Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 825
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(1-2): 213-226.e18, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30554876

RESUMEN

Transcriptional regulation in metazoans occurs through long-range genomic contacts between enhancers and promoters, and most genes are transcribed in episodic "bursts" of RNA synthesis. To understand the relationship between these two phenomena and the dynamic regulation of genes in response to upstream signals, we describe the use of live-cell RNA imaging coupled with Hi-C measurements and dissect the endogenous regulation of the estrogen-responsive TFF1 gene. Although TFF1 is highly induced, we observe short active periods and variable inactive periods ranging from minutes to days. The heterogeneity in inactive times gives rise to the widely observed "noise" in human gene expression and explains the distribution of protein levels in human tissue. We derive a mathematical model of regulation that relates transcription, chromosome structure, and the cell's ability to sense changes in estrogen and predicts that hypervariability is largely dynamic and does not reflect a stable biological state.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Expresión Génica/fisiología , Transcripción Genética/fisiología , Receptor alfa de Estrógeno/metabolismo , Estrógenos , Expresión Génica/genética , Humanos , Modelos Teóricos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Análisis de la Célula Individual/métodos , Transcripción Genética/genética , Activación Transcripcional/fisiología , Factor Trefoil-1/genética
2.
Gastroenterology ; 167(3): 505-521.e19, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38583723

RESUMEN

BACKGROUND & AIMS: Gastric cancer is often accompanied by a loss of mucin 6 (MUC6), but its pathogenic role in gastric carcinogenesis remains unclear. METHODS: Muc6 knockout (Muc6-/-) mice and Muc6-dsRED mice were newly generated. Tff1Cre, Golph3-/-, R26-Golgi-mCherry, Hes1flox/flox, Cosmcflox/flox, and A4gnt-/- mice were also used. Histology, DNA and RNA, proteins, and sugar chains were analyzed by whole-exon DNA sequence, RNA sequence, immunohistochemistry, lectin-binding assays, and liquid chromatography-mass spectrometry analysis. Gastric organoids and cell lines were used for in vitro assays and xenograft experiments. RESULTS: Deletion of Muc6 in mice spontaneously causes pan-gastritis and invasive gastric cancers. Muc6-deficient tumor growth was dependent on mitogen-activated protein kinase activation, mediated by Golgi stress-induced up-regulation of Golgi phosphoprotein 3. Glycomic profiling revealed aberrant expression of mannose-rich N-linked glycans in gastric tumors, detected with banana lectin in association with lack of MUC6 expression. We identified a precursor of clusterin as a binding partner of mannose glycans. Mitogen-activated protein kinase activation, Golgi stress responses, and aberrant mannose expression are found in separate Cosmc- and A4gnt-deficient mouse models that lack normal O-glycosylation. Banana lectin-drug conjugates proved an effective treatment for mannose-rich murine and human gastric cancer. CONCLUSIONS: We propose that Golgi stress responses and aberrant glycans are important drivers of and promising new therapeutic targets for gastric cancer.


Asunto(s)
Ratones Noqueados , Mucina 6 , Neoplasias Gástricas , Animales , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Glicosilación , Humanos , Mucina 6/metabolismo , Mucina 6/genética , Ratones , Línea Celular Tumoral , Carcinogénesis/metabolismo , Carcinogénesis/genética , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Factor Trefoil-1/metabolismo , Factor Trefoil-1/genética , Organoides/metabolismo , Aparato de Golgi/metabolismo , Mucinas Gástricas/metabolismo , Modelos Animales de Enfermedad
3.
J Transl Med ; 22(1): 917, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39385235

RESUMEN

Spread through air spaces (STAS) is a recognized aggressive pattern in lung cancer, serving as a crucial risk factor for postoperative recurrence. However, its phenotype and related spatial structure have remained elusive. To address these limitations, we conducted a comprehensive study based on spatial data, analyzing over 30,000 spots from 14 non-STAS samples and one STAS sample. We observed increased proliferation activities and angiogenesis in STAS, identifying S100P as a potential biomarker for STAS. Furthermore, our investigation into the heterogeneity of STAS tumor cells revealed a subset identified as S100P + TFF1 +, exhibiting a negative impact on patients' survival in public datasets. This subtype exhibited the highest activities in the TGFb and hypoxia, suggesting its potential pro-tumor role within the tumor microenvironment. To assess the role of S100P + TFF1 + tumor cells in therapy response, we included data from two clinical trial cohorts (BPI-7711 for EGFR-TKI therapy and ORIENT-3 for immunotherapy). The presence of S100P + TFF1 + tumor cells correlated with worse responses to both EGFR-TKI therapy and immunotherapy. Notably, TFF1 emerged as a serum marker for predicting EGFR-TKI response. Cell-cell communication analysis revealed that the TGFb signaling pathway was the most activated in S100P + TFF1 + tumor cells, with TGFB2-TGFBR2 identified as the main ligand-receptor pair. This was further validated by multiplex immunofluorescence performed on twenty NSCLC samples. In summary, our study identified S100P as the biomarker for STAS and highlighted the adverse role of S100P + TFF1 + tumor cells in survival outcomes.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Factor Trefoil-1 , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Factor Trefoil-1/metabolismo , Proteínas de Unión al Calcio/metabolismo , Resultado del Tratamiento , Proliferación Celular/efectos de los fármacos , Masculino , Femenino , Proteínas de Neoplasias
4.
PLoS Genet ; 16(1): e1008516, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31905229

RESUMEN

Unliganded Estrogen receptor alpha (ERα) has been implicated in ligand-dependent gene regulation. Upon ligand exposure, ERα binds to several EREs relatively proximal to the pre-marked, unliganded ERα-bound sites and affects transient but robust gene expression. However, the underlying mechanisms are not fully understood. Here we demonstrate that upon ligand stimulation, persistent sites interact extensively, via chromatin looping, with the proximal transiently ERα-bound sites, forming Ligand Dependent ERα Enhancer Cluster in 3D (LDEC). The E2-target genes are regulated by these clustered enhancers but not by the H3K27Ac super-enhancers. Further, CRISPR-based deletion of TFF1 persistent site disrupts the formation of its LDEC resulting in the loss of E2-dependent expression of TFF1 and its neighboring genes within the same TAD. The LDEC overlap with nuclear ERα condensates that coalesce in a ligand and persistent site dependent manner. Furthermore, formation of clustered enhancers, as well as condensates, coincide with the active phase of signaling and their later disappearance results in the loss of gene expression even though persistent sites remain bound by ERα. Our results establish, at TFF1 and NRIP1 locus, a direct link between ERα condensates, ERα enhancer clusters, and transient, but robust, gene expression in a ligand-dependent fashion.


Asunto(s)
Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética , Eliminación de Gen , Histonas/metabolismo , Humanos , Ligandos , Células MCF-7 , Factor Trefoil-1/genética
5.
Int J Mol Sci ; 24(16)2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37628863

RESUMEN

Tff1 is a typical gastric peptide secreted together with the mucin, Muc5ac. Tff1-deficient (Tff1KO) mice are well known for their prominent gastric phenotype and represent a recognized model for antral tumorigenesis. Notably, intestinal abnormalities have also been reported in the past in these animals. Here, we have compared the expression of selected genes in Tff1KO mice and their corresponding wild-type littermates (RT-PCR analyses), focusing on different mucosal protection systems along the murine intestine. As hallmarks, genes were identified with maximum expression in the proximal colon and/or the duodenum: Agr2, Muc6/A4gnt/Tff2, Tff1, Fut2, Gkn2, Gkn3, Duox2/Lpo, Nox1. This is indicative of different protection systems such as Tff2/Muc6, Tff1-Fcgbp, gastrokines, fucosylation, and reactive oxygen species (ROS) in the proximal colon and/or duodenum. Few significant transcriptional changes were observed in the intestine of Tff1KO mice when compared with wild-type littermates, Clca1 (Gob5), Gkn1, Gkn2, Nox1, Tff2. We also analyzed the expression of Tff1, Tff2, and Tff3 in the pancreas, liver, and lung of Tff1KO and wild-type animals, indicating a cross-regulation of Tff gene expression. Furthermore, on the protein level, heteromeric Tff1-Fcgbp and various monomeric Tff1 forms were identified in the duodenum and a high-molecular-mass Tff2/Muc6 complex was identified in the proximal colon (FPLC, proteomics).


Asunto(s)
Intestinos , Animales , Ratones , Duodeno , Colon , Animales Salvajes , Transporte Biológico , Factor Trefoil-1/genética
6.
Lab Invest ; 102(8): 885-895, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35279702

RESUMEN

Trefoil factor family 1 (TFF1) is one of three members of the trefoil factor family that are abundantly expressed in the gastrointestinal mucosal epithelium. Recent studies have shown that TFF1 acts as a tumor suppressor in gastric, pancreatic and hepatocellular carcinogenesis; however, little is known about its function in esophageal carcinogenesis, especially in esophageal adenocarcinoma (EAC). Barrett's epithelium is the metaplastic columnar epithelium of the esophagus and a known premalignant lesion of EAC. To investigate the role of TFF1 in EAC development, a mouse model of Barrett's epithelium was employed, and human specimens of EAC were assessed by immunohistochemistry (IHC) and methylation-specific PCR. Wild-type (WT) mice underwent gastrojejunostomy on the forestomach, resulting in the development of Barrett's epithelium-like (BE-like) epithelium adjacent to the anastomotic site. BE-like epithelium in these mice expressed TFF1, indicating the association of TFF1 with esophageal adenocarcinoma. TFF1-knockout (TFF1KO) mice underwent the same procedure as well, revealing that a deficiency in TFF1 resulted in the development of adenocarcinoma in the anastomotic site, presumably from BE-like epithelium. IHC of human samples revealed strong TFF1 expression in Barrett's epithelium, which was lost in some EACs, confirming the association between TFF1 and EAC development. Aberrant DNA hypermethylation in TFF1 promoter lesions was detected in TFF1-negative human EAC samples, further confirming not only the role of TFF1 in EAC but also the underlying mechanisms of TFF1 regulation. In addition, IHC revealed the nuclear translocation of ß-catenin in human and mouse EAC, suggesting that activation of the Wnt/ß-catenin pathway was induced by the loss of TFF1. In conclusion, these results indicate that TFF1 functions as a tumor suppressor to inhibit the development of esophageal carcinogenesis from Barrett's epithelium.


Asunto(s)
Adenocarcinoma , Esófago de Barrett , Neoplasias Esofágicas , Factor Trefoil-1 , Adenocarcinoma/etiología , Adenocarcinoma/genética , Animales , Esófago de Barrett/complicaciones , Esófago de Barrett/genética , Carcinogénesis , Metilación de ADN , Epitelio/metabolismo , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Factor Trefoil-1/genética , Vía de Señalización Wnt , beta Catenina
7.
Biochem Biophys Res Commun ; 588: 75-82, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34952473

RESUMEN

Germline mutations to the breast cancer 2 (BRCA2) gene have been associated with hereditary breast cancer. In addition to estrogen uptake, BRCA2 expression increases in the S phase of the cell cycle and largely contributes to DNA damage repair associated with DNA replication. However, the role of BRCA2 in estrogen induction remains unclear. An expression plasmid was created to induce BRCA2 activation upon the addition of estradiol by introducing mutations to the binding sequences for the transcription factors USF1, E2F1, and NF-κB within the promoter region of BRCA2. Then, the estrogen receptor (ER) sites of the proteins that interact with BRCA2 upon the addition of estradiol were identified. Both proteins were bound by the helical domain of BRCA2 and activation function-2 of the ER, suggesting that this binding may regulate the transcriptional activity of pS2, a target gene of the estradiol-ER, by suppressing the binding of SRC-1, a coactivator required for activation of the transcription factor.


Asunto(s)
Proteína BRCA2/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Proteínas/genética , Transcripción Genética , Factor Trefoil-1/genética , Proteína BRCA2/química , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Coactivador 1 de Receptor Nuclear/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos , Proteínas/metabolismo , Factores de Transcripción/metabolismo , Factor Trefoil-1/metabolismo
8.
Exp Eye Res ; 217: 108969, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35114215

RESUMEN

Diabetic retinopathy (DR) represents a major complication of diabetes, and molecular mechanisms related to vascular dysfunction, particularly endothelial dysfunction, in DR remains unclear. In the present work, we generated a DR animal model using mice and a cell model in mouse retinal microvascular endothelial cells (mRMECs) to examine the role of Trefoil factor family 1 (Tff1) in DR. Tff1 was poorly expressed in DR mice and high glucose (HG)-treated mRMECs. Overexpression of Tff1 significantly attenuated streptozotocin-induced retinal proliferation and angiogenesis in DR mice and reduced the secretion of inflammatory factors. In HG-treated mRMECs, overexpression of Tff1 remarkably reduced the proliferation and angiogenesis of mRMECs. In further experiments, we found that Tff1 was transcriptionally repressed by Runt-related transcription factor 1 (Runx1) directly, and Tff1 expression was indirectly modulated by Runx1 via the core-binding factor subunit beta (CBF-ß)/nuclear factor, erythroid 2/microRNA-423-5p axis and the CBF-ß/estrogen receptor 1 (ESR1) axis. Moreover, Tff1 could inhibit the activation of NF-κB signaling pathway, which in turn attenuated retinal endothelial cell proliferation and angiogenesis. It was thus proposed that Runx1/Tff1/NF-κB axis may be a potential target for the treatment strategy of DR, and further studies are needed.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Diabetes Mellitus , Retinopatía Diabética , MicroARNs , Factor Trefoil-1 , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Diabetes Mellitus/metabolismo , Retinopatía Diabética/metabolismo , Células Endoteliales/metabolismo , Ratones , MicroARNs/metabolismo , FN-kappa B/metabolismo , Neovascularización Patológica/metabolismo , Retina/metabolismo , Factor Trefoil-1/metabolismo
9.
Mol Biol Rep ; 49(10): 10127-10131, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36057754

RESUMEN

INTRODUCTION: Trefoil Factor 1 (TFF1) is a secretory peptide with gastrointestinal protective functions. Abnormal TFF1 expression is reported in some cancers and functional promoter polymorphism in TFF1 is believed to be associated with risk of gastric cancer. We evaluated rs3761376 in a sample of Iranian patients with colorectal cancer. METHODS: Peripheral blood samples were taken from pathology confirmed cases of colorectal cancer and healthy volunteers. Genotyping was carried out using Restriction Fragment Length Polymorphism (RFLP) PCR. Any association with clinicopathologic data was assessed by SPSS version 19. RESULTS: A total of 245 participants, including 122 patients with cancer and 123 non-cancer subjects were enrolled. Age, body mass index, and smoking habits were not significantly different between the two groups (P > 0.05). Distribution of TFF1 genotypes was not found to be associated with colorectal cancer. However, distant metastasis was more prevalent in carriers of the mutant allele. CONCLUSION: TFF1 rs3761376 was not associated with colorectal cancer but it may be involved in metastasis. Therefore, further investigation is warranted to determine this relationship.


Asunto(s)
Neoplasias Colorrectales , Polimorfismo de Nucleótido Simple , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Humanos , Irán , Péptidos/genética , Péptidos/metabolismo , Polimorfismo de Nucleótido Simple/genética , Factor Trefoil-1/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
BMC Urol ; 22(1): 127, 2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-35987613

RESUMEN

Trefoil Factor 1 (TFF1) is considered to be able to inhibit the formation of kidney stone. However, genetic variants in TFF1 and corresponding function in kidney stone development are still not well studied. In this study, the discovery set including 230 cases and 250 controls was used to analyze the association between seven tagSNPs of TFF1 gene and the nephrolithiasis risk. Further evaluation was confirmed by the validation set comprising 307 cases and 461 controls. The consequences of the two-stage case-control study indicated that individuals with the rs3761376 A allele have significantly increased nephrolithiasis risk than those with the GG genotypes [adjusted odds ratio (OR) = 1.35, 95% confidence interval (CI) = 1.05-1.73]. Moreover, we also carried out a stratified analysis and found the increased nephrolithiasis risks at A allele among males, overweight individuals, no hypertensive individuals, nondiabetic individuals, smokers, and drinkers. In the following functional experiments, the notably lower expression of TFF1 was exhibited by the vectors carrying A allele compared with those carrying G allele in both luciferase (P = 0.022) and expression vectors (P = 0.041). In addition to tissue detection, we confirmed a significant inverse association of rs3761376 G > A and TFF1 gene expression (P < 0.001). These results suggest that TFF1 rs3761376 may serve as a potential biomarker to predict the risk of nephrolithiasis.


Asunto(s)
Cálculos Renales , Nefrolitiasis , Factor Trefoil-1 , Estudios de Casos y Controles , China , Humanos , Cálculos Renales/genética , Masculino , Nefrolitiasis/genética , Polimorfismo de Nucleótido Simple/genética , Factor Trefoil-1/genética
11.
Oral Dis ; 28(4): 1240-1249, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-33660336

RESUMEN

OBJECTIVE: This study aimed to investigate the levels of trefoil factor family (TFF)-1, TFF-3 and interleukin (IL)-1ß in gingival crevicular fluid (GCF), saliva and serum of patients with gingivitis, stage 3 periodontitis and healthy individuals. MATERIALS AND METHODS: A total of 100 individuals consisting of 25 periodontally healthy, 25 gingivitis and 50 stage 3 periodontitis, were enrolled in the study. Clinical periodontal examinations were recorded and GCF, saliva and serum samples were obtained. TFF-1, TFF-3 and IL-1ß were measured by ELISA. RESULTS: TFF-1 and TFF-3 levels in both GCF, saliva and serum were higher in periodontitis patients than healthy controls (p < .001) and gingivitis group (p < .01). The levels of these peptides in all biofluids were similar between gingivitis and healthy control groups (p > .05). GCF, saliva and serum IL-1ß levels were also higher in periodontitis patients than the controls (p < .01). Periodontitis patients had elevated GCF and saliva IL-ß levels than gingivitis group (p < .001). CONCLUSION: Elevated TFF-1 and TFF-3 levels both locally and systemically in periodontitis in parallel to increased IL-1ß levels might suggest that these peptides are involved in host response during the periodontal tissue destruction.


Asunto(s)
Periodontitis Crónica , Gingivitis , Factores Trefoil , Periodontitis Crónica/metabolismo , Líquido del Surco Gingival , Gingivitis/metabolismo , Humanos , Saliva/metabolismo , Factor Trefoil-1/metabolismo , Factor Trefoil-3/metabolismo , Factores Trefoil/metabolismo , Regulación hacia Arriba
12.
Biochem Genet ; 60(1): 315-335, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34219206

RESUMEN

Some circular RNAs (circRNAs) have been verified to act as essential regulators in the progression of breast cancer (BC). We aimed to investigate the role of circRNA trefoil factor 1 (circ-TFF1) in BC progression. The expression of circ-TFF1, microRNA-338-3p (miR-338-3p) and fibroblast growth factor receptor 1 (FGFR1) mRNA was measured by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was evaluated by methylthiazolyldiphenyl-tetrazolium bromide (MTT), colony formation, and 5-Ethynyl-2'-deoxyuridine (EDU) assays. Cell apoptosis and invasion were assessed by flow cytometry and transwell assay, respectively. Cellular glycolysis, including glucose consumption, lactate production, and ATP/ADP ratio, was detected by commercial kits. All protein levels were measured by western blot assay. The relationship between miR-338-3p and circ-TFF1 or FGFR1 was predicted by online bioinformatics tool and verified by dual-luciferase reporter assay. Xenograft tumor model was established to verify the function of circ-TFF1 in vivo. Circ-TFF1 was overexpressed in BC tissues and cells. Circ-TFF1 knockdown inhibited cell proliferation, invasion and glycolysis and induced apoptosis in BC cells. Circ-TFF1 acted as a sponge of miR-338-3p, and the effects of circ-TFF1 knockdown on BC cell proliferation, apoptosis, invasion, and glycolysis were abolished by miR-338-3p inhibition. FGFR1 was confirmed to be a target gene of miR-338-3p, and miR-338-3p played a tumor-suppressive role in BC by targeting FGFR1. Moreover, circ-TFF1 regulated FGFR1 expression by targeting miR-338-3p. Additionally, circ-TFF1 knockdown hampered tumorigenesis in vivo. Circ-TFF1 knockdown suppressed BC progression by regulating miR-338-3p/FGFR1 axis, providing a promising therapeutic target for BC.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Neoplasias de la Mama/genética , Femenino , Humanos , MicroARNs/genética , ARN Circular , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Factor Trefoil-1
13.
Biochem Genet ; 60(6): 2155-2170, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35279761

RESUMEN

Breast cancer (BC) is a common malignant tumor, and circular RNA-trefoil factor 1 (circ-TFF1; hsa_circ_0061825) has been found to be highly expressed in BC tissues and cells and is associated with the poor prognosis of BC patients. However, the interaction between circ-TFF1 and microRNA in BC has not been studied. Quantitative real-time PCR was used to detect the expression of circ-TFF1, miR-129-2-3p, and interleukin (IL)-1 receptor-associated kinase 1 (IRAK1). Through the detection of cell proliferation, migration, invasion, tube formation, and apoptosis, cell function was assessed. The expression levels of angiogenesis-related proteins were detected by western blot. The interaction between miR-129-2-3p and circ-TFF1 or IRAK1 was verified by dual-luciferase reporter assay and RNA immunoprecipitation assay. Xenotransplantation experiments were used to confirm the function of circ-TFF1 in vivo. Circ-TFF1 and IRAK1 were significantly high expressed in BC tissues and cells. Silencing of circ-TFF1 reduced the proliferation, migration, invasion and tube formation, while increased the apoptosis of MDA-MB-361 and SK-Br-3 cells. MiR-129-2-3p was a target of circ-TFF1. Silencing of circ-TFF1 inhibited the malignant behavior of BC cells by releasing miR-129-2-3p. In addition, IRAK1 was a target of miR-129-2-3p. Overexpression of IRAK1 partially restored the inhibitory effect of miR-129-2-3p on cell progression. Animal experiments confirmed the anti-tumor effect of circ-TFF1 knockdown in vivo. Circ-TFF1 regulated the expression of IRAK1 by sponging miR-129-2-3p, thereby, promoting the development of BC. These data provided a novel targeted therapy for BC.


Asunto(s)
MicroARNs , Neoplasias , Animales , Factor Trefoil-1/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , MicroARNs/genética , Proliferación Celular/genética , Neoplasias/genética , Línea Celular Tumoral
14.
Int J Mol Sci ; 23(10)2022 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-35628183

RESUMEN

In 2020, gastric cancer was the fourth leading cause of cancer deaths globally. About 90% of gastric cancers are sporadic and the vast majority are correlated with Helicobacter pylori infection; whereas familial clustering is observed in about 10% of cases. Gastric cancer is now considered to be a disease originating from dysregulated self-renewal of the gastric glands in the setting of an inflammatory environment. The human stomach contains two types of gastric units, which show bi-directional self-renewal from a complex variety of stem cells. This review focuses on recent progress concerning the characterization of the different stem cell populations and the mainly mesenchymal signals triggering their stepwise differentiation as well as the genesis of pre-cancerous lesions and carcinogenesis. Furthermore, a model is presented (Lectin-triggered Receptor Blocking Hypothesis) explaining the role of the lectin TFF1 as an antral tumor suppressor possibly regulating Lgr5+ antral stem cells in a paracrine or maybe autocrine fashion, with neighboring antral gland cells having a role as niche cells.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Epitelio/patología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Humanos , Lectinas , Neoplasias Gástricas/patología , Factor Trefoil-1
15.
J Sci Food Agric ; 102(3): 1255-1262, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34358346

RESUMEN

BACKGROUND: Artemisia capillaris is among the most abundantly used traditional medicines, utilized in East Asia to treat diverse illnesses, including gastrointestinal tract diseases. We previously reported that an aqueous extract of A. capillaris (AEAC) inhibited gastric inflammation induced by HCl/ethanol via reactive oxygen species scavenging and NF-κB downregulation. To date, the pharmacological potential of AEAC for promoting mucosal integrity has not been studied. RESULTS: Here, we report that a single treatment with AEAC increased mucus production, and repeated administration of AEAC abolished HCl/ethanol-induced mucosal injury in vivo. Single- and multiple-dose AEAC treatments measurably increased the expression of mucosal stabilizing factors in vivo, including mucin (MUC) 5 AC, MUC6, and trefoil factor (TFF) 1 and TFF2 (but not TFF3). AEAC also induced mucosal stabilizing factors in both SNU-601 cells and RGM cells through phosphorylation of extracellular signal-regulated kinases. CONCLUSION: Taken together, our results suggest that AEAC protects against HCl/ethanol-induced gastritis by upregulating MUCs and TFFs and stabilizing the mucosal epithelium. © 2021 Society of Chemical Industry.


Asunto(s)
Artemisia/química , Medicamentos Herbarios Chinos/farmacología , Mucosa Gástrica/efectos de los fármacos , Gastropatías/tratamiento farmacológico , Animales , Mucosa Gástrica/inmunología , Mucosa Gástrica/lesiones , Humanos , Masculino , Mucinas/genética , Mucinas/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Hojas de la Planta/química , Ratas , Ratas Sprague-Dawley , Gastropatías/genética , Gastropatías/inmunología , Factor Trefoil-1/genética , Factor Trefoil-1/inmunología
16.
J Neurosci ; 40(9): 1956-1974, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31980586

RESUMEN

TREM2 is an Alzheimer's disease (AD) risk gene expressed in microglia. To study the role of Trem2 in a mouse model of ß-amyloidosis, we compared PS2APP transgenic mice versus PS2APP mice lacking Trem2 (PS2APP;Trem2ko) at ages ranging from 4 to 22 months. Microgliosis was impaired in PS2APP;Trem2ko mice, with Trem2-deficient microglia showing compromised expression of proliferation/Wnt-related genes and marked accumulation of ApoE. Plaque abundance was elevated in PS2APP;Trem2ko females at 6-7 months; but by 12 or 19-22 months of age, it was notably diminished in female and male PS2APP;Trem2ko mice, respectively. Across all ages, plaque morphology was more diffuse in PS2APP;Trem2ko brains, and the Aß42:Aß40 ratio was elevated. The amount of soluble, fibrillar Aß oligomers also increased in PS2APP;Trem2ko hippocampi. Associated with these changes, axonal dystrophy was exacerbated from 6 to 7 months onward in PS2APP;Trem2ko mice, notwithstanding the reduced plaque load at later ages. PS2APP;Trem2ko mice also exhibited more dendritic spine loss around plaque and more neurofilament light chain in CSF. Thus, aggravated neuritic dystrophy is a more consistent outcome of Trem2 deficiency than amyloid plaque load, suggesting that the microglial packing of Aß into dense plaque is an important neuroprotective activity.SIGNIFICANCE STATEMENT Genetic studies indicate that TREM2 gene mutations confer increased Alzheimer's disease (AD) risk. We studied the effects of Trem2 deletion in the PS2APP mouse AD model, in which overproduction of Aß peptide leads to amyloid plaque formation and associated neuritic dystrophy. Interestingly, neuritic dystrophies were intensified in the brains of Trem2-deficient mice, despite these mice displaying reduced plaque accumulation at later ages (12-22 months). Microglial clustering around plaques was impaired, plaques were more diffuse, and the Aß42:Aß40 ratio and amount of soluble, fibrillar Aß oligomers were elevated in Trem2-deficient brains. These results suggest that the Trem2-dependent compaction of Aß into dense plaques is a protective microglial activity, limiting the exposure of neurons to toxic Aß species.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Axones/patología , Espinas Dendríticas/patología , Glicoproteínas de Membrana/genética , Fragmentos de Péptidos/metabolismo , Placa Amiloide/genética , Receptores Inmunológicos/genética , Factor Trefoil-1/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Neuritas/patología , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Placa Amiloide/patología
17.
Biochem Biophys Res Commun ; 563: 15-22, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34058470

RESUMEN

Helicobacter pylori infection is a crucial factor in the development of gastric cancer (GC). Molecular therapeutic targets and mechanisms contributing to H. pylori infection-associated GC induction are poorly understood and this study aimed to fill that research gap. We found that the nuclear receptor estrogen-related receptor gamma (ESRRG) is a candidate factor influencing H. pylori infection-driven GC. ESRRG suppressed H. pylori infection and cell growth induced by H. pylori infection in GC cells and organoid models In addition, H. pylori infection downregulates ESRRG expression. Gene expression profiling revealed that trefoil factor 1 (TFF1), a well-known tumor suppressor in GC, is a downstream target of ESRRG. Mechanistically, ESRRG directly binds to the TFF1 promoter and induces TFF1 gene expression. Furthermore, TFF1 activation by ESRRG was inhibited by nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/p65, which is induced by inflammation, such as by H. pylori infection. Our current study provides new molecular insights into how ESRRG regulates H. pylori infection, contributing to GC development. We suggest that modulation of ESRRG-suppressing H. pylori infection could be a therapeutic target for the treatment of GC patients.


Asunto(s)
Infecciones por Helicobacter/metabolismo , Receptores de Estrógenos/metabolismo , Neoplasias Gástricas/metabolismo , Factor Trefoil-1/metabolismo , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias Gástricas/patología
18.
Hepatology ; 72(2): 503-517, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31733149

RESUMEN

BACKGROUND AND AIMS: Recent studies have suggested that trefoil factor family 1 (TFF1) functions as a tumor suppressor in gastric and pancreatic carcinogenesis. APPROACH AND RESULTS: To investigate the role of TFF1 in hepatocarcinogenesis, we performed immunohistochemical staining of surgically resected human liver samples, transfected a TFF1 expression vector into hepatocellular carcinoma (HCC) cell lines, and employed a mouse model of spontaneous HCC development (albumin-cyclization recombination/Lox-Stop-Lox sequence-Kirsten rat sarcoma viral oncogene homologG12D [KC]); the model mouse strain was bred with a TFF1-knockout mouse strain to generate a TFF1-deficient HCC mouse model (KC/TFF1-/- ). TFF1 expression was found in some human samples with HCC. Interestingly, TFF1-positive cancer cells showed a staining pattern contradictory to that of proliferating cell nuclear antigen, and aberrant DNA hypermethylation in TFF1 promoter lesions was detected in HCC samples, indicating the tumor-suppressive role of TFF1. In vitro, induction of TFF1 expression resulted in impaired proliferative activity and enhanced apoptosis in HCC cell lines (HuH7, HepG2, and HLE). These anticancer effects of TFF1 were accompanied by the loss of nuclear ß-catenin expression, indicating inactivation of the ß-catenin signaling pathway by TFF1. In vivo, TFF1 deficiency in KC mice accelerated the early development and growth of HCC, resulting in poor survival rates. In addition, immunohistochemistry revealed that the amount of nuclear-localized ß-catenin was significantly higher in KC/TFF1-/- mice than in KC mice and that human HCC tissue showed contradictory expression patterns for ß-catenin and TFF1, confirming the in vitro observations. CONCLUSIONS: TFF1 might function as a tumor suppressor that inhibits the development of HCC by regulating ß-catenin activity.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Factor Trefoil-1/fisiología , Proteínas Supresoras de Tumor/fisiología , beta Catenina/fisiología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Células Tumorales Cultivadas
19.
Scand J Clin Lab Invest ; 81(6): 446-450, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34242119

RESUMEN

The trefoil factor family proteins: TFF1, TFF2 and TFF3 are secreted by epithelial cells in the respiratory tract. Here, we explore circulating concentrations of the trefoil factors in relation to lung cancer, age and lung function. We included 751 patients suspected of lung cancer. Lung cancer diagnosis was based on data reported to a national database. Serum TFF1, TFF2 and TFF3 concentrations were measured by ELISA, and spirometry was performed within ±3 days of blood sampling. Forced expiratory volume in the first second (FEV1) in relation to forced vital capacity (FVC), FEV1/FVC (a parameter used to quantify reduced lung function) was recorded. Lung cancer was diagnosed in 163 (22%) patients. Circulating concentrations of TFF3 (p = .021), but not TFF1 and TFF2, were significantly elevated in cancer patients. All three trefoil factors showed an increase in concentration with increasing age (p < .001) and declining lung function (p < .004). In the present cohort, concentrations of all three peptides were elevated compared with previous results published for healthy individuals. In conclusion, we report higher concentrations of TFF3 in patients with lung cancer, while increasing age and reduced lung function are associated with increasing concentrations of all trefoil factors in this specific patient population. The results emphasize that age and lung function should be taken into consideration when evaluating concentrations of trefoil factors in patients. However, the increases in trefoil factor concentrations were relatively small, and consequently, it is unlikely that circulating trefoil factor concentrations may have a role in the diagnosis of lung cancer and lung function impairment.


Asunto(s)
Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/fisiopatología , Derivación y Consulta , Pruebas de Función Respiratoria , Factor Trefoil-1/sangre , Factor Trefoil-2/sangre , Factor Trefoil-3/sangre , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Intervalos de Confianza , Estudios Transversales , Humanos , Neoplasias Pulmonares/patología , Persona de Mediana Edad , Adulto Joven
20.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673347

RESUMEN

Gastric cancer is considered one of the most common malignancies in humans and Helicobacter pylori infection is the major environmental risk factor of gastric cancer development. Given the high spread of this bacterium whose infection is mostly asymptomatic, H. pylori colonization persists for a long time, becoming chronic and predisposing to malignant transformation. The first defensive barrier from bacterial infection is constituted by the gastric mucosa that secretes several protective factors, among which is the trefoil factor 1 (TFF1), that, as mucin 5AC, binds the bacterium. Even if the protective role of TFF1 is well-documented, the molecular mechanisms that confer a beneficial function to the interaction among TFF1 and H. pylori remain still unclear. Here we analyze the effects of this interaction on H. pylori at morphological and molecular levels by means of microscopic observation, chemiotaxis and motility assays and real-time PCR analysis. Our results show that TFF1 favors aggregation of H. pylori and significantly slows down the motility of the bacterium across the mucus. Such aggregates significantly reduce both flgE and flaB gene transcription compared with bacteria not incubated with TFF1. Finally, our results suggest that the interaction between TFF1 and the bacterium may explain the frequent persistence of H. pylori in the human host without inducing disease.


Asunto(s)
Proteínas Bacterianas/metabolismo , Flagelina/metabolismo , Mucosa Gástrica , Helicobacter pylori/metabolismo , Factor Trefoil-1/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Células HT29 , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA