a clinical diagnosis of stage IVA (cT1c,N3,M1a). Because the biopsy tissue was insufficient for NGS analysis, a blood-based genetic analysis was performed, revealing the presence of KRAS p.Q61R mutations. The patient received carboplatin and pemetrexed with pembrolizumab as first-line therapy, followed by maintenance therapy of pembrolizumab monotherapy. Although the tumor initially showed significant shrinkage, it unfortunately progressed further after 11 months. Subsequently, the patient was given carboplatin and pemetrexed with pembrolizumab again, but the tumor progression continued. An NGS using a rebiopsy of the left upper lobe tumor suggested a KANK1::ALK fusion. Alectinib was prescribed in January 2022, and a durable partial response was observed after 18 months. ALK rearrangements were observed in the broader spectrum of lung cancers. This study provided a potential treatment option for patients with KANK1::ALK fusions. Further studies are needed to understand the function of these fusions.


Subject(s)
Adenocarcinoma of Lung , Carbazoles , Lung Neoplasms , Piperidines , Female , Humans , Aged , Pemetrexed , Carboplatin/therapeutic use , Positron Emission Tomography Computed Tomography , Anaplastic Lymphoma Kinase/genetics , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Cytoskeletal Proteins/therapeutic use , Adaptor Proteins, Signal Transducing/therapeutic use
3.
Pediatr Neurosurg ; 59(1): 27-34, 2024.
Article in English | MEDLINE | ID: mdl-38109857

ABSTRACT

INTRODUCTION: Infant-type hemispheric glioma (IHG) is a rare form of cancer that affects newborns and infants. It is classified as a pediatric-type high-grade glioma and typically harbors receptor tyrosine kinase (RTK) gene fusions. Here, we present the finding of a novel gene fusion IHG treated with a targeted therapy that has yet to be implemented for any other IHG case to date. CASE PRESENTATION: We report the case of a 12-month-old boy with IHG who presented with obstructive hydrocephalus due to a large mass in the right frontal lobe. The patient initially underwent mass resection, but subsequent imaging showed rapid interval progression of the residual tumor. Comprehensive molecular analysis of the tumor tissue revealed a novel GAB1-ABL2 gene fusion, and the patient was started on dasatinib, an ABL kinase inhibitor. Shortly after initiation of dasatinib treatment, there was a significant reduction in tumor size and enhancement, followed by stabilization of disease. DISCUSSION: The patient's robust response to treatment suggests that dasatinib is an effective targeted therapy for IHG harboring a GAB1-ABL2 gene fusion. This finding may inform future investigations into the disease processes of IHG and help guide the diagnosis and treatment of IHG in the absence of previously identified gene fusions, improving clinical management of this vulnerable patient population.


Subject(s)
Glioma , Humans , Infant , Male , Adaptor Proteins, Signal Transducing/therapeutic use , Dasatinib/therapeutic use , Glioma/diagnostic imaging , Glioma/drug therapy , Glioma/genetics , Protein Kinase Inhibitors/therapeutic use
4.
OMICS ; 27(8): 338-360, 2023 08.
Article in English | MEDLINE | ID: mdl-37581495

ABSTRACT

Cigarette smoking is the major cause of chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD). It is paramount to develop pharmacological interventions and delivery strategies against the cigarette smoke (CS) associated oxidative stress in COPD. This study in Wistar rats examined cysteamine in nanoemulsions to counteract the CS distressed microenvironment. In vivo, 28 days of CS and 15 days of cysteamine nanoemulsions treatment starting on 29th day consisting of oral and inhalation routes were established in Wistar rats. In addition, we conducted inflammatory and epithelial-to-mesenchymal transition (EMT) studies in vitro in human bronchial epithelial cell lines (BEAS2B) using 5% CS extract. Inflammatory and anti-inflammatory markers, such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-1ß, IL-8, IL-10, and IL-13, have been quantified in bronchoalveolar lavage fluid (BALF) to evaluate the effects of the cysteamine nanoemulsions in normalizing the diseased condition. Histopathological analysis of the alveoli and the trachea showed the distorted, lung parenchyma and ciliated epithelial barrier, respectively. To obtain mechanistic insights into the CS COPD rat model, "shotgun" proteomics of the lung tissues have been carried out using high-resolution mass spectrometry wherein genes such as ABI1, PPP3CA, PSMA2, FBLN5, ACTG1, CSNK2A1, and ECM1 exhibited significant differences across all the groups. Pathway analysis showed autophagy, signaling by receptor tyrosine kinase, cytokine signaling in immune system, extracellular matrix organization, and hemostasis, as the major contributing pathways across all the studied groups. This work offers new preclinical findings on how cysteamine taken orally or inhaled can combat CS-induced oxidative stress.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Rats , Humans , Animals , Rats, Wistar , Cysteamine/pharmacology , Cysteamine/therapeutic use , Proteomics , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Nicotiana , Interleukin-6/metabolism , Anti-Inflammatory Agents/therapeutic use , Cytoskeletal Proteins , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/therapeutic use , Extracellular Matrix Proteins
5.
Mol Omics ; 19(8): 624-639, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37232035

ABSTRACT

Colorectal cancer (CRC), a common malignant tumour of the gastrointestinal tract, is a life-threatening cancer worldwide. Mutations in KRAS and BRAF, the major driver mutation subtypes in CRC, activate the RAS pathway, contribute to tumorigenesis in CRC and are being investigated as potential therapeutic targets. Despite recent advances in clinical trials targeting KRASG12C or RAS downstream signalling molecules for KRAS-mutant CRC, there is a lack of effective therapeutic interventions. Therefore, understanding the unique molecular characteristics of KRAS-mutant CRC is essential for identifying molecular targets and developing novel therapeutic interventions. We obtained in-depth proteomics and phosphoproteomics quantitative data for over 7900 proteins and 38 700 phosphorylation sites in cells from 35 CRC cell lines and performed informatic analyses, including proteomics-based coexpression analysis and correlation analysis between phosphoproteomics data and cancer dependency scores of the corresponding phosphoproteins. Our results revealed novel dysregulated protein-protein associations enriched specifically in KRAS-mutant cells. Our phosphoproteomics analysis revealed activation of EPHA2 kinase and downstream tight junction signalling in KRAS-mutant cells. Furthermore, the results implicate the phosphorylation site Y378 in the tight junction protein PARD3 as a cancer vulnerability in KRAS-mutant cells. Together, our large-scale phosphoproteomics and proteomics data across 35 steady-state CRC cell lines represent a valuable resource for understanding the molecular characteristics of oncogenic mutations. Our approach to predicting cancer dependency from phosphoproteomics data identified the EPHA2-PARD3 axis as a cancer vulnerability in KRAS-mutant CRC.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/therapeutic use , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/therapeutic use , Signal Transduction
6.
West Afr J Med ; 40(4): 375-381, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37119129

ABSTRACT

Comorbid depression among diabetes mellitus (DM) patients is on the increase. This has been linked with poor glycaemic control, greater risk of complications, high burden of medical cost and health care utilisation, and worsening prevalence of other comorbidities resulting in decreased life expectancy. This study determined the antidepressant effect of amitriptyline on depression and glycaemic control among the depressed type 2 DM patients attending Federal Teaching Hospital, Ido-Ekiti (FETHI), Nigeria. It was an interventional study involving 51 depressed type 2 DM patients randomly screened using Patient Health Questionnaire-9 (PHQ-9). They had health education and oral amitriptyline 50mg at night for two months. Postintervention assessment was done using the same tool. Respondents' age ranged between 44 and 78 years with a mean age of 58±8.4 years. Post-intervention assessment showed improved depressive symptoms; 50% of the respondents had significantly improved glycaemic control with a statistically significant effect on depression (the median score of PHQ-9 reduced from 6.0 to 3.0).


La dépression comorbide chez les diabétiques est en augmentation. Elle a été associée à un mauvais contrôle de la glycémie, à un risque accru de complications, à une charge élevée en termes de coûts médicaux et d'utilisation des soins de santé, ainsi qu'à un taux de mortalité plus élevé chez les personnes souffrant de comorbidité. Cette étude a déterminé l'effet de l'antidépresseur (Amitriptyline) sur la dépression et le contrôle de la glycémie chez les patients dépressifs atteints de diabète de type 2 qui fréquentent l'hôpital universitaire fédéral d'Ido-Ekiti (FETHI). Il s'agit d'une étude interventionnelle portant sur 51 patients atteints de diabète de type 2 et déprimés, sélectionnés au hasard à l'aide du questionnaire sur la santé des patients 9 (PHQ-9). Ils ont bénéficié d'une éducation à la santé et ont pris 50 mg d'amitriptyline par voie orale pendant deux mois. L'évaluation post-intervention a été réalisée à l'aide du même outil. L'âge des personnes interrogées était compris entre 44 et 78 ans, avec un âge moyen de 58± 8,4 ans. L'évaluation postintervention a montré une amélioration des symptômes dépressifs, 50% des personnes interrogées ont eu un contrôle glycémique significativement amélioré avec un effet statistiquement significatif sur la dépression (le score médian du PHQ est passé de 6,0 à 3,0). Mots clés: Diabète sucré, dépression, contrôle glycémique, observance thérapeutique.


Subject(s)
Depression , Diabetes Mellitus, Type 2 , Humans , Middle Aged , Aged , Adult , Depression/drug therapy , Depression/epidemiology , Amitriptyline/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Antidepressive Agents/therapeutic use , Medication Adherence , Hospitals, Teaching , Golgi Matrix Proteins , Adaptor Proteins, Signal Transducing/therapeutic use
7.
Ocul Surf ; 28: 131-140, 2023 04.
Article in English | MEDLINE | ID: mdl-36990276

ABSTRACT

PURPOSE: To investigate the efficacy and mechanisms of human umbilical cord-derived MSC-derived extracellular vesicles (hucMSC-EVs) in a mouse model of desiccation-induced dry eye disease (DED). METHODS: hucMSC-EVs were enriched by ultracentrifugation. The DED model was induced by desiccating environment combined with scopolamine administration. The DED mice were divided into the hucMSC-EVs group, fluorometholone (FML) group, PBS group, and blank control group. Tear secretion, corneal fluorescein staining, the cytokine profiles in tears and goblet cells, TUNEL-positive cell, and CD4+ cells were examined to assess therapeutic efficiency. The miRNAs in the hucMSC-EVs were sequenced, and the top 10 were used for miRNA enrichment analysis and annotation. The targeted DED-related signaling pathway was further verified by using RT‒qPCR and western blotting. RESULTS: Treatment with hucMSC-EVs increased the tear volume and maintained corneal integrity in DED mice. The cytokine profile in the tears of the hucMSC-EVs group presented with a lower level of proinflammatory cytokines than PBS group. Moreover, hucMSC-EVs treatment increased goblet cell density and inhibited cell apoptosis and CD4+ cell infiltration. Functional analysis of the top 10 miRNAs in hucMSC-EVs showed a high correlation with immunity. Among them, miR-125 b, let-7b, and miR-6873 were conserved between humans and mice and were associated with the IRAK1/TAB2/NF-κB pathway that was activated in DED. Furthermore, IRAK1/TAB2/NF-κB pathway activation and the abnormal expression of IL-4, IL-8, IL-10, IL-13, IL-17, and TNF-α were reversed by hucMSC-EVs. CONCLUSIONS: hucMSCs-EVs alleviate DED signs, suppress inflammation and restore homeostasis of the corneal surface by multitargeting the IRAK1/TAB2/NF-κB pathway via certain miRNAs.


Subject(s)
Dry Eye Syndromes , Extracellular Vesicles , Mesenchymal Stem Cells , MicroRNAs , Humans , Mice , Animals , MicroRNAs/genetics , NF-kappa B/metabolism , NF-kappa B/therapeutic use , Inflammation/metabolism , Cytokines/metabolism , Extracellular Vesicles/metabolism , Dry Eye Syndromes/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/therapeutic use
8.
Eur J Pharm Sci ; 182: 106372, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36621614

ABSTRACT

Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKIs) was one of the main drugs in the treatment of non-small cell lung cancer (NSCLC). Previous studies had demonstrated that PDZ and LIM Domain 5 (PDLIM5) played an important role in EGFR TKIs resistance. However, there was no feasible method to eliminate EGFR TKIs resistance by suppressing this gene. Here, we formulated a novel mesoporous silica-loaded PDLIM5 siRNA (Small interfering RNA) nanoplatforms. The results have shown that PDLIM5 siRNA could be effectively bound to the nanoplatforms and had good biocompatibility. Further exploration suggested that the nano-platform combined with ultrasonic irradiation could be very effective for siRNA delivery and ultrasound imaging. Moreover, Epithelial-mesenchymal transformation (EMT) changes occurred in PC-9 Gefitinib resistance (PC-9/GR) cells during the development of drug resistance. When PDLIM5 siRNA entered PC-9/GR cells, the sensitivity of drug-resistant cells to gefitinib could be restored through the transforming growth factor-ß (TGF-ß)/EMT pathway. Therefore, PDLIM5 may be an important reason for the resistance of NSCLC cells to gefitinib, and this nanoplatform may become a novel treatment for EGFR TKIs resistance in NSCLC patients.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Gefitinib/pharmacology , Gefitinib/therapeutic use , RNA, Small Interfering/genetics , Lung Neoplasms/metabolism , Epithelial-Mesenchymal Transition , ErbB Receptors , Quinazolines , Drug Resistance, Neoplasm , Ultrasonography , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Adaptor Proteins, Signal Transducing/therapeutic use , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/pharmacology
9.
J Nat Med ; 77(1): 28-40, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36068393

ABSTRACT

Hepatocellular carcinoma (HCC) was the third most common cause of cancer death. But it has only limited therapeutic options, aggressive nature, and very low overall survival. Dihydroartemisinin (DHA), an anti-malarial drug approved by the Food and Drug Administration (FDA), inhibited cell growth in HCC. The Warburg effect was one of the ten new hallmarks of cancer. Solute carrier family 2 member 1 (SLC2A1) was a crucial carrier for glucose to enter target cells in the Warburg effect. Yes-associated transcriptional regulator 1 (YAP1), an effector molecule of the hippo pathway, played a crucial role in promoting the development of HCC. This study sought to determine the role of DHA in the SLC2A1 mediated Warburg effect in HCC. In this study, DHA inhibited the Warburg effect and SLC2A1 in HepG2215 cells and mice with liver tumors in situ. Meanwhile, DHA inhibited YAP1 expression by inhibiting YAP1 promoter binding protein GA binding protein transcription factor subunit beta 1 (GABPB1) and cAMP responsive element binding protein 1 (CREB1). Further, YAP1 knockdown/knockout reduced the Warburg effect and SLC2A1 expression by shYAP1-HepG2215 cells and Yap1LKO mice with liver tumors. Taken together, our data indicated that YAP1 knockdown/knockout reduced the SLC2A1 mediated Warburg effect by shYAP1-HepG2215 cells and Yap1LKO mice with liver tumors induced by DEN/TCPOBOP. DHA, as a potential YAP1 inhibitor, suppressed the SLC2A1 mediated Warburg effect in HCC.


Subject(s)
Artemisinins , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Adaptor Proteins, Signal Transducing/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Glucose Transporter Type 1 , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Transcription Factors/metabolism , Transcription Factors/therapeutic use , Humans
10.
J Nat Prod ; 85(8): 2006-2017, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35976233

ABSTRACT

Bladder cancer, specifically, muscle-invasive bladder cancer (MIBC), is among the most common malignant tumors. Patients with MIBC who cannot tolerate standard drugs require novel treatments. Targeting apoptosis may help treat cancer, which may be achieved with the use of some natural products. Nodosin, found in Isodon serra (Maxim.) Kudo (known as Xihuangcao), may inhibit bladder cancer cells. Transcriptomics and proteomics dual-omic analyses revealed the network pharmacological mechanism: (1) blocking the S phase by up-regulating RPA2, CLSPN, MDC1, PDCD2L, and E2F6 gene expressions, suppressing cancer cell proliferation; (2) inducing apoptosis and autophagy and restraining ferroptosis by up-regulating HMOX1, G0S2, SQSTM1, FTL, SLC7A11, and AIFM2 gene expressions; (3) preventing cancer cell migration by down-regulating NEXN, LIMA1, CFL2, PALLD, and ITGA3 gene expressions. In vivo, nodosin inhibited bladder cancer cell growth in a model of xenograft tumor in nude mice. This study is the first to report basic research findings on the network pharmacological mechanism of cytotoxicity of bladder cancer cells by nodosin, providing novel evidence for the application of nodosin in the field of oncology; however, other mechanisms may be involved in the effects of nodosin for further research. These findings provide a foundation for the development of novel MIBC drugs.


Subject(s)
Biological Products , Urinary Bladder Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Adaptor Proteins, Signal Transducing/therapeutic use , Animals , Biological Products/pharmacology , Cell Line, Tumor , Cell Proliferation , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/pharmacology , Cytoskeletal Proteins/therapeutic use , Diterpenes , Humans , Mice , Mice, Nude , Microfilament Proteins/metabolism , Microfilament Proteins/pharmacology , Microfilament Proteins/therapeutic use , Muscles/metabolism , Muscles/pathology , Network Pharmacology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
11.
Cancer Genet ; 264-265: 50-59, 2022 06.
Article in English | MEDLINE | ID: mdl-35366592

ABSTRACT

Gene fusions involving the neurotrophic receptor tyrosine kinase genes NTRK1, NTRK2, and NTRK3, are well established oncogenic drivers in a broad range of pediatric and adult tumors. These fusions are also important actionable markers, predicting often dramatic response to FDA approved kinase inhibitors. Accurate interpretation of the clinical significance of NTRK fusions is a high priority for diagnostic laboratories, but remains challenging and time consuming given the rapid pace of new data accumulation, the diversity of fusion partners and tumor types, and heterogeneous and incomplete information in variant databases and knowledgebases. The ClinGen NTRK Fusions Somatic Cancer Variant Curation Expert Panel (SC-VCEP) was formed to systematically address these challenges and create an expert-curated resource to support clinicians, researchers, patients and their families in making accurate interpretations and informed treatment decisions for NTRK fusion-driven tumors. We describe a system for NTRK fusion interpretation (including compilation of key elements and annotations) developed by the NTRK fusions SC-VCEP. We illustrate this stepwise process on examples of LMNA::NTRK1 and KANK1::NTRK2 fusions. Finally, we provide detailed analysis of current representation of NTRK fusions in public fusion databases and the CIViC knowledgebase, performed by the NTRK fusions SC-VCEP to determine existing gaps and prioritize future curation activities.


Subject(s)
Neoplasms , Receptor, trkA , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/therapeutic use , Adult , Biomarkers, Tumor/genetics , Carcinogenesis , Child , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/therapeutic use , Gene Fusion , Humans , Neoplasms/diagnosis , Oncogene Proteins, Fusion/genetics , Receptor, trkA/genetics , Receptor, trkA/therapeutic use
12.
Acta Pharm ; 72(3): 427-436, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36651547

ABSTRACT

Hepatocellular carcinoma (HCC) remains a notably global health challenge with high mortality rates and poor prognosis. The deregulation of the Hippo signalling pathway, especially the overexpression and activation of downstream effector Yes-associated protein (YAP), has been demonstrated to result in the rapid malignant evolution of HCC. In this context, multiple efforts have been dedicated to targeting YAP for HCC therapy, but effective YAP inhibitors are still lacking. In this study, through a YAP-TEAD (8×GTIIC) luciferase reporter assay, we identified fingolimod, an immunomodulatory drug approved for the treatment of multiple sclerosis, as a novel YAP inhibitor. Fingolimod suppressed the proliferation of HCC cell lines by downregulating the protein levels as well as the trans-activating function of YAP. Overall, our current study not only identifies fingolimod as a novel YAP-targeting in hibitor, but also indicates that this clinically-approved drug could be utilized as a potential and feasible therapeutic drug for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/therapeutic use , Liver Neoplasms/drug therapy , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Transcription Factors/metabolism , Transcription Factors/therapeutic use , Trans-Activators/metabolism , Trans-Activators/therapeutic use , YAP-Signaling Proteins , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Cell Line, Tumor
13.
Clin Transl Med ; 11(11): e591, 2021 11.
Article in English | MEDLINE | ID: mdl-34841684

ABSTRACT

BACKGROUND: The methylation of lysine residues has been involved in the multiple biological and diseases processes. Recently, some particular non-histone proteins have been elucidated to be methylated by SMYD2, a SET and MYND domain protein with lysine methyltransferase activity. METHODS: SMYD2 was evaluated in synovial tissue and cells derived from rheumatoid arthritis patients. We confirmed TRAF2 could be methylated by SMYD2 using Mass spectrometry, pull-down, immunoprecipitation, methyltransferase assay, ubiquitination assay, luciferase reporter assays, and western blot analyses. Using loss- and gain-of function studies, we explored the biological functions of SMYD2 in vitro and in vivo. Using acute and chronic inflammation with different mice models to determine the impact of SMYD2. RESULTS: Here, we first time confirmed that the cytoplasmic protein TRAF2 as the kernel node for NF-κB signaling pathway could be methylated by SMYD2. SMYD2-mediated TRAF2 methylation contributed to the durative sensitization of NF-κB signaling transduction through restraining its own proteolysis and enhancing the activity. In addition, we found knocking down of SMYD2 has different degrees of mitigation in acute and chronic inflammation mice models. Furthermore, as the lysine-specific demethylase, LSD1 could resist methylation on TRAF2 induced by SMYD2. CONCLUSIONS: Our data uncovered an unprecedented cytoplasmic protein network that employed methylation of TRAF2 for the maintenance of NF-κB activation during inflammatory diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/drug effects , Histone-Lysine N-Methyltransferase/pharmacology , Inflammation/drug therapy , Methylation/drug effects , Adaptor Proteins, Signal Transducing/therapeutic use , Histone-Lysine N-Methyltransferase/therapeutic use , Humans , Inflammation/immunology , Inflammation/physiopathology , Signal Transduction/drug effects
14.
Cells ; 10(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33572372

ABSTRACT

The present study explored the effects of endophilin A1 (SH3GL2) against oxidative damage brought about by H2O2 in HT22 cells and ischemic damage induced upon transient forebrain ischemia in gerbils. Tat-SH3GL2 and its control protein (Control-SH3GL2) were synthesized to deliver it to the cells by penetrating the cell membrane and blood-brain barrier. Tat-SH3GL2, but not Control-SH3GL2, could be delivered into HT22 cells in a concentration- and time-dependent manner and the hippocampus 8 h after treatment in gerbils. Tat-SH3GL2 was stably present in HT22 cells and degraded with time, by 36 h post treatment. Pre-incubation with Tat-SH3GL2, but not Control-SH3GL2, significantly ameliorated H2O2-induced cell death, DNA fragmentation, and reactive oxygen species formation. SH3GL2 immunoreactivity was decreased in the gerbil hippocampal CA1 region with time after ischemia, but it was maintained in the other regions after ischemia. Tat-SH3GL2 treatment in gerbils appreciably improved ischemia-induced hyperactivity 1 day after ischemia and the percentage of NeuN-immunoreactive surviving cells increased 4 days after ischemia. In addition, Tat-SH3GL2 treatment in gerbils alleviated the increase in lipid peroxidation as assessed by the levels of malondialdehyde and 8-iso-prostaglandin F2α and in pro-inflammatory cytokines such as tumor necrosis factor-α, interleukin-1ß, and interleukin-6; while the reduction of protein levels in markers for synaptic plasticity, such as postsynaptic density 95, synaptophysin, and synaptosome associated protein 25 after transient forebrain ischemia was also observed. These results suggest that Tat-SH3GL2 protects neurons from oxidative and ischemic damage by reducing lipid peroxidation and inflammation and improving synaptic plasticity after ischemia.


Subject(s)
Adaptor Proteins, Signal Transducing/therapeutic use , Brain Ischemia/drug therapy , Hippocampus/pathology , Lipid Peroxidation , Neuronal Plasticity , Neurons/pathology , Neuroprotective Agents/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adaptor Proteins, Signal Transducing/pharmacology , Animals , Brain Ischemia/physiopathology , Cell Death/drug effects , Cell Line , Gene Products, tat/metabolism , Gerbillinae , Hippocampus/physiopathology , Hydrogen Peroxide/toxicity , Lipid Peroxidation/drug effects , Mice , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Recombinant Fusion Proteins/pharmacology , Time Factors
15.
J Nat Med ; 74(4): 777-787, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32666278

ABSTRACT

Lung adenocarcinoma (LUAD) is the top prevalent histological kind of lung cancer worldwide. Recent evidences have demonstrated that Sauchinone plays an anticancer role in tumor cell invasion and migration. Therefore, we performed this investigation to explain the potential role of Sauchinone in LUAD as well as the potential mechanism involved. Cell counting kit 8 (CCK-8) and transwell experiments were implemented to measure the proliferative, invasive and migratory abilities of LUAD cells. qRT-PCR and Western blot were performed to detect the transfection efficiency of si-EIF4EBP1s. Additionally, Western blot was also implemented to evaluate the effect of Sauchinone on EIF4EBP1 expression level as well as cell cycle-related proteins. Our findings showed that Sauchinone remarkably suppressed the proliferative ability of LUAD cells in a dose-dependent and time-dependent manner. EIF4EBP1 was a candidate target gene of Sauchinone. EIF4EBP1 expression was increased in LUAD tissues, and its high expression induced a poorer prognosis of LUAD patients. EIF4EBP1 expression was positively associated with cell cycle in LUAD. Sauchinone treatment attenuated EIF4EBP1 expression and cell cycle-related protein levels. Knockdown of EIF4EBP1 repressed the proliferation, invasion and migration of LUAD cells; furthermore, Sauchinone stimulation enforced its inhibitory effect. Meanwhile, the treatment of Sauchinone intensified the arrest of cell cycle induced by EIF4EBP1 knockdown. To sum up, our discovery indicated that Sauchinone exerts an anticancer role through down-regulating EIF4EBP1 and mediating cell cycle in LUAD.


Subject(s)
Adaptor Proteins, Signal Transducing/therapeutic use , Adenocarcinoma of Lung/drug therapy , Benzopyrans/therapeutic use , Cell Cycle Proteins/therapeutic use , Dioxoles/therapeutic use , Lung Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/pharmacology , Adenocarcinoma of Lung/pathology , Benzopyrans/pharmacology , Cell Cycle Proteins/pharmacology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Dioxoles/pharmacology , Down-Regulation , Humans , Lung Neoplasms/pathology , Neoplasm Invasiveness , Prospective Studies , Transfection
17.
Andrologia ; 52(2): e13488, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31797427

ABSTRACT

We aimed to assess the effects of gametogenetin-binding protein 2 (GGNBP2) on the proliferation, invasion and migration of prostate cancer PC-3 cells. PcDNA3-HisC-GGNBP2 was transfected to overexpress GGNBP2. Proliferation was tested by MTT assay, and migration and invasion were detected by Transwell assay. Cell cycle was detected by flow cytometry. The protein expressions of COX-2, cyclin D1, PI3K, Akt and p-Akt were detected by Western blot. A subcutaneous xenograft model of prostate cancer was established. Mice were randomly divided into three groups (n = 9) and intratumorally injected with pcDNA3-HisC-GGNBP2, pcDNA3-HisC and normal saline respectively. The xenograft tumour volume was measured every 3 days, and weight was measured after 2 weeks. After GGNBP2 overexpression, the proliferation, migration and invasion capacities of PC-3 cells decreased, and cell cycle was arrested in the G1 phase. The protein expressions of COX-2, cyclin D1, PI3K, Akt and p-Akt all reduced. The tumour volume and weight of pcDNA3-HisC-GGNBP2 group were significantly lower than those of pcDNA3-HisC group (p < .05). The proliferation capacity of GGNBP2-overexpressing prostate cancer cells is significantly attenuated, tumour growth is significantly inhibited, and cell cycle is arrested in the G1 phase. GGNBP2 overexpression affects the growth of castration-resistant prostate cancer via the PI3K/Akt signalling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/therapeutic use , Prostatic Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement , Cell Proliferation , Drug Screening Assays, Antitumor , Humans , Male , Mice, Inbred BALB C , PC-3 Cells , Prostatic Neoplasms/metabolism
18.
Arch Med Res ; 50(4): 225-233, 2019 05.
Article in English | MEDLINE | ID: mdl-31518897

ABSTRACT

BACKGROUND: High vascularization is a major characteristic of renal cell carcinoma (RCC). Thus, exploration of molecules promoting the tumor vascularization in RCC is urgent. Yes-associated Protein (YAP) is an oncogene in many cancer types, and high YAP expression was correlated with worse overall survival of RCC patients according to The Cancer Genome Atlas (TCGA) database. However, whether YAP promotes tumor angiogenesis of RCC is still unknown. METHODS: Western blotting assay, real-time Quantitive PCR analysis, and ELISA assay were used to detect the related gene expression. The function of YAP on tumor angiogenesis was investigated by HUVEC recruitment, tube formation, and rabbit cornea assay. The clinical relevance of several genes was analyzed in a public database. RESULTS: knockdown of YAP decreased RCC cell-inducing HUVEC recruitment and tube formation. Moreover, tumor angiogenesis ability of 786-O cells was crippled by YAP knockdown in vivo. In addition, the expression of Vascular endothelial growth factors A (VEGFA) was positively correlated with YAP expression in RCC tumor tissues, and YAP promoted expression and secretion of VEGFA in RCC cells. Mechanistically, GLI family zinc finger 2 (Gli2) knockdown in RCC cells reduced both basic and YAP-induced VEGFA expression, HUVECs recruitment, and tube formation, indicating that Gli2 is necessary for YAP to promote expression of VEGFA. CONCLUSION: Taken together, our results demonstrate that YAP/Gli2 promotes VEGFA expression and tumor angiogenesis in RCC cells, which could provide novel therapeutic targets in RCC treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/therapeutic use , Carcinoma, Renal Cell/genetics , Neovascularization, Pathologic/genetics , Transcription Factors/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Zinc Finger Protein Gli2/genetics , Adaptor Proteins, Signal Transducing/pharmacology , Animals , Cell Line, Tumor , Female , Humans , Male , Rabbits , Transcription Factors/pharmacology , Transfection , YAP-Signaling Proteins
19.
Nat Med ; 24(10): 1507-1512, 2018 10.
Article in English | MEDLINE | ID: mdl-30297895

ABSTRACT

Retinal gene therapy is increasingly recognized as a novel molecular intervention that has huge potential in treating common causes of blindness, the majority of which have a genetic aetiology1-5. Choroideremia is a chronic X-linked retinal degeneration that was first described in 18726. It leads to progressive blindness due to deficiency of Rab-escort protein 1 (REP1). We designed an adeno-associated viral vector to express REP1 and assessed it in a gene therapy clinical trial by subretinal injection in 14 patients with choroideremia. The primary endpoint was vision change in treated eyes 2 years after surgery compared to unoperated fellow eyes. Despite complications in two patients, visual acuity improved in the 14 treated eyes over controls (median 4.5 letter gain, versus 1.5 letter loss, P = 0.04), with 6 treated eyes gaining more than one line of vision (>5 letters). The results suggest that retinal gene therapy can sustain and improve visual acuity in a cohort of predominantly late-stage choroideremia patients in whom rapid visual acuity loss would ordinarily be predicted.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Choroideremia/therapy , Genetic Therapy , Retinal Degeneration/physiopathology , Visual Acuity/genetics , Adaptor Proteins, Signal Transducing/therapeutic use , Adult , Aged , Choroideremia/genetics , Choroideremia/physiopathology , Choroideremia/surgery , Dependovirus/genetics , Genetic Vectors/therapeutic use , Humans , Male , Middle Aged , Retina/physiopathology , Retinal Degeneration/genetics , Retinal Degeneration/surgery , Vision, Ocular/genetics , Vision, Ocular/physiology
20.
Crit Care ; 21(1): 171, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28679406

ABSTRACT

BACKGROUND: Formyl peptide receptor 2-lipoxin receptor (FPR2/ALX) modulates the anti-inflammatory response and therefore may be a target for treating sepsis. The purpose of this study was to investigate the association between genetic variants of the FPR2/ALX gene and sepsis after severe trauma as well as to further analyze the functions of sepsis-related genetic polymorphisms. METHODS: Three tag single-nucleotide polymorphisms (tag SNPs) that captured all common alleles across the FPR2/ALX genomic region were genotyped using pyrosequencing in an initial sample consisting of 275 patients with severe trauma. The rs11666254 polymorphism, which had statistical significance, was genotyped in an additional 371 patients, and logistic regression analysis was performed to determine associations between the FPR2/ALX gene polymorphism and sepsis susceptibility after severe trauma. The messenger RNA (mRNA) and protein levels of FPR2/ALX in the lipopolysaccharide-stimulated white blood cells of trauma patients were determined by performing quantitative polymerase chain reactions and Western blot analysis. Tumor necrosis factor (TNF)-α production was measured by enzyme-linked immunosorbent assay. The effects of the promoter polymorphism rs11666254 on the transcription activity of FPR2/ALX were analyzed using a luciferase reporter assay. RESULTS: Among the three tag SNPs, only the rs11666254 polymorphism was found to be significantly associated with sepsis in trauma patients, and this association persisted after a pooled analysis of all 646 trauma patients, which showed that patients who carried the A allele of rs11666254 had a significantly higher risk of developing sepsis than individuals who carried the G allele. This SNP was also significantly associated with lower FPR2/ALX mRNA and protein expression as well as higher TNF-α production from the peripheral blood leukocyte response to bacterial lipoprotein stimulation. In addition, the rs11666254 polymorphism could significantly decrease the promoter activity of the FPR2/ALX gene. CONCLUSIONS: The rs11666254 polymorphism in the FPR2/ALX gene is a functional SNP that increases sepsis susceptibility in patients after traumatic injury.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Receptors, Formyl Peptide/genetics , Receptors, Lipoxin/genetics , Sepsis/genetics , Wounds and Injuries/genetics , Adaptor Proteins, Signal Transducing/pharmacology , Adaptor Proteins, Signal Transducing/therapeutic use , Adolescent , Adult , Aged , China , Female , Flow Cytometry/methods , Humans , Injury Severity Score , Male , Middle Aged , Polymorphism, Single Nucleotide/physiology , Sepsis/metabolism , Wounds and Injuries/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...