Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 30(10): 3284-3299, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-35765243

RESUMEN

Existing evidence indicates that gut fungal dysbiosis might play a key role in the pathogenesis of colorectal cancer (CRC). We sought to explore whether reversing the fungal dysbiosis by terbinafine, an approved antifungal drug, might inhibit the development of CRC. A population-based study from Sweden identified a total of 185 patients who received terbinafine after their CRC diagnosis and found that they had a decreased risk of death (hazard ratio = 0.50) and metastasis (hazard ratio = 0.44) compared with patients without terbinafine administration. In multiple mouse models of CRC, administration of terbinafine decreased the fungal load, the fungus-induced myeloid-derived suppressor cell (MDSC) expansion, and the tumor burden. Fecal microbiota transplantation from mice without terbinafine treatment reversed MDSC infiltration and partially restored tumor proliferation. Mechanistically, terbinafine directly impaired tumor cell proliferation by reducing the ratio of nicotinamide adenine dinucleotide phosphate (NADP+) to reduced form of nicotinamide adenine dinucleotide phosphate (NADPH), suppressing the activity of glucose-6-phosphate dehydrogenase (G6PD), resulting in nucleotide synthesis disruption, deoxyribonucleotide (dNTP) starvation, and cell-cycle arrest. Collectively, terbinafine can inhibit CRC by reversing fungal dysbiosis, suppressing tumor cell proliferation, inhibiting fungus-induced MDSC infiltration, and restoring antitumor immune response.


Asunto(s)
Neoplasias Colorrectales , Terbinafina , Animales , Antifúngicos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Desoxirribonucleótidos , Disbiosis , Glucosafosfato Deshidrogenasa , Ratones , NADP , Terbinafina/farmacología
2.
Biochem Biophys Res Commun ; 551: 100-106, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33725570

RESUMEN

Colorectal cancer (CRC) is prevalent worldwide and novel diagnostic and prognostic biomarkers are needed to improve precision medicine. Circular RNAs (circRNAs) are currently being considered as emerging tumor biomarkers. Herein, we aimed to explore the possible clinical application of circRNAs in the early diagnosis and prognostic prediction of CRC. First, candidate circRNA was selected by integrating analysis of Gene Expression Omnibus (GEO) database using GEO2R program. ROC curve analysis demonstrated the predictive values and likelihood ratios of circ_001659 were satisfactory for the diagnosis of CRC, including patients in early-stage disease or patients with carcinoembryonic antigen (CEA)-negative status. Moreover, serum circ_001659 may be a novel biomarker in the assessment of successful treatment and remission of cancer tracking. We further investigated the oncogenic role of circ_001659. In vivo and in vitro experiments indicated that circ_001659 could promote CRC cell invasion and migration. Mechanistically, circ_001659 was localized in the nucleus, recruited the RBBP5 to Vimentin promoter and increased H3K4 trimethylation level on the Vimentin promoter region, which epigenetically activated Vimentin transcription. Our findings demonstrate that circ_001659 could be a useful serum biomarker for CRC diagnosis and prognosis. Targeting circ_001659 and its pathway may be meaningful for treating patients with CRC.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/diagnóstico , Metástasis de la Neoplasia , ARN Circular/sangre , Animales , Biomarcadores de Tumor/genética , Antígeno Carcinoembrionario/sangre , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia/genética , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Circular/genética , Transcripción Genética , Vimentina/genética
3.
J Immunol ; 199(11): 3849-3857, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29070673

RESUMEN

This study identifies a novel mechanism linking IL-17A with colon tissue repair and tumor development. Abrogation of IL-17A signaling in mice attenuated tissue repair of dextran sulfate sodium (DSS)-induced damage in colon epithelium and markedly reduced tumor development in an azoxymethane/DSS model of colitis-associated cancer. A novel IL-17A target gene, PLET1 (a progenitor cell marker involved in wound healing), was highly induced in DSS-treated colon tissues and tumors in an IL-17RC-dependent manner. PLET1 expression was induced in LGR5+ colon epithelial cells after DSS treatment. LGR5+PLET1+ marks a highly proliferative cell population with enhanced expression of IL-17A target genes. PLET1 deficiency impaired tissue repair of DSS-induced damage in colon epithelium and reduced tumor formation in an azoxymethane/DSS model of colitis-associated cancer. Our results suggest that IL-17A-induced PLET1 expression contributes to tissue repair and colon tumorigenesis.


Asunto(s)
Colitis/inmunología , Colon/metabolismo , Neoplasias del Colon/inmunología , Células Epiteliales/inmunología , Interleucina-17/metabolismo , Proteínas Gestacionales/metabolismo , Animales , Azoximetano , Carcinogénesis , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Colitis/inducido químicamente , Colon/patología , Neoplasias del Colon/inducido químicamente , Sulfato de Dextran , Regulación Neoplásica de la Expresión Génica , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Gestacionales/genética , Receptores de Interleucina/genética , Cicatrización de Heridas
4.
Bioorg Med Chem Lett ; 27(24): 5446-5449, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29138025

RESUMEN

To follow up on our recent discovery of the 18-amino acid all-hydrocarbon [i, i + 4]-stapled p110α[E545K] peptide 1 that was shown to potently block the intracellular p110α[E545K]-IRS1 interaction (a protein-protein interaction uniquely present in cancer cells expressing p110α[E545K]) and the growth of the xenograft tumors formed by cancers harboring this mutation, in the current study we prepared and examined six derivatives of 1, i.e. stapled peptides 2-A, 2-B, 3-A, 3-B, 4-A, 4-B. We found that 2-A, 2-B, 4-A, and 4-B had higher % α-helicity than 1; moreover, the enhanced % α-helicity also led to an enhanced proteolytic stability. When compared with 1, the structurally simplified 14-amino acid 4-A and 4-B were found to more potently deactivate the AKT phosphorylation at Ser473 in the p110α[E545K]-expressing colon cancer cells, whose activation was previously demonstrated by us to be specifically derived from the p110α[E545K]-IRS1 interaction. The preliminary findings from the current study have laid a foundation for future more extensive studies on the stapled p110α[E545K] peptides newly identified in the current study.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Hidrocarburos/química , Proteínas Sustrato del Receptor de Insulina/metabolismo , Péptidos/química , Línea Celular Tumoral , Dicroismo Circular , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/genética , Humanos , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Mutación , Resonancia Magnética Nuclear Biomolecular , Péptidos/síntesis química , Péptidos/farmacología , Fosforilación/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Bioorg Med Chem Lett ; 25(16): 3319-24, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26081291

RESUMEN

N(ε)-Thiocarbamoyl-lysine was recently demonstrated by our laboratory to be a potent catalytic mechanism-based SIRT1/2/3 inhibitory warhead, in the current study, among the prepared analogs of N(ε)-thiocarbamoyl-lysine with its terminal NH2 mono-substituted with alkyl and aryl groups, we found that N(ε)-methyl-thiocarbamoyl-lysine and N(ε)-carboxyethyl-thiocarbamoyl-lysine, respectively, also behaved as strong inhibitory warheads against SIRT1/2/3 and SIRT5, typical deacetylases and deacylase in the human sirtuin family, respectively. Moreover, N(ε)-methyl-thiocarbamoyl-lysine was found in the study to be a ∼ 2.5-18.4-fold stronger SIRT1/2/3 inhibitory warhead than its lead warhead N(ε)-thiocarbamoyl-lysine.


Asunto(s)
Sirtuina 1/antagonistas & inhibidores , Sirtuina 2/antagonistas & inhibidores , Tiourea/química , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Tiourea/farmacología
6.
Toxicol Pathol ; 42(1): 140-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24178578

RESUMEN

Phosphoinositide-3-kinase, catalytic, alpha polypeptide, which encodes the catalytic p110α subunit of phosphatidylinositol 3-kinase α, is the most frequently mutated oncogene in human cancers. Targeting mutant p110α holds great promise for cancer therapy. However, it is challenging to develop p110α isoform-specific inhibitors. Most p110α mutations occur at two hot spot regions: an acidic cluster (E542, E545, and Q546) in the helical domain and a histidine residue (H1047) in the kinase domain. We recently discovered that p110α helical domain mutant proteins, but not the kinase domain mutant proteins, directly associate with insulin receptor substrate 1 (IRS1). Moreover, we demonstrated that disruption of protein-protein interaction between p110α helical domain mutant and IRS1 inhibits the growth of tumors with such mutations. The direct protein interaction between IRS1 and p110α helical domain mutants may provide a more accessible target for developing novel precision cancer therapy.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/genética , Terapia Molecular Dirigida , Mutación , Neoplasias/terapia , Fosfatidilinositol 3-Quinasas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Dominio Catalítico , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas Mutantes/metabolismo , Neoplasias/genética , Oncogenes , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal
7.
Cell Rep Med ; 5(5): 101510, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38614093

RESUMEN

Key gene mutations are essential for colorectal cancer (CRC) development; however, how the mutated tumor cells impact the surrounding normal cells to promote tumor progression has not been well defined. Here, we report that PIK3CA mutant tumor cells transmit oncogenic signals and result in malignant transformation of intestinal epithelial cells (IECs) via paracrine exosomal arachidonic acid (AA)-induced H3K4 trimethylation. Mechanistically, PIK3CA mutations sustain SGK3-FBW7-mediated stability of the cPLA2 protein, leading to the synthetic increase in AA, which is transported through exosome and accumulated in IECs. Transferred AA directly binds Menin and strengthens the interactions of Menin and MLL1/2 methyltransferase. Finally, the combination of VTP50469, an inhibitor of the Menin-MLL interaction, and alpelisib synergistically represses PDX tumors harboring PIK3CA mutations. Together, these findings unveil the metabolic link between PIK3CA mutant tumor cells and the IECs, highlighting AA as the potential target for the treatment of patients with CRC harboring PIK3CA mutations.


Asunto(s)
Ácido Araquidónico , Transformación Celular Neoplásica , Ensamble y Desensamble de Cromatina , Fosfatidilinositol 3-Quinasa Clase I , Mutación , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Ácido Araquidónico/metabolismo , Animales , Mutación/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ensamble y Desensamble de Cromatina/genética , Ratones , Línea Celular Tumoral , Colon/patología , Colon/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Exosomas/metabolismo , Exosomas/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Histonas/metabolismo , Histonas/genética
8.
Redox Biol ; 75: 103286, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39079386

RESUMEN

Metabolic reprogramming is a hallmark of human cancer, and cancer-specific metabolism provides opportunities for cancer diagnosis, prognosis, and treatment. However, the underlying mechanisms by which metabolic pathways affect the initiation and progression of colorectal cancer (CRC) remain largely unknown. Here, we demonstrate that cysteine is highly enriched in colorectal tumors compared to adjacent non-tumor tissues, thereby promoting tumorigenesis of CRC. Synchronously importing both cysteine and cystine in colorectal cancer cells is necessary to maintain intracellular cysteine levels. Hypoxia-induced reactive oxygen species (ROS) and ER stress regulate the co-upregulation of genes encoding cystine transporters (SLC7A11, SLC3A2) and genes encoding cysteine transporters (SLC1A4, SLC1A5) through the transcription factor ATF4. Furthermore, the metabolic flux from cysteine to reduced glutathione (GSH), which is critical to support CRC growth, is increased due to overexpression of glutathione synthetase GSS in CRC. Depletion of cystine/cysteine by recombinant cyst(e)inase effectively inhibits the growth of colorectal tumors by inducing autophagy in colorectal cancer cells through mTOR-ULK signaling axis. This study demonstrates the underlying mechanisms of cysteine metabolism in tumorigenesis of CRC, and evaluates the potential of cysteine metabolism as a biomarker or a therapeutic target for CRC.


Asunto(s)
Neoplasias Colorrectales , Cisteína , Reprogramación Metabólica , Animales , Humanos , Ratones , Factor de Transcripción Activador 4/metabolismo , Factor de Transcripción Activador 4/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Carcinogénesis/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/genética , Cisteína/metabolismo , Cistina/metabolismo , Estrés del Retículo Endoplásmico , Regulación Neoplásica de la Expresión Génica , Glutatión/metabolismo , Hipoxia/metabolismo , Hipoxia/genética , Antígenos de Histocompatibilidad Menor , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
9.
Cell Biosci ; 13(1): 164, 2023 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-37689735

RESUMEN

BACKGROUND: PI3K/AKT signaling pathway plays important role in tumorigenesis of human cancer. Protein phosphorylation is crucial for signaling transduction of this pathway. PIK3CA, encoding the catalytic subunit p110α of PI3K complex, is one of the most frequently mutated oncogenes in human cancers. However, phosphorylation sites of PIK3CA/p110α and their underlying mechanism in tumorigenesis are largely unknown. METHODS: Tyrosine phosphorylation sites of PIK3CA/p110α are identified with Mass-Spectrum. Crispr/CAS9 strategy is applied to generate Y317F and Y508F mutant knock-in cell clones. The growth and metastasis abilities of cells are evaluated in vitro and in vivo. Phospho-proteomics analysis and Western blots are used to demonstrate downstream signaling pathways of PIK3CA/p110α tyrosine phosphorylation. In vitro kinase assay is applied to identify the kinase of PIK3CA/p110α tyrosine phosphorylation. RESULTS: Tyrosine phosphorylation of PIK3CA/p110α is stimulated by growth factors such as EGF, HGF and PDGF. Two tyrosine residues, Y317 and Y508, are identified on PIK3CA/p110α. Either Y317 or Y508 phosphorylation is essential for tumorigenesis of CRC. Mutation at Y317 of p110α reduces the proliferation, migration, and invasion of cancer cells through Src-MLC2 pathway, while mutation at Y508 of p110α impairs AKT signaling. Moreover, Src interacts with and phosphorylates p110α. CONCLUSIONS: PIK3CA/p110α phosphorylation at Y317 and Y508 play important role in tumorigenesis of colorectal cancer through two independent pathways.

10.
Cell Rep ; 42(3): 112188, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36857183

RESUMEN

PI3K regulatory subunit p85s normally stabilizes and regulates catalytic subunit p110s in the cytoplasm. Recent studies show that p110-free p85s in the nucleus plays important roles in biological processes. However, the mechanisms by which p85s translocate into the nucleus remain elusive. Here, we describe the mechanism by which p85ß translocates into the nucleus to promote ccRCC tumorigenesis. Phosphorylation of p85ß at the Y464 by FAK facilitates its nuclear translocation in the kidney through enhancing the binding of p85ß to KPNA1. PIK3R2/p85ß is highly expressed in ccRCC samples and associated with overall survival of ccRCC patients. Nuclear but not cytoplasmic p85ß performs oncogenic functions by repressing RB1 expression and regulating the G1/S cell cycle transition. Nuclear p85ß represses RB1 expression by stabilizing histone methyltransferase EZH1/EZH2 proteins. Last, the FAK inhibitor defactinib significantly suppresses the tumor growth of ccRCC with high p85ß Y464 levels.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinogénesis , Transformación Celular Neoplásica , Fosforilación , Proteínas de Unión a Retinoblastoma , Transducción de Señal , Ubiquitina-Proteína Ligasas
11.
Front Pharmacol ; 14: 1161897, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251313

RESUMEN

Background: Hemodialysis patients have a high risk of severe/critical COVID-19 and related high mortality, but nirmatrelvir/ritonavir is not recommended for hemodialysis patients with COVID-19 infection because of lack of evidence of safety. Objectives: Our study aims to evaluate the minimum plasma concentration (Cmin) of nirmatrelvir and its safety of different doses of nirmatrelvir/ritonavir in hemodialysis patients with mild COVID-19. Method: This was a prospective, two step, nonrandomized, open-label study. Participants were treated with nirmatrelvir 150 mg or 300 mg once a day (another 75 mg or 150 mg supplied after hemodialysis) and ritonavir 100 mg twice daily for 5 days, respectively. The primary outcome was the safety of nirmatrelvir/ritonavir, including the Cmin of nirmatrelvir and the number of adverse events (AE). The secondary outcome was the time of viral elimination in hemodialysis patients. Results: Adverse events were happened in 3 and 7 participants in the step 1 and step 2 group, respectively (p = 0.025). Among them, 2 and 6 participants were identified as drug-related adverse events (p = 0.054). No SAE or liver function damage happened. The Cmin of nirmatrelvir in step 1 and step 2 group were 5,294.65 ± 2,370.59 ng/mL and 7,675.67 ± 2,745.22 ng/mL (p = 0.125). The Cmin of the control group was 2,274.10 ± 1,347.25 ng/mL (p = 0.001 compared to step 2 and p = 0.059 compared to step 1). Compared to hemodialysis patients without nirmatrelvir/ritonavir, there were no statistical differences in overall viral elimination time (p = 0.232). Conclusion: In our study, two doses of nirmatrelvir/ritonavir appeared to be excessive for hemodialysis patients. Although all of the patients tolerated 5-day administration, nearly half of the patients experienced drug-related adverse events. In addition, the medication group did not show a significant advantage in the time of viral elimination.

12.
Cancer Cell ; 41(6): 1118-1133.e12, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37267951

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is the major sensor for cytosolic DNA and activates type I interferon signaling and plays an essential role in antitumor immunity. However, it remains unclear whether the cGAS-mediated antitumor activity is affected by nutrient status. Here, our study reports that methionine deprivation enhances cGAS activity by blocking its methylation, which is catalyzed by methyltransferase SUV39H1. We further show that methylation enhances the chromatin sequestration of cGAS in a UHRF1-dependent manner. Blocking cGAS methylation enhances cGAS-mediated antitumor immunity and suppresses colorectal tumorigenesis. Clinically, cGAS methylation in human cancers correlates with poor prognosis. Thus, our results indicate that nutrient stress promotes cGAS activation via reversible methylation, and suggest a potential therapeutic strategy for targeting cGAS methylation in cancer treatment.


Asunto(s)
Cromatina , Metionina , Humanos , Cromatina/genética , Metionina/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , ADN , Inmunidad Innata , Desmetilación , Proteínas Potenciadoras de Unión a CCAAT/genética , Ubiquitina-Proteína Ligasas/genética
13.
JHEP Rep ; 5(10): 100843, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37675273

RESUMEN

Background & Aims: Exploiting key regulators responsible for hepatocarcinogenesis is of great importance for the prevention and treatment of hepatocellular carcinoma (HCC). However, the key players contributing to hepatocarcinogenesis remain poorly understood. We explored the molecular mechanisms underlying the carcinogenesis and progression of HCC for the development of potential new therapeutic targets. Methods: The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) and Genotype-Tissue Expression (GTEx) databases were used to identify genes with enhanced expression in the liver associated with HCC progression. A murine liver-specific Ftcd knockout (Ftcd-LKO) model was generated to investigate the role of formimidoyltransferase cyclodeaminase (FTCD) in HCC. Multi-omics analysis of transcriptomics, metabolomics, and proteomics data were applied to further analyse the molecular effects of FTCD expression on hepatocarcinogenesis. Functional and biochemical studies were performed to determine the significance of loss of FTCD expression and the therapeutic potential of Akt inhibitors in FTCD-deficient cancer cells. Results: FTCD is highly expressed in the liver but significantly downregulated in HCC. Patients with HCC and low levels of FTCD exhibited worse prognosis, and patients with liver cirrhosis and low FTCD levels exhibited a notable higher probability of developing HCC. Hepatocyte-specific knockout of FTCD promoted both chronic diethylnitrosamine-induced and spontaneous hepatocarcinogenesis in mice. Multi-omics analysis showed that loss of FTCD affected fatty acid and cholesterol metabolism in hepatocarcinogenesis. Mechanistically, loss of FTCD upregulated peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element-binding protein 2 (SREBP2) by regulating the PTEN/Akt/mTOR signalling axis, leading to lipid accumulation and hepatocarcinogenesis. Conclusions: Taken together, we identified a FTCD-regulated lipid metabolic mechanism involving PPARγ and SREBP2 signaling in hepatocarcinogenesis and provide a rationale for therapeutically targeting of HCC driven by downregulation of FTCD. Impact and implications: Exploiting key molecules responsible for hepatocarcinogenesis is significant for the prevention and treatment of HCC. Herein, we identified formimidoyltransferase cyclodeaminase (FTCD) as the top enhanced gene, which could serve as a predictive and prognostic marker for patients with HCC. We generated and characterised the first Ftcd liver-specific knockout murine model. We found loss of FTCD expression upregulated peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element-binding protein 2 (SREBP2) by regulating the PTEN/Akt/mTOR signalling axis, leading to lipid accumulation and hepatocarcinogenesis, and provided a rationale for therapeutic targeting of HCC driven by downregulation of FTCD.

14.
J Proteome Res ; 11(12): 5630-41, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23106643

RESUMEN

Affinity-purification mass spectrometry (AP-MS) is the preeminent technique for identification of eukaryotic protein complexes in vivo. AP-MS workflows typically express epitope-tagged bait proteins, immunopurify, and then identify associated protein complexes using mass spectrometry. However, challenges of existing strategies include the construction of expression vectors for large open reading frames and the possibility that overexpression of bait proteins may result in expression of nonphysiological levels of the bait protein with concomitant perturbation of endogenous protein complexes. To address these issues, we use human cell lines with epitope-tagged endogenous genes as AP-MS substrates to develop a platform that we call "knock-in AP-MS", thereby avoiding the challenges of expression vector construction and ensuring that expression of tagged proteins is driven by endogenous regulatory mechanisms. Using three different bait genes (MRE11A, DNMT1 and APC), we show that cell lines expressing epitope-tagged endogenous genes make good substrates for sensitive and reproducible identification of protein interactions using AP-MS. In particular, we identify novel interactors of the important oncoprotein Adenomatous Polyposis Coli (APC), including an interaction with Flightless-1 homologue (FLII) that is enriched in nuclear fractions.


Asunto(s)
Cromatografía de Afinidad/métodos , Neoplasias del Colon/patología , Mapeo Epitopo/métodos , Genoma Humano , Espectrometría de Masas/métodos , Complejos Multiproteicos/aislamiento & purificación , Proteínas Adaptadoras Transductoras de Señales/aislamiento & purificación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/patología , Neoplasias del Colon/metabolismo , Citoplasma/metabolismo , Citoplasma/patología , Dependovirus , Vectores Genéticos , Células HCT116 , Células HEK293 , Humanos , Proteínas de Microfilamentos/aislamiento & purificación , Proteínas de Microfilamentos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/aislamiento & purificación , Proteínas de Neoplasias/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteómica/métodos , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Transactivadores , Transfección
15.
Plant J ; 68(2): 302-13, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21707801

RESUMEN

NAC transcription factors play important roles in plant growth, development and stress responses. Previously, we identified multiple NAC genes in soybean (Glycine max). Here, we identify the roles of two genes, GmNAC11 and GmNAC20, in stress responses and other processes. The two genes were differentially induced by multiple abiotic stresses and plant hormones, and their transcripts were abundant in roots and cotyledons. Both genes encoded proteins that localized to the nucleus and bound to the core DNA sequence CGT[G/A]. In the protoplast assay system, GmNAC11 acts as a transcriptional activator, whereas GmNAC20 functions as a mild repressor; however, the C-terminal end of GmANC20 has transcriptional activation activity. Over-expression of GmNAC20 enhances salt and freezing tolerance in transgenic Arabidopsis plants; however, GmNAC11 over-expression only improves salt tolerance. Over-expression of GmNAC20 also promotes lateral root formation. GmNAC20 may regulate stress tolerance through activation of the DREB/CBF-COR pathway, and may control lateral root development by altering auxin signaling-related genes. GmNAC11 probably regulates DREB1A and other stress-related genes. The roles of the two GmNAC genes in stress tolerance were further analyzed in soybean transgenic hairy roots. These results provide a basis for genetic manipulation to improve the agronomic traits of important crops.


Asunto(s)
Regulación de la Expresión Génica de las Plantas/genética , Glycine max/metabolismo , Proteínas de Plantas/metabolismo , Estrés Fisiológico/fisiología , Factores de Transcripción/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Núcleo Celular/metabolismo , Cotiledón/genética , Cotiledón/crecimiento & desarrollo , Cotiledón/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Sequías , Flores/genética , Flores/crecimiento & desarrollo , Flores/metabolismo , Congelación , Proteínas Fluorescentes Verdes , Ácidos Indolacéticos/metabolismo , Motivos de Nucleótidos/genética , Reguladores del Crecimiento de las Plantas/metabolismo , Proteínas de Plantas/genética , Raíces de Plantas/genética , Raíces de Plantas/crecimiento & desarrollo , Raíces de Plantas/metabolismo , Tallos de la Planta/genética , Tallos de la Planta/crecimiento & desarrollo , Tallos de la Planta/metabolismo , Plantas Modificadas Genéticamente , Protoplastos , Tolerancia a la Sal , Plantones/genética , Plantones/crecimiento & desarrollo , Plantones/metabolismo , Cloruro de Sodio/farmacología , Proteínas de Soja/genética , Proteínas de Soja/metabolismo , Glycine max/genética , Glycine max/crecimiento & desarrollo , Factores de Transcripción/genética , Activación Transcripcional
16.
Plant Cell Environ ; 35(6): 1156-70, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22220579

RESUMEN

WRKY-type transcription factors are involved in multiple aspects of plant growth, development and stress response. WRKY genes have been found to be responsive to abiotic stresses; however, their roles in abiotic stress tolerance are largely unknown especially in crops. Here, we identified stress-responsive WRKY genes from wheat (Triticum aestivum L.) and studied their functions in stress tolerance. Forty-three putative TaWRKY genes were identified and two multiple stress-induced genes, TaWRKY2 and TaWRKY19, were further characterized. TaWRKY2 and TaWRKY19 are nuclear proteins, and displayed specific binding to typical cis-element W box. Transgenic Arabidopsis plants overexpressing TaWRKY2 exhibited salt and drought tolerance compared with controls. Overexpression of TaWRKY19 conferred tolerance to salt, drought and freezing stresses in transgenic plants. TaWRKY2 enhanced expressions of STZ and RD29B, and bound to their promoters. TaWRKY19 activated expressions of DREB2A, RD29A, RD29B and Cor6.6, and bound to DREB2A and Cor6.6 promoters. The two TaWRKY proteins may regulate the downstream genes through direct binding to the gene promoter or via indirect mechanism. Manipulation of TaWRKY2 and TaWRKY19 in wheat or other crops should improve their performance under various abiotic stress conditions.


Asunto(s)
Arabidopsis/fisiología , Proteínas de Plantas/metabolismo , Estrés Fisiológico , Factores de Transcripción/metabolismo , Triticum/genética , Arabidopsis/genética , Clonación Molecular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Sequías , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Presión Osmótica , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/fisiología , Regiones Promotoras Genéticas , Cloruro de Sodio/farmacología , Factores de Transcripción/genética
17.
Foods ; 11(7)2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35407005

RESUMEN

To provide necessary information for further pasteurization experiments and computer simulations based on radio frequency (RF) and microwave (MW) energy, dielectric and thermal properties of walnut components were measured at frequencies between 10 and 3000 MHz, temperatures between 20 and 80 °C, and moisture contents of whole walnuts between 8.04% and 20.01% on a dry basis (d.b.). Results demonstrated that dielectric constants and loss factors of walnut kernels and shells decreased dramatically with raised frequency within the RF range from 10 to 300 MHz, but then reduced slightly within the MW range from 300 to 3000 MHz. Dielectric constant, loss factor, specific heat capacity, and thermal conductivity increased with raised temperature and moisture content. Dielectric loss factors of kernels were greater than those of shells, leading to a higher RF or MW heating rate. Penetration depth of electromagnetic waves in walnut components was found to be greater at lower frequencies, temperatures, and moisture contents. The established regression models with experimental results could predict both dielectric and thermal properties with large coefficients of determination (R2 > 0.966). Therefore, this study offered essential data and effective guidance in developing and optimizing RF and MW pasteurization techniques for walnuts using both experiments and mathematical simulations.

18.
CNS Neurosci Ther ; 28(3): 332-342, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35044099

RESUMEN

BACKGROUND: The compatibility of deep brain stimulation (DBS) hardware and MRI scans has greatly improved the diagnostic rate of postoperative peri-lead edema (PLE). However, the etiology, incidence, and prognostic outcomes of this complication have not been established. OBJECTIVE: The incidence of PLE and associated symptoms, the process of occurrence and progression of this complication, as well as treatment strategies were evaluated. METHODS: We conducted a Preferred Reporting Items for Systematic Reviews and Meta-Analyses compliant systematic review of all studies that reported on incidences of PLE and associated symptoms after DBS implantation. Through systematic literature review, we evaluated its causes, neuropsychiatric symptoms, duration, treatment methods, and prognostic outcomes. RESULTS: Our search retrieved 10 articles, including 5 articles on PLE and 10 articles on symptomatic PLE. The incidence of PLE was 35.8% (95% CI: 17.0%-54.6%), while the incidence of symptomatic PLE was 3.1% (95% CI: 1.5%-4.7%) accounting for 8.7% of PLE. CONCLUSIONS: This complication is not as rare as previously reported. Therefore, it requires significant attention after DBS implantation. The correlation between its causes, duration, symptoms, and the area involved in edema should be assessed in long-term prospective clinical studies with large sample sizes.


Asunto(s)
Edema Encefálico , Estimulación Encefálica Profunda , Edema Encefálico/diagnóstico por imagen , Edema Encefálico/epidemiología , Edema Encefálico/etiología , Estimulación Encefálica Profunda/efectos adversos , Estimulación Encefálica Profunda/métodos , Edema/etiología , Humanos , Imagen por Resonancia Magnética/efectos adversos , Estudios Prospectivos
19.
Front Hum Neurosci ; 16: 849069, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35601911

RESUMEN

Background: The past four decades have seen the growing use of tissue or cell transplants in Parkinson's disease (PD) treatment. Parkinson's cell therapy is a promising new treatment; however, efficacy of cell transplantation for Parkinson's disease are entirely unclear. Objective: To conduct a meta-analysis and a systematic review of the efficacy of cell therapy in patients with PD. Methods: A systematic literature review and meta-analysis of 10 studies were performed to assess the efficacy of cell therapy in Parkinson's patients. To achieve this, we compared the change in Unified Parkinson's Disease Rating Scale (UPDRS) II and III scale scores to baseline and assessed the incidence of transplant-related adverse events. The MINORS score and the I2 index were applied to evaluate the quality of studies between-study heterogeneity, respectively. Results: The literature search yielded 10 articles (n = 120). The improvement in motor function based on the UPDRSIII assessment was -14.044 (95% CI: -20.761, -7.327) (p < 0.001), whereas improvement in daily living ability based on the UPDRSII assessment was -5.661 (95% CI: -7.632, -3.689) (p < 0.001). Conclusion: The present findings demonstrate important clues on the therapeutic effect of cell therapy in alleviating motor impairment and daily living ability in PD patients.

20.
Nat Commun ; 13(1): 1974, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35418124

RESUMEN

PI3Ks consist of p110 catalytic subunits and p85 regulatory subunits. PIK3CA, encoding p110α, is frequently mutated in human cancers. Most PIK3CA mutations are clustered in the helical domain or the kinase domain. Here, we report that p85ß disassociates from p110α helical domain mutant protein and translocates into the nucleus through a nuclear localization sequence (NLS). Nuclear p85ß recruits deubiquitinase USP7 to stabilize EZH1 and EZH2 and enhances H3K27 trimethylation. Knockout of p85ß or p85ß NLS mutant reduces the growth of tumors harboring a PIK3CA helical domain mutation. Our studies illuminate a novel mechanism by which PIK3CA helical domain mutations exert their oncogenic function. Finally, a combination of Alpelisib, a p110α-specific inhibitor, and an EZH inhibitor, Tazemetostat, induces regression of xenograft tumors harboring a PIK3CA helical domain mutation, but not tumors with either a WT PIK3CA or a PIK3CA kinase domain mutation, suggesting that the drug combination could be an effective therapeutic approach for PIK3CA helical domain mutant tumors.


Asunto(s)
Neoplasias , Fosfatidilinositol 3-Quinasas , Carcinogénesis/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Oncogenes , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Peptidasa Específica de Ubiquitina 7/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA