Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Theranostics ; 14(2): 662-680, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38169511

RESUMEN

Rationale: Cancer local recurrence increases the mortality of patients, and might be caused by field cancerization, a pre-malignant alteration of normal epithelial cells. It has been suggested that cancer-derived small extracellular vesicles (CDEs) may contribute to field cancerization, but the underlying mechanisms remain poorly understood. In this study, we aim to identify the key regulatory factors within recipient cells under the instigation of CDEs. Methods: In vitro experiments were performed to demonstrate that CDEs promote the expression of CREPT in normal epithelial cells. TMT-based quantitative mass spectrometry was employed to investigate the proteomic differences between normal cells and tumor cells. Loss-of-function approaches by CRISPR-Cas9 system were used to assess the role of CREPT in CDEs-induced field cancerization. RNA-seq was performed to explore the genes regulated by CREPT during field cancerization. Results: CDEs promote field cancerization by inducing the expression of CREPT in non-malignant epithelial cells through activating the ERK signaling pathway. Intriguingly, CDEs failed to induce field cancerization when CREPT was deleted, highlighting the importance of CREPT. Transcriptomic analyses revealed that CDEs elicited inflammatory responses, primarily through activation of the TNF signaling pathway. CREPT, in turn, regulates the transduction of downstream signals of TNF by modulating the expression of TNFR2 and PI3K, thereby promoting inflammation-to-cancer transition. Conclusion: CREPT not only serves as a biomarker for field cancerization, but also emerges as a target for preventing the cancer local recurrence.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Humanos , Línea Celular Tumoral , Proteómica , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/genética , Proteínas de Neoplasias/genética , Vesículas Extracelulares/metabolismo , Neoplasias/genética
2.
Mol Pharm ; 20(8): 4007-4020, 2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37427910

RESUMEN

Distant metastasis of malignant tumors is considered to be the main culprit for the failure of current antitumor treatments. Conventional single treatments often exhibit limited efficacy in inhibiting tumor metastasis. Therefore, there is a growing interest in developing collaborative antitumor strategies based on photothermal therapy (PTT) and free-radical-generated photodynamic therapy (PDT), especially utilizing oxygen-independent nanoplatforms, to address this challenge. Such antitumor strategies can enhance the therapeutic outcomes by ensuring the cytotoxicity of free radicals even in the hypoxic tumor microenvironment, thereby improving the effective suppression of primary tumors. Additionally, these approaches can stimulate the production of tumor-associated antigens and amplify the immunogenic cell death (ICD) effects, potentially feasible for enhancing the therapeutic outcomes of immunotherapy. Herein, we fabricated a functional nanosystem that co-loads IR780 and 2,2'-azobis[2-(2-imidazolin-2-yl)propane]-dihydrochloride (AIPH) to realize PTT-triggered thermodynamic combination therapy via the oxygen-independent pathway for the elimination of primary tumors. Furthermore, the nanocomposites were surface-decorated with a predesigned complex peptide (PLGVRGC-anti-PD-L1 peptide, MMP-sensitive), which facilitated the immunotherapy targeting distant tumors. Through the specific recognition of matrix metalloproteinase (MMP), the sensitive segment on the obtained aNC@IR780A was cleaved. As a result, the freed anti-PD-L1 peptide effectively blocked immune checkpoints, leading to the infiltration and activation of T cells (CTLs). This nanosystem was proven to be effective at inhibiting both primary tumors and distant tumors, providing a promising combination strategy for tumor PTT/TDT/immunotherapy.


Asunto(s)
Nanopartículas , Fototerapia , Línea Celular Tumoral , Inmunoterapia , Oxígeno , Péptidos , Polímeros , Termodinámica , Microambiente Tumoral , Humanos
3.
Cell Signal ; 106: 110592, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36641081

RESUMEN

Natural compounds that interfere with tumor cell growth have potential to be used as therapeutic agents to treat cancers. Lachnochromonin (p71) is a small molecule isolated from Lachnum virgineum. Here, we reported the effect of p71 on human tumor cells, especially on breast cancer MCF-7 cells. We found that p71 significantly suppresses cell growth and induces apoptosis. The luciferase results demonstrated that p71 specifically attenuates the activation of JAK/STAT3 signaling. Biochemical analysis revealed that p71 blocks the phosphorylation of STAT3 tyrosine 705 and serine 727, resulting in down-regulation of c-Myc and Cyclin D1 expression level. Importantly, p71 inhibited cell growth, colony-formation, and migration through affecting STAT3 activity. These results implied that p71 may be used as a therapeutic agent against breast cancer.


Asunto(s)
Apoptosis , Neoplasias de la Mama , Humanos , Femenino , Línea Celular Tumoral , Transducción de Señal , Proliferación Celular , Fosforilación , Neoplasias de la Mama/patología , Factor de Transcripción STAT3/metabolismo
4.
Cancers (Basel) ; 14(19)2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36230720

RESUMEN

Histone deacetylases 1 (HDAC1), an enzyme that functions to remove acetyl molecules from ε-NH3 groups of lysine in histones, eliminates the histone acetylation at the promoter regions of tumor suppressor genes to block their expression during tumorigenesis. However, it remains unclear why HDAC1 fails to impair oncogene expression. Here we report that HDAC1 is unable to occupy at the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with CREPT (cell cycle-related and expression-elevated protein in tumor, also named RPRD1B), an oncoprotein highly expressed in tumors. We observed that CREPT competed with HDAC1 for binding to oncogene (such as CCND1, CLDN1, VEGFA, PPARD and BMP4) promoters but not the tumor suppressor gene (such as p21 and p27) promoters by a chromatin immunoprecipitation (ChIP) qPCR experiment. Using immunoprecipitation experiments, we deciphered that CREPT specifically occupied at the oncogene promoter via TCF4, a transcription factor activated by Wnt signaling. In addition, we performed a real-time quantitative PCR (qRT-PCR) analysis on cells that stably over-expressed CREPT and/or HDAC1, and we propose that HDAC1 inhibits CREPT to activate oncogene expression under Wnt signaling activation. Our findings revealed that HDAC1 functions differentially on tumor suppressors and oncogenes due to its interaction with the oncoprotein CREPT.

5.
FEBS Lett ; 596(20): 2668-2677, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35918178

RESUMEN

Exosomes released by mesenchymal stem cells (MSCs) are thought to function as extensions of the MSCs. However, it remains unclear whether exosomes derived from human umbilical cord MSCs (HUMSCs) possess immunoregulatory functions in rheumatoid arthritis. We report that when mice with collagen-induced arthritis were injected with exosomes derived from HUMSC (HUMSC-Exo), their paws became less swollen, and they had lower serum pro-inflammatory cytokine and anti-collagen IgG levels, and decreased synovial hyperplasia. The HUMSC-Exo appeared to restore the balance between Th17 and Treg cells, and this effect was accompanied by reduced IL-17 and enhanced TGF-ß and IL-10 levels. These findings suggest that HUMSC-Exo function as important regulator of the balance between Th1/Th17 and Treg cells during immune and inflammatory responses.


Asunto(s)
Artritis Experimental , Exosomas , Células Madre Mesenquimatosas , Animales , Humanos , Ratones , Artritis Experimental/terapia , Citocinas , Inmunoglobulina G , Interleucina-10/genética , Interleucina-17 , Linfocitos T Reguladores , Factor de Crecimiento Transformador beta , Cordón Umbilical , Células Th17
6.
Stem Cells Int ; 2022: 1617229, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35694239

RESUMEN

Mesenchymal stem cells (MSCs) have been documented to be effective for the therapy of inflammation-related diseases but raised concerns on possible tumorigenic effects. Since most of the tumors are induced or promoted by chronic inflammation, one could expect that MSCs might be beneficial for the cancer therapy because of their potent roles on inhibiting inflammation. This study is aimed at performing a safety evaluation and evaluating the role of human umbilical cord mesenchymal stem cells (HUC-MSCs) on tumorigenesis. We found that HUC-MSCs cultured within 20 generations had no significant changes in proliferation, cell cycle, cellular senescence, apoptosis, and expression of mesenchymal stem cell markers. HUC-MSCs were unable to form any tumor in immunodeficiency or normal mice with or without inflammatory stimulation. Intriguingly, we observed that HUC-MSCs inhibited tumorigenesis in B16-derived or AOM/DSS-induced colon cancer models. We reasoned that the effect of HUC-MSCs on tumorigenesis might be through regulating the inflammatory response. Indeed, HUC-MSCs dramatically ameliorated the disease symptoms and pathological changes of DSS-induced colitis mice. We deciphered the mechanism that HUC-MSCs inhibited tumorigenesis through reducing the proportion of macrophages, which were decreased in the mice suffered from AOM/DSS-induced colon cancer. Correspondingly, the expression levels of TNF-α and IL-6, which were secreted by macrophages, were significantly decreased in the plasma of colon cancer and colitis mice after injection of HUC-MSCs. This study revealed the role of inhibiting macrophages and shed light on the therapeutic application of HUC-MSCs in inflammation-induced tumorigenesis.

7.
FEBS Lett ; 596(4): 427-436, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34939667

RESUMEN

T helper cells, especially Th1 and Th17 cells, were reported to play a pivotal role in the pathogenesis of inflammatory bowel disease (IBD). However, the underlying factors regulating T cell functions in IBD progression remain to be fully elucidated. Here, we revealed that IL-17RD/Sef exacerbates DSS-induced colitis by regulating the balance of T cell subsets and their secretion of associated cytokines. We also observed that IL-17RD/Sef promotes colitis-associated tumorigenesis and negatively correlates with survival in both mouse and colorectal cancer patients. Our results suggested that IL-17RD/Sef functions as a regulator of T cell subsets to promote the inflammatory responses in the pathogenesis of IBD and colitis-associated colon cancer.


Asunto(s)
Carcinogénesis/inmunología , Colitis/inmunología , Proteínas de la Membrana/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Colitis/inducido químicamente , Colitis/genética , Colitis/mortalidad , Colon/inmunología , Colon/patología , Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inflamación , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Recuento de Linfocitos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Análisis de Supervivencia , Células TH1/patología , Células Th17/patología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
8.
Br J Cancer ; 124(8): 1437-1448, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33531691

RESUMEN

BACKGROUND: Signal transducer and activator of transcription 3 (STAT3) has been shown to upregulate gene transcription during tumorigenesis. However, how STAT3 initiates transcription remains to be exploited. This study is to reveal the role of CREPT (cell cycle-related and elevated-expression protein in tumours, or RPRD1B) in promoting STAT3 transcriptional activity. METHODS: BALB/c nude mice, CREPT overexpression or deletion cells were employed for the assay of tumour formation, chromatin immunoprecipitation, assay for transposase-accessible chromatin using sequencing. RESULTS: We demonstrate that CREPT, a recently identified oncoprotein, enhances STAT3 transcriptional activity to promote tumorigenesis. CREPT expression is positively correlated with activation of STAT3 signalling in tumours. Deletion of CREPT led to a decrease, but overexpression of CREPT resulted in an increase, in STAT3-initiated tumour cell proliferation, colony formation and tumour growth. Mechanistically, CREPT interacts with phosphorylated STAT3 (p-STAT3) and facilitates p-STAT3 to recruit p300 to occupy at the promoters of STAT3-targeted genes. Therefore, CREPT and STAT3 coordinately facilitate p300-mediated acetylation of histone 3 (H3K18ac and H3K27ac), further augmenting RNA polymerase II recruitment. Accordingly, depletion of p300 abolished CREPT-enhanced STAT3 transcriptional activity. CONCLUSIONS: We propose that CREPT is a co-activator of STAT3 for recruiting p300. Our study provides an alternative strategy for the therapy of cancers related to STAT3.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica/patología , Proteína p300 Asociada a E1A/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Femenino , Células HEK293 , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células 3T3 NIH , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Fosforilación , Transcripción Genética
9.
Nat Commun ; 12(1): 270, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33431892

RESUMEN

Intestinal stem cells (ISCs) residing in the crypts are critical for the continual self-renewal and rapid recovery of the intestinal epithelium. The regulatory mechanism of ISCs is not fully understood. Here we report that CREPT, a recently identified tumor-promoting protein, is required for the maintenance of murine ISCs. CREPT is preferably expressed in the crypts but not in the villi. Deletion of CREPT in the intestinal epithelium of mice (Vil-CREPTKO) results in lower body weight and slow migration of epithelial cells in the intestine. Vil-CREPTKO intestine fails to regenerate after X-ray irradiation and dextran sulfate sodium (DSS) treatment. Accordingly, the deletion of CREPT decreases the expression of genes related to the proliferation and differentiation of ISCs and reduces Lgr5+ cell numbers at homeostasis. We identify that CREPT deficiency downregulates Wnt signaling by impairing ß-catenin accumulation in the nucleus of the crypt cells during regeneration. Our study provides a previously undefined regulator of ISCs.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Intestinos/fisiología , Proteínas de Neoplasias/metabolismo , Regeneración/fisiología , Células Madre/metabolismo , Animales , Recuento de Células , Proteínas de Ciclo Celular/deficiencia , Diferenciación Celular , Proliferación Celular , Epitelio/metabolismo , Eliminación de Gen , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Proteínas de Neoplasias/deficiencia , Organoides/metabolismo , Células Madre/citología , Vía de Señalización Wnt , Rayos X , beta Catenina/metabolismo
10.
Cell Mol Immunol ; 18(8): 1883-1895, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32963355

RESUMEN

Rheumatoid arthritis (RA) is exacerbated by TNF-alpha signaling. However, it remains unclear whether TNF-α-activated TNFR1 and TNFR2 are regulated by extracellular factors. Here, we showed that soluble glycosylated interleukin-17 receptor D (sIL-17RD), which was produced by proteolytic cleavage, enhanced TNF-α-induced RA. We revealed that IL-17RD shedding was induced by the proteolytic enzyme TACE and enhanced by TNF-α expression in macrophages. Intriguingly, sIL-17RD was elevated in the sera of arthritic mice and rats. Recombinant sIL-17RD significantly enhanced the TNF-α-induced proinflammatory response by promoting TNF-α-TNFR-sIL-17RD complex formation and receptor clustering, leading to the accelerated development of collagen-induced arthritis. Our observations revealed that ectodomain shedding of IL-17RD occurred in RA to boost the TNF-α-induced inflammatory response. Targeting sIL-17RD may provide a new strategy for the therapy of RA.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Receptores de Interleucina-17 , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Artritis Reumatoide/metabolismo , Análisis por Conglomerados , Ratones , Ratas , Receptores de Interleucina-17/sangre , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Cell Biochem ; 121(2): 1541-1551, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31535402

RESUMEN

Mesenchymal stem cells (MSCs) have been proved to exert anti-inflammatory effects and regulate immune reactions. Traditional Chinese medicine (TCM), qi-fang-bi-min-tang, is effective for some patients with allergic diseases. However, it remains unclear whether MSCs combined with TCM could benefit the treatment of allergic rhinitis (AR). In this study, we reported an additional effect of TCM (qi-fang-bi-min-tang) on the therapy of AR under MSCs treatment. Intriguingly, we observed that TCM-treated MSCs significantly inhibited the symptoms of AR and reduced the pathological changes of nasal mucosa in ovalbumin (OVA)-induced rats. The expression levels of interferon Î³ (IFN-γ), interleukin-17 (IL-17), and IL-4 were significantly decreased in the plasma of AR rats after injection of TCM-treated MSCs. TCM-treated MSCs reduced the levels of histamine secreted by mast cells and immunoglobulin E (IgE) secreted by plasma cells. In addition, we found that MSCs combined with TCM had a better therapeutic effect than TCM alone on AR in an OVA-induced mouse model. After OVA induction, MSCs combined with TCM significantly reduced the ratio of T helper type 1 (Th1), Th2, and Th17, but increased the proportion of Treg in the spleen of mice. Consistently, the expression levels of IFN-γ, IL-4, and IL-17 were significantly decreased, but transforming growth factor-ß1 was significantly increased in the plasma of AR mice after treated with TCM and MSCs. Our results from both rats and mice indicated that the effects of TCM combined with MSCs on the AR might be through regulating the secretion of Th1, Th2, and Th17 cytokines. This study suggested that TCM (qi-fang-bi-min-tang)-treated MSCs could be used in the clinical therapy of AR.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Rinitis Alérgica/terapia , Aloinjertos , Animales , Citocinas/inmunología , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Sprague-Dawley , Rinitis Alérgica/inmunología , Rinitis Alérgica/patología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología
12.
Theranostics ; 9(5): 1369-1384, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30867837

RESUMEN

Nuclear factor-kappa B (NF-κB) activation is critical for innate immune responses. However, cellular-intrinsic regulation of NF-κB activity during inflammatory diseases remains incompletely understood. Ubiquitin-like protein 4A (UBL4A, GdX) is a small adaptor protein involved in protein folding, biogenesis and transcription. Yet, whether GdX has a role during innate immune response is largely unknown. Methods: To investigate the involvement of GdX in innate immunity, we challenged GdX-deficient mice with lipopolysaccharides (LPS). To investigate the underlying mechanism, we performed RNA sequencing, real-time PCR, ELISA, luciferase reporter assay, immunoprecipitation and immunoblot analyses, flow cytometry, and structure analyses. To investigate whether GdX functions in inflammatory bowel disease, we generated dendritic cell (DC), macrophage (Mφ), epithelial-cell specific GdX-deficient mice and induced colitis with dextran sulfate sodium. Results: GdX enhances DC and Mφ-mediated innate immune defenses by positively regulating NF-κB signaling. GdX-deficient mice were resistant to LPS-induced endotoxin shock and DSS-induced colitis. DC- or Mφ- specific GdX-deficient mice displayed alleviated mucosal inflammation. The production of pro-inflammatory cytokines by GdX-deficient DCs and Mφ was reduced. Mechanistically, we found that tyrosine-protein phosphatase non-receptor type 2 (PTPN2, TC45) and protein phosphatase 2A (PP2A) form a complex with RelA (p65) to mediate its dephosphorylation whereas GdX interrupts the TC45/PP2A/p65 complex formation and restrict p65 dephosphorylation by trapping TC45. Conclusion: Our study provides a mechanism by which NF-κB signaling is positively regulated by an adaptor protein GdX in DC or Mφ to maintain the innate immune response. Targeting GdX could be a strategy to reduce over-activated immune response in inflammatory diseases.


Asunto(s)
Colitis/patología , Células Dendríticas/inmunología , Inmunidad Innata , Macrófagos/inmunología , FN-kappa B/metabolismo , Transducción de Señal , Ubiquitinas/metabolismo , Animales , Colitis/inducido químicamente , Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Ubiquitinas/deficiencia
13.
Cell Death Dis ; 9(12): 1172, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518842

RESUMEN

Gastric cancer, like most of other cancers, has an uncontrolled cell cycle regulated by cyclins and cyclin-dependent kinases (CDKs). In this study, we reported that gastric cancer cells showed an accelerated G2/M transition promoted by CREPT/RPRD1B and Aurora kinase B (Aurora B). We found that CREPT/RPRD1B and Aurora B were coordinately expressed during the cell cycle in gastric cancer cells. Deletion of CREPT/RPRD1B disturbed the cell progression and extended the length of cell cycle, leading to a significant accumulation of mitotic cells. Mechanistically, we revealed that CREPT/RPRD1B interacted with Aurora B to regulate the expression of Cyclin B1 in gastric cancer cells. Interestingly, Aurora B phosphorylates S145 in a well-conserved motif of CREPT/RPRD1B. We proposed that phosphorylation of CREPT/RPRD1B by Aurora B is required for promoting the transcription of Cyclin B1, which is critical for the regulation of gastric tumorigenesis. Our study provides a mechanism by which gastric tumor cells maintain their high proliferation rate via coordination of Aurora B and CREPT/RPRD1B on the expression of Cyclin B1. Targeting the interaction of Aurora B and CREPT/RPRD1B might be a strategy for anti-gastric cancer therapy in the future.


Asunto(s)
Aurora Quinasa B/genética , Proteínas de Ciclo Celular/genética , Ciclina B1/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética , Animales , Aurora Quinasa B/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Ciclina B1/metabolismo , Humanos , Metástasis Linfática , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Estadificación de Neoplasias , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Carcinog ; 57(10): 1408-1420, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29938829

RESUMEN

CREPT (Cell-cycle-related and expression-elevated protein in tumor)/RPRD1B, a novel protein that enhances the transcription of Cyclin D1 to promote cell proliferation during tumorigenesis, was demonstrated highly expressed in most of tumors. However, it remains unclear how CREPT is regulated in colorectal cancers. In this study, we report that miR-383 negatively regulates CREPT expression. We observed that CREPT was up-regulated but the expression of miR-383 was down regulated in both colon cancer cell lines and colon tumor tissues. Intriguingly, we found that enforced expression of miR-383 inhibited the expression of CREPT at both the mRNA and protein level. Using a luciferase reporter, we showed that miR-383 targeted the 3'-UTR of CREPT mRNA directly. Consistently we observed that over expression of miR-383 shortened the half-life of CREPT mRNA in varieties of colorectal cancer cells. Furthermore, restoration of miR-383 inhibited cell growth and colony formation of colon cancer cells accompanied by inhibition of expression of CREPT and related downstream genes. Finally, we demonstrated that stable over expression of miR-383 in colon cancer cells decreased the growth of the tumors. Our results revealed that the abundant expression of CREPT in colorectal cancers is attributed to the decreased level of miR-383. This study shed a new light on the potential therapeutic therapy strategy for colorectal cancers using introduced miRNA.


Asunto(s)
Proteínas de Ciclo Celular/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MicroARNs/genética , Proteínas de Neoplasias/genética , Regiones no Traducidas 3'/genética , Anciano , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Femenino , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Estabilidad del ARN/genética
15.
J Biol Chem ; 293(20): 7618-7628, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29618509

RESUMEN

We previously demonstrated that p15RS, a newly discovered tumor suppressor, inhibits Wnt/ß-catenin signaling by interrupting the formation of ß-catenin·TCF4 complex. However, it remains unclear how p15RS helps exert such an inhibitory effect on Wnt signaling based on its molecular structure. In this study, we reported that dimerization of p15RS is required for its inhibition on the transcription regulation of Wnt-targeted genes. We found that p15RS forms a dimer through a highly conserved leucine zipper-like motif in the coiled-coil terminus domain. In particular, residues Leu-248 and Leu-255 were identified as being responsible for p15RS dimerization, as mutation of these two leucines into prolines disrupted the homodimer formation of p15RS and weakened its suppression of Wnt signaling. Functional studies further confirmed that mutations of p15RS at these residues results in diminishment of its inhibition on cell proliferation and tumor formation. We therefore concluded that dimerization of p15RS governed by the leucine zipper-like motif is critical for its inhibition of Wnt/ß-catenin signaling and tumorigenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Leucina Zippers , Melanoma/prevención & control , Proteínas Represoras/química , Animales , Apoptosis , Proliferación Celular , Femenino , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Multimerización de Proteína , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción 4/antagonistas & inhibidores , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Células Tumorales Cultivadas , Vía de Señalización Wnt , beta Catenina/antagonistas & inhibidores , beta Catenina/metabolismo
16.
Oncogene ; 37(26): 3485-3500, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29563608

RESUMEN

Using whole genome sequencing, we identified gene amplification of CREPT in colorectal cancer (CRC). In this study, we aim to clarify its clinical significance, biological effects, and mechanism in CRC. CREPT was upregulated in CRC cell lines and in 47.37% (72/152) of primary CRC tumors. Amplification of CREPT was detected in 48.28% (56/116) of primary CRC tumors, which was positively correlated with its overexpression (P < 0.001). Multivariate analysis showed that CRC patients with CREPT protein overexpression were significantly associated with poor disease-free survival (P < 0.05). CREPT significantly accelerated CRC cell proliferation and metastasis both in vitro and in vivo. RNA-sequencing (seq) analysis uncovered that the tumor-promoting effect by CREPT was attributed to enhancing Wnt/ß-catenin signaling. Using co-immunoprecipitation coupled with mass spectroscopy, we identified p300 protein was a novel CREPT interacting partner. CREPT greatly increased the interaction between p300 and ß-catenin, thus promoting p300-mediated ß-catenin acetylation and stabilization. Moreover, CREPT cooperated with p300, leading to elevated active histone acetylation markers H3K27ac and H4Ac and decreased repressive histone marker H3K9me3 at the promoters of Wnt downstream targets. In summary, CREPT plays a pivotal oncogenic role in colorectal carcinogenesis through promoting Wnt/ß-catenin pathway via cooperating with p300. CREPT may serve as a prognostic biomarker of patients with CRC.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales/patología , Proteína p300 Asociada a E1A/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Acetilación , Animales , Células CACO-2 , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/mortalidad , Supervivencia sin Enfermedad , Amplificación de Genes/genética , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Trasplante Heterólogo , Regulación hacia Arriba , Secuenciación Completa del Genoma
17.
Cell Biochem Funct ; 35(7): 420-425, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28940415

RESUMEN

Mesenchymal stromal cells (MSCs) have been extensively investigated as a potential antiinflammatory treatment in many inflammatory-related diseases; however, it remains unclear whether MSCs could be used to treat acute allergic rhinitis. A rat model of allergic rhinitis was treated with MSCs. The effect of MSCs on the inflammation of allergic rhinitis was evaluated by sneezing, nose rubbing, the pathology of the nasal mucosa, and the expression of interleukin 4, tumour necrosis factor alpha, and immunoglobulin E in the serum of rats. Also, the population of MSCs isolated from umbilical cords of humans was evaluated to determine if they could inhibit the symptoms and inflammation of acute allergic rhinitis in a rat model. We observed that this population of cells inhibited sneezing, nose rubbing, and changes in the pathology of the nasal mucosa. Intriguingly, we observed that MSCs reduced the expression of interleukin 4, tumour necrosis factor alpha, and immunoglobulin E in the serum. Furthermore, MSCs reduced the expression of histamine and the recruitment of macrophages in the nasal mucosa of allergic rhinitis rats. We reasoned that the effect of MSCs on allergic rhinitis might be through its regulation of the secretion of related cytokines from macrophages during the process of acute allergic rhinitis. This work suggested that MSCs from the umbilical cords of humans could be used as a positive clinical therapy for the human disease.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Rinitis Alérgica/terapia , Enfermedad Aguda , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Histamina/sangre , Humanos , Inmunoglobulina E/sangre , Interleucina-4/sangre , Macrófagos/citología , Macrófagos/inmunología , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Mucosa Nasal/citología , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Ratas , Ratas Sprague-Dawley , Rinitis Alérgica/metabolismo , Rinitis Alérgica/patología , Trasplante Heterólogo , Factor de Necrosis Tumoral alfa/sangre , Cordón Umbilical/citología
18.
FEBS Lett ; 589(15): 1890-6, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26026268

RESUMEN

STAT3 is associated with embryo development and survival as well as proliferation and metastasis of tumor cells. In a previous study, we demonstrated that STAT3-Interacting Protein As a Repressor (SIPAR) enhances the dephosphorylation of STAT3 and negatively regulates its activity. However, it remains unclear how SIPAR inhibits phosphorylation of STAT3. Here we demonstrate that SIPAR directly interacts with T cell protein tyrosine phosphatase TC45 and enhances its association with STAT3. This interaction triggers an accelerated dephosphorylation process for STAT3. Furthermore, SIPAR inhibits the transcriptional activity of STAT3 in wild-type MEF cells but not in TC45 null MEF cells. These results suggest that SIPAR terminates the activation of STAT3 through a dephosphorylation process that is dependent upon interaction with TC45 in the nucleus.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Núcleo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones
19.
J Biol Chem ; 290(15): 9701-13, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25697359

RESUMEN

We previously reported that p15RS (p15INK4b-related sequence), a regulation of nuclear pre-mRNA domain containing protein, inhibited Wnt signaling by interrupting the formation of the ß-catenin·TCF4 complex. However, how p15RS functions as an intrinsic repressor to repress transcription remains unclear. In this study, we show that p15RS, through a specific interaction with HDAC2 (histone deacetylase 2), a deacetylase that regulates gene transcription, maintains histone H3 in a deacetylated state in the promoter region of Wnt-targeted genes where ß-catenin·TCF4 is bound. We observed that histone deacetylase inhibitors impair the ability of p15RS in inhibiting Wnt/ß-catenin signaling. Depletion of HDAC2 markedly disabled p15RS inhibition of Wnt/ß-catenin-mediated transcription. Interestingly, overexpression of p15RS decreases the level of acetylated histone H3 in the c-MYC promoter. Finally, we demonstrate that p15RS significantly enhances the association of HDAC2 and TCF4 and enhances the occupancy of HDAC2 to DNA, resulting in the deacetylation of histone H3 and the failure of ß-catenin interaction. We propose that p15RS acts as an intrinsic transcriptional repressor for Wnt/ß-catenin-mediated gene transcription at least partially through recruiting HDAC2 to occupy the promoter and maintaining deacetylated histone H3.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas Represoras/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Acetilación , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Western Blotting , Proteínas de Ciclo Celular/genética , Proliferación Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Expresión Génica , Células HEK293 , Histona Desacetilasa 2/genética , Histonas/metabolismo , Humanos , Células MCF-7 , Microscopía Confocal , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Interferencia de ARN , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción 4 , Factores de Transcripción/genética , beta Catenina/genética
20.
J Biol Chem ; 290(2): 861-71, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25378394

RESUMEN

TNF receptor 2 (TNFR2) exerts diverse roles in the pathogenesis of inflammatory and autoimmune diseases. Here, we report that TNFR2 but not TNFR1 forms a heteromer with interleukin-17 receptor D (IL-17RD), also named Sef, to activate NF-κB signaling. TNFR2 associates with IL-17RD, leading to mutual receptor aggregation and TRAF2 recruitment, which further activate the downstream cascade of NF-κB signaling. Depletion of IL-17RD impaired TNFR2-mediated activation of NF-κB signaling. Importantly, IL-17RD was markedly increased in renal tubular epithelial cells in nephritis rats, and a strong interaction of TNFR2 and IL-17RD was observed in the renal epithelia. The IL-17RD·TNFR2 complex in activation of NF-κB may explain the role of TNFR2 in inflammatory diseases including nephritis.


Asunto(s)
Inflamación/metabolismo , FN-kappa B/metabolismo , Nefritis/metabolismo , Receptores de Interleucina-17/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Inflamación/etiología , Inflamación/patología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , FN-kappa B/genética , Nefritis/etiología , Nefritis/patología , Multimerización de Proteína , Ratas , Receptores de Interleucina-17/química , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/química , Transducción de Señal/genética , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA