Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Acta Pharmacol Sin ; 45(2): 378-390, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37798352

RESUMEN

Reinforced cellular responses to endoplasmic reticulum (ER) stress are caused by a variety of pathological conditions including cancers. Human rhomboid family-1 protein (RHBDF1), a multiple transmembrane protein located mainly on the ER, has been shown to promote cancer development, while the binding immunoglobulin protein (BiP) is a key regulator of cellular unfolded protein response (UPR) for the maintenance of ER protein homeostasis. In this study, we investigated the role of RHBDF1 in maintaining ER protein homeostasis in breast cancer cells. We showed that deleting or silencing RHBDF1 in breast cancer cell lines MCF-7 and MDA-MB-231 caused marked aggregation of unfolded proteins in proximity to the ER. We demonstrated that RHBDF1 directly interacted with BiP, and this interaction had a stabilizing effect on the BiP protein. Based on the primary structural motifs of RHBDF1 involved in BiP binding, we found a pentapeptide (PE5) targeted BiP and inhibited BiP ATPase activity. SPR assay revealed a binding affinity of PE5 toward BiP (Kd = 57.7 µM). PE5 (50, 100, 200 µM) dose-dependently promoted ER protein aggregation and ER stress-mediated cell apoptosis in MCF-7 and MDA-MB-231 cells. In mouse 4T1 breast cancer xenograft model, injection of PE5 (10 mg/kg, s.c., every 2 days for 2 weeks) significantly inhibited the tumor growth with markedly increased ER stress and apoptosis-related proteins in tumor tissues. Our results suggest that the ability of RHBDF1 to maintain BiP protein stability is critical to ER protein homeostasis in breast cancer cells, and that the pentapeptide PE5 may serve as a scaffold for the development of a new class of anti-BiP inhibitors.


Asunto(s)
Neoplasias de la Mama , Proteínas Portadoras , Humanos , Animales , Ratones , Femenino , Proteínas Portadoras/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Estrés del Retículo Endoplásmico , Apoptosis , Respuesta de Proteína Desplegada , Proteínas Reguladoras de la Apoptosis/metabolismo , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo
2.
Pharmacol Res ; 198: 106995, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37979663

RESUMEN

Melanoma is a dangerous form of skin cancer, making it important to investigate new mechanisms and approaches to enhance the effectiveness of treatment. Here, we establish a positive correlation between the human rhomboid family-1 (RHBDF1) protein and melanoma malignancy. We demonstrate that the melanoma RHBDF1 decrease dramatically inhibits tumor growth and the development of lung metastases, which may be related to the impaired glycolysis. We show that RHBDF1 function is essential to the maintenance of high levels of glycolytic enzymes, especially glucose-6-phosphate isomerase (GPI). Additionally, we discover that the E3 ubiquitin ligase tripartite motif-containing 32 (TRIM32) mediates the K27/K63-linked ubiquitination of GPI and the ensuing lysosomal degradation process. We prove that the multi-transmembrane domain of RHBDF1 is in competition with GPI, preventing the latter from interacting with NCL1-HT2A-LIN41 (NHL) domain of TRIM32. We also note that the mouse RHBDF1's R747 and Y799 are crucial for competitive binding and GPI protection. Artificially silencing the Rhbdf1 gene in a mouse melanoma model results in declined lactic acid levels, elevated cytotoxic lymphocyte infiltration, and improved tumor responsiveness to immunotherapy. These results provide credence to the hypothesis that RHBDF1 plays a significant role in melanoma regulation and suggest that blocking RHBDF1 may be an efficient technique for reestablishing the tumor immune microenvironment (TIME) in melanoma and halting its progression.


Asunto(s)
Glucosa-6-Fosfato Isomerasa , Melanoma , Humanos , Animales , Ratones , Glucosa-6-Fosfato Isomerasa/genética , Glucosa-6-Fosfato Isomerasa/metabolismo , Proteínas de la Membrana/metabolismo , Ubiquitinación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Melanoma/genética , Melanoma/terapia , Inmunoterapia , Microambiente Tumoral , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Factores de Transcripción/metabolismo
3.
Microbiome ; 10(1): 149, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36114582

RESUMEN

BACKGROUND: The pathogenesis of inflammatory bowel diseases (IBD) is multifactorial, and diagnostic and treatment strategies for IBD remain to be developed. RhoB regulates multiple cell functions; however, its role in colitis is unexplored. RESULTS: Here, we found RhoB was dramatically increased in colon tissues of ulcerative colitis (UC) patients and mice with DSS-induced colitis. Compared with wild type mice, RhoB+/- and RhoB-/- mice developed milder DSS-induced colitis and increased goblet cell numbers and IEC proliferation. Decreased RhoB promoted goblet cell differentiation and epithelial regeneration through inhibiting Wnt signaling pathway and activating p38 MAPK signaling pathway. Moreover, increased SCFA-producing bacteria and SCFA concentrations were detected in intestinal microbiome of both RhoB+/- and RhoB-/- mice and upregulated SCFA receptor expression was also observed. CONCLUSIONS: Taken together, a higher level of RhoB is associated with UC, which also contributes to UC development through modulating cell signaling and altering intestinal bacterial composition and metabolites. These observations suggest that RhoB has potential as a biomarker and a treatment target for UC. Video Abstract.


Asunto(s)
Colitis Ulcerosa , Colitis , Microbioma Gastrointestinal , Proteína de Unión al GTP rhoB/metabolismo , Animales , Biomarcadores , Colitis/inducido químicamente , Colitis/patología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Sulfato de Dextran , Humanos , Ratones , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Cell Rep ; 40(9): 111308, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36044853

RESUMEN

Commensal intestinal bacteria play key roles in regulating host immune tolerance; however, bacterial strains and related metabolites directly involved in this regulation are largely unknown. Here, using a mouse model of dextran sulfate sodium (DSS)-induced colitis combined with different antibiotic treatment, Enterobacter ludwigii, abundant in microbiota of mice treated with metronidazole, is screened out to have prophylactic and therapeutic effects on DSS-induced colitis with or without the presence of complex intestinal bacteria. E. ludwigii is found to induce CD103+DC and regulatory T (Treg)-mediated immune tolerance for colitis remission using in vitro and in vivo experiments. Moreover, choline, one metabolite of E. ludwigii, is identified to increase dendritic cells' (DCs) immune tolerance to promote Treg differentiation. E. ludwigii is found to induce DCs' immune tolerance ability for Treg differentiation through choline and α7nAChR-mediated retinoic acid (RA) and transforming growth factor beta (TGF-ß) upregulation, resulting in protecting mice against DSS-induced colitis. This study suggests potential therapeutic approaches for inflammatory bowel diseases (IBDs).


Asunto(s)
Colina , Colitis , Animales , Colina/metabolismo , Células Dendríticas/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Enterobacter , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores
5.
Cancer Cell Int ; 21(1): 590, 2021 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-34736454

RESUMEN

BACKGROUND: The rhomboids are a family of multi-transmembrane proteins, many of which have been implicated in facilitating tumor progression. Little is yet known, however, about rhomboid-associated biomarkers in cancers. An analysis of such biomarkers could yield important insights into the role of the rhomboids in cancer pathology. METHODS: In this study, we carried out the univariate Cox regression analysis and compared gene expression patterns of several rhomboid genes in 30 types of cancers by using The Cancer Genome Atlas (TCGA) database and the methods delineated in Gene Expression Profiling Interactive Analysis (GEPIA). We then used datasets GSE47032, GSE126964, GSE68417 and 75 paired pathological specimens to verify the influences of the rhomboid genes in cancer progression. Moreover, we carried out Weighted Gene Correlation Network Analysis (WGCNA) to investigate gene-related functions and we exploited potential correlations between rhomboid genes expression and immune cell infiltration in cancer tissues. Furthermore, we constructed gene-knockdown cancer cell lines to investigate rhomboid gene functions. RESULTS: We find that kidney renal clear cell carcinoma (KIRC) disease progression is affected by fluctuations in the expression of a number of the rhomboid family of genes and, more specifically, high levels of RHBDF2 gene expression are a good indicator of poor prognosis of the disease, as patients with high RHBDF2 expression levels exhibit less favorable survival rates compared to those with low RHBDF2 levels. Silencing of the RHBDF2 gene in KIRC cell lines leads to significantly diminished cell proliferation and migration; this is in good agreement with the identification of an enhanced presence of a number of cell growth and migration promoting signaling molecules in KIRC tumors. We found that, although high level of RHBDF2 correlated with increased infiltration of lymphocytes in cancer tissues, artificially overexpressed RHBDF2 led to an inhibition of the activity of the infiltrated immune cells through sustaining PD-L1 protein level. Furthermore, we show that RHBDF2 related cell migration and PD-L1 regulation were potentially mediated by EGFR signaling pathway. CONCLUSIONS: RHBDF2 gene functions are correlated to facilitated renal clear cell carcinoma progression and may serve as a critical prognostic biomarker for the disease.

6.
ASN Neuro ; 13: 17590914211038441, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34596444

RESUMEN

Destabilization of blood vessels by the activities of vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs) following intracerebral hemorrhage (ICH) has been considered the main causes of aggravated secondary brain injury. Here, we show that tumor necrosis factor superfamily-15 (TNFSF15; also known as vascular endothelial growth inhibitor), an inhibitor of VEGF-induced vascular hyper-permeability, when overexpressed in transgenic mice, exhibits a neuroprotective function post-ICH. In this study, we set-up a collagenase-induced ICH model with TNFSF15-transgenic mice and their transgene-negative littermates. We observed less lesion volume and neural function perturbations, together with less severe secondary injuries in the acute phase that are associated with brain edema and inflammation, including vascular permeability, oxidative stress, microglia/macrophage activation and neutrophil infiltration, and neuron degeneration, in the TNFSF15 group compared with the littermate group. Additionally, we show that there is an inhibition of VEGF-induced elevation of MMP-9 in the perihematomal blood vessels of the TNFSF15 mice following ICH, concomitant with enhanced pericyte coverage of the perihematomal blood vessels. These findings are consistent with the view that TNFSF15 may have a potential as a therapeutic agent for the treatment of secondary injuries in the early phase of ICH.


Asunto(s)
Edema Encefálico , Lesiones Encefálicas , Animales , Edema Encefálico/etiología , Permeabilidad Capilar , Hemorragia Cerebral/complicaciones , Modelos Animales de Enfermedad , Ratones , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral , Factor A de Crecimiento Endotelial Vascular
8.
Nat Commun ; 12(1): 2587, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972537

RESUMEN

Host cells use several anti-bacterial pathways to defend against pathogens. Here, using a uropathogenic Escherichia coli (UPEC) infection model, we demonstrate that bacterial infection upregulates RhoB, which subsequently promotes intracellular bacteria clearance by inducing LC3 lipidation and autophagosome formation. RhoB binds with Beclin 1 through its residues at 118 to 140 and the Beclin 1 CCD domain, with RhoB Arg133 being the key binding residue. Binding of RhoB to Beclin 1 enhances the Hsp90-Beclin 1 interaction, preventing Beclin 1 degradation. RhoB also directly interacts with Hsp90, maintaining RhoB levels. UPEC infections increase RhoB, Beclin 1 and LC3 levels in bladder epithelium in vivo, whereas Beclin 1 and LC3 levels as well as UPEC clearance are substantially reduced in RhoB+/- and RhoB-/- mice upon infection. We conclude that when stimulated by UPEC infections, host cells promote UPEC clearance through the RhoB-Beclin 1-HSP90 complex, indicating RhoB may be a useful target when developing UPEC treatment strategies.


Asunto(s)
Autofagosomas/metabolismo , Beclina-1/metabolismo , Infecciones por Escherichia coli/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Infecciones Urinarias/metabolismo , Escherichia coli Uropatógena/crecimiento & desarrollo , Proteína de Unión al GTP rhoB/metabolismo , Animales , Autofagosomas/genética , Autofagosomas/ultraestructura , Beclina-1/genética , Línea Celular , Epitelio/metabolismo , Epitelio/microbiología , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Femenino , Técnicas de Silenciamiento del Gen , Proteínas HSP90 de Choque Térmico/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Estabilidad Proteica , ARN Interferente Pequeño , Proteínas Recombinantes , Vejiga Urinaria/metabolismo , Vejiga Urinaria/microbiología , Infecciones Urinarias/genética , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/patogenicidad , Proteína de Unión al GTP rhoB/genética
9.
J Mater Chem B ; 9(1): 80-84, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33313613

RESUMEN

Specific and expeditious identification and enrichment of target proteins in living cells is often a challenging task. The hexahistidine (6His) tag is frequently used to label artificially engineered proteins produced in prokaryotic or eukaryotic cells. Utilizing the interaction between 6His-tag and nitrilotriacetic acid (NTA) mediated by divalent metal ions (Ni2+, Cu2+, Zn2+ or Co2+), we designed and synthesized a series of Nap-G/Biotin/ANA-FFpYGK-NTA probes that, assisted by alkaline phosphatase (ALP), self-assemble into nanofibers. The probe consists of an NTA group that specifically binds to 6His-tag, an FFpY group that promotes self-assembly facilitated by ALP, and a hydrophobic (Nap-G/ANA/Biotin) capping group for various applications. We demonstrate that the ANA-FFpYGK-NTA(Ni2+) nanofibers are fit for real-time tracking of His-tagged protein in living cells, and the Biotin-FFpYGK-NTA(Ni2+) nanofibers are for isolating His-tagged proteins and other proteins that they interact with.


Asunto(s)
Quelantes/metabolismo , Citoplasma/metabolismo , Histidina/metabolismo , Nanofibras , Ácido Nitrilotriacético/metabolismo , Oligopéptidos/metabolismo , Quelantes/análisis , Citoplasma/química , Colorantes Fluorescentes/análisis , Colorantes Fluorescentes/metabolismo , Histidina/análisis , Humanos , Células MCF-7 , Nanofibras/análisis , Ácido Nitrilotriacético/análisis , Oligopéptidos/análisis
10.
Chem Commun (Camb) ; 57(6): 797-800, 2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33355564

RESUMEN

Caspase-3/8 are key members of the cysteine-aspartyl protease family with pivotal roles in apoptosis. We have designed and synthesized self-assembling probes, Nap-GFFpYDEVD-AFC and Nap-GFFpYIETD-AFC, with fluorescence 'turn-on' properties for real-time monitoring of Caspase-3/8 activity in living cells.


Asunto(s)
Caspasa 3/análisis , Caspasa 8/análisis , Pruebas de Enzimas/métodos , Colorantes Fluorescentes/química , Nanofibras/química , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Colorantes Fluorescentes/síntesis química , Células HeLa , Humanos , Microscopía Confocal , Estructura Molecular , Factores de Tiempo
11.
Chem Commun (Camb) ; 56(97): 15321-15324, 2020 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-33205785

RESUMEN

We developed a new strategy to overcome the MDR of etoposide using self-assembling nanofibers. Compared with the original etoposide, the inhibitory activity of Nap-GFFpYK-etoposide1/2 against murine Lewis lung cancer or breast cancer cells was increased 10 times, and 20 times on these cells with artificially overexpressed MDR1. Our method to synthesize and separate etoposide isomers provides a new strategy for the modification of this drug.


Asunto(s)
Antineoplásicos/síntesis química , Portadores de Fármacos/química , Etopósido/química , Nanofibras/química , Péptidos/síntesis química , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Composición de Medicamentos , Liberación de Fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Imagen Óptica , Péptidos/farmacología , Técnicas de Síntesis en Fase Sólida , Espectrometría de Fluorescencia
12.
FASEB J ; 34(6): 7927-7940, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32314833

RESUMEN

Uropathogenic Escherichia coli (UPEC), a leading cause of urinary tract infections, is associated with prostate and bladder cancers. Cytotoxic necrotizing factor 1 (CNF1) is a key UPEC toxin; however, its role in bladder cancer is unknown. In the present study, we found CNF1 induced bladder cancer cells to secrete vascular endothelial growth factor (VEGF) through activating Ras homolog family member C (RhoC), leading to subsequent angiogenesis in the bladder cancer microenvironment. We then investigated that CNF1-mediated RhoC activation modulated the stabilization of hypoxia-inducible factor 1α (HIF1α) to upregulate the VEGF. We demonstrated in vitro that active RhoC increased heat shock factor 1 (HSF1) phosphorylation, which induced the heat shock protein 90α (HSP90α) expression, leading to stabilization of HIF1α. Active RhoC elevated HSP90α, HIF1α, VEGF expression, and angiogenesis in the human bladder cancer xenografts. In addition, HSP90α, HIF1α, and VEGF expression were also found positively correlated with the human bladder cancer development. These results provide a potential mechanism through which UPEC contributes to bladder cancer progression, and may provide potential therapeutic targets for bladder cancer.


Asunto(s)
Toxinas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Vejiga Urinaria/metabolismo , Proteína rhoC de Unión a GTP/metabolismo , Animales , Línea Celular , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/microbiología , Neutrófilos/metabolismo , Microambiente Tumoral/fisiología , Vejiga Urinaria/microbiología , Neoplasias de la Vejiga Urinaria/microbiología , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología
13.
Mucosal Immunol ; 13(1): 22-33, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31719643

RESUMEN

Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs), inducing acute pyelonephritis and may result in permanent renal scarring and failure. Alpha-hemolysin (HlyA), a key UPEC toxin, causes serious tissue damage; however, the mechanism through which HlyA induces kidney injury remains unclear. In the present study, granulocyte-macrophage colony-stimulating factor (GM-CSF) secreted by renal epithelial cells was upregulated by HlyA in vitro and in vivo, which induced M1 macrophage accumulation in kidney, and ADAM10 was found involved in HlyA-induced GM-CSF. Macrophage elimination or GM-CSF neutralization protected against acute kidney injury in mice, and increased GM-CSF was detected in urine of patients infected by hlyA-positive UPEC. In addition, HlyA was found to promote UPEC invasion into renal epithelial cells by interacting with Nectin-2 in vitro. However, HlyA did not affect bacterial titers during acute kidney infections, and HlyA-induced invasion did not contribute to GM-CSF upregulation in vitro, which indicate that HlyA-induced GM-CSF is independent of bacteria invasion. The role of GM-CSF in HlyA-mediated kidney injury may lead to novel strategies to treat acute pyelonephritis.


Asunto(s)
Lesión Renal Aguda/metabolismo , Células Epiteliales/metabolismo , Infecciones por Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiología , Proteínas Hemolisinas/metabolismo , Riñón/patología , Pielonefritis/metabolismo , Lesión Renal Aguda/microbiología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/patología , Infecciones por Escherichia coli/microbiología , Femenino , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Nectinas/metabolismo , Pielonefritis/microbiología , Transducción de Señal , Infecciones Urinarias
14.
EBioMedicine ; 50: 23-33, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31757778

RESUMEN

BACKGROUND: Uropathogenic Escherichia coli (UPEC) is the leading cause of urinary tract infections (UTIs), and fimbrial tip adhesins, play important roles in UPEC colonization. Few fimbrial tip adhesins and their receptors on host cells, which have the potential to be the therapeutic targets, have been identified. METHODS: the UPEC wild-type strain CFT073, ΔyadC and the complemented strain were used to perform assays in vitro and in vivo. The effects of D-xylose targeting YadC on UPEC colonization were evaluated. A YadC receptor was identified by far-western blotting, LC-MS/MS and co-immunoprecipitation. The effects of compounds targeting the receptor on UPEC colonization were tested. FINDINGS: YadC was investigated for its mediation of UPEC adhesion and invasion to bladder epithelial cells in vitro; and its promotion of UPEC colonization in bladder in vivo. D-xylose, targeting YadC, showed prophylactic and therapeutic effects on UPEC colonization. Annexin A2 (ANXA2) was identified as a YadC receptor, involved in UPEC infection. ANXA2 inhibitors attenuated UPEC infections. The yadC gene was widely present in UPEC clinical isolates and phylogenetic analysis of yadC was performed. INTERPRETATION: YadC and its receptor ANXA2 play important roles in UPEC colonization in bladder, leading to novel treatment strategies targeting YadC or ANXA2 for acute UTIs. FUND: This study was supported by grants from the National Natural Science Foundation of China (NSFC) Programs (31670071 and 31970133), the National Key Technologies R&D Program, Intergovernmental international innovation cooperation (2018YFE0102000), Tianjin Science and Technology Commissioner Project (18JCZDJC36000), the Science & Technology Development Fund of Tianjin Education Commission for Higher Education (2017ZD12). The Science Foundation of Tianjin Medical University (2016KY2M08).


Asunto(s)
Anexina A2/metabolismo , Cistitis/metabolismo , Cistitis/microbiología , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas Fimbrias/antagonistas & inhibidores , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Biomarcadores , Línea Celular , Modelos Animales de Enfermedad , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Femenino , Proteínas Fimbrias/química , Proteínas Fimbrias/genética , Humanos , Inmunohistoquímica , Ratones , Membrana Mucosa/metabolismo , Membrana Mucosa/microbiología , Filogenia , Escherichia coli Uropatógena/clasificación , Escherichia coli Uropatógena/genética
15.
FASEB J ; 33(9): 10505-10514, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31242765

RESUMEN

Hemangioblastoma (HB) is an abnormal intracranial buildup of blood vessels that exhibit a great potential for hemorrhage. Surgical options are limited, and few medications are available for treatment. We show here by immunohistochemical analysis that HB lesions display highly increased levels of VEGF expression and macrophage/microglia infiltration compared with those in normal brain tissues. In the meantime, TNF superfamily 15 (TNFSF15) (also known as vascular endothelial growth inhibitor), an antiangiogenic cytokine, is highly expressed in normal brain blood vessels but diminished in HB lesions. We set up a brain hemangioma model by using mouse bEnd.3 cells of a T antigen-transformed endothelial cell line that produce a large amount of VEGF. When implanted in mouse brains, these cells form lesions that closely resemble the pathologic characteristics of HB. Retroviral infection of bEnd.3 cells with TNFSF15 leads to inhibition of VEGF production and retardation of hemangioma formation. Similar results are obtained when wild-type bEnd.3 cells are implanted in the brains of transgenic mice overexpressing TNFSF15. Additionally, TNFSF15 treatment results in enhanced pericyte coverage of the blood vessels in the lesions together with reduced inflammatory cell infiltration and decreased hemorrhage. These findings indicate that the ability of TNFSF15 to counterbalance the abnormally highly angiogenic and inflammatory potential of the microenvironment of HB is of therapeutic value for the treatment of this disease.-Yang, G.-L., Han, Z., Xiong, J., Wang, S., Wei, H., Qin, T.-T., Xiao, H., Liu, Y., Xu, L.-X., Qi, J.-W., Zhang, Z.-S., Jiang, R., Zhang, J., Li, L.-Y. Inhibition of intracranial hemangioma growth and hemorrhage by TNFSF15.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Células Endoteliales/trasplante , Hemangioma/prevención & control , Hemorragias Intracraneales/prevención & control , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Animales , Apoptosis , Proliferación Celular , Células Endoteliales/citología , Hemangioma/metabolismo , Hemangioma/patología , Humanos , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pronóstico , Células Tumorales Cultivadas , Microambiente Tumoral , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/administración & dosificación
16.
EBioMedicine ; 36: 229-240, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30279141

RESUMEN

BACKGROUND: Epidermal growth factor receptor (EGFR) signalling is critical in epithelial cancer development. Human rhomboid family-1 (RHBDF1) facilitates the secretion of TGFα, an EGFR ligand, in breast cancer; however, the underlying mechanism remains unclear. We evaluated the role for RHBDF1 in clathrin-coated vesicle (CCV)-dependent pro-TGFα membrane trafficking in breast cancer cells upon stimulation by G-protein coupled receptor (GPCR) agonists. METHODS: RHBDF1 was silenced in various breast cancer cells using shRNA. TGFα levels, subcellular localization, and secretion were evaluated using ELISA, immunofluorescent staining, and coimmunoprecipitation. Phosphorylation and expression of relevant proteins were measured by western blotting. RHBDF1-dependent cell viability and invasion were measured. FINDINGS: RHBDF1 mediates GPCR agonist-induced EGFR phosphorylation by promoting TGFα secretion in various types of breast cancer cells. RHBDF1 not only mediates ADAM17-dependent shedding of TGFα, but is essential in membrane trafficking of pro-TGFα. RHBDF1 silencing results in blocking of clathrin uncoating from CCV, a crucial step for the plasma membrane release of pro-TGFα. Interaction of RHBDF1 with auxilin-2, a CCV protein, determines the recruitment of HSC70 to CCV to facilitate clathrin uncoating. RHBDF1 function is required for the proliferation and mobility of breast cancer cells upon stimulation by Sphingosine 1 Phosphate (S1P), a GPCR agonist. We demonstrate a significant correlation between RHBDF1 overexpression and EGFR activation in breast cancer tissues. INTERPRETATION: RHBDF1 is an indispensable component of the protein trafficking machinery involved in GPCR-mediated EGFR transactivation, and is an attractive therapeutic target for cancer. FUND: National Natural Science Foundation of China (81,672,740 to ZSZ, 81,272,356 and 81,330,029 to LYL).


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Vesículas Cubiertas por Clatrina/metabolismo , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Proteína ADAM17/metabolismo , Auxilinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Femenino , Proteínas del Choque Térmico HSC70/metabolismo , Humanos , Ligandos , Modelos Biológicos , Unión Proteica , Transporte de Proteínas , ARN Interferente Pequeño/genética
17.
Front Immunol ; 9: 1987, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30233583

RESUMEN

Urinary tract infections (UTIs) caused by uropathogenic Escherichia coli (UPEC) induce cystitis, pyelonephritis, and can cause kidney scarring and failure if inflammation is not under control. The detailed effects of cytotoxic necrotizing factor 1 (CNF1), the key UPEC toxin, on the pathogenicity of UPEC remain unclear. CD36 is an important scavenger receptor, responsible for pathogen and apoptotic cell clearance, and plays an essential role in host immune defense and homeostasis. Regulation of CD36 by bacterial toxins has not been reported. In this study, using a pyelonephritis mouse model, CNF1 was observed to contribute to increasing neutrophils and bacterial titers in infected bladder and kidney tissues, resulting in severe inflammation and tissue damage. CD36 expression in macrophages was found to be decreased by CNF1 in vitro and in vivo. We demonstrated that CNF1 attenuated CD36 transcription by decreasing expressions of its upstream transcription factors LXRß and C/EBPα and their recruitment to the CD36 promotor. In addition, Cdc42 was found to be involved in CNF1-mediated downregulation of LXRß. Our study investigated the pathogenesis of cnf1-carrying UPEC, which affected host innate immune defenses and homeostasis through regulation of CD36 in macrophages during acute UTIs.


Asunto(s)
Toxinas Bacterianas/metabolismo , Antígenos CD36/genética , Infecciones por Escherichia coli/inmunología , Proteínas de Escherichia coli/metabolismo , Inflamación/inmunología , Macrófagos Peritoneales/inmunología , Pielonefritis/inmunología , Infecciones Urinarias/inmunología , Escherichia coli Uropatógena/fisiología , Enfermedad Aguda , Animales , Toxinas Bacterianas/genética , Antígenos CD36/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Proteínas de Escherichia coli/genética , Femenino , Homeostasis , Humanos , Inmunidad Innata , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
18.
FASEB J ; 31(5): 2001-2012, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28183800

RESUMEN

Vascular hyperpermeability is critical in ischemic diseases, including stroke and myocardial infarction, as well as in inflammation and cancer. It is well known that the VEGF-VEGFR2 signaling pathways are pivotal in promoting vascular permeability; however, counterbalancing mechanisms that restrict vascular permeability to maintain the integrity of blood vessels are not yet fully understood. We report that TNF superfamily member 15 (TNFSF15), a cytokine largely produced by vascular endothelial cells and a specific inhibitor of the proliferation of these same cells, can inhibit VEGF-induced vascular permeability in vitro and in vivo, and that death receptor 3 (DR3), a cell surface receptor of TNFSF15, mediates TNFSF15-induced dephosphorylation of VEGFR2. Src homology region 2 domain-containing phosphatase-1 (SHP-1) becomes associated with DR3 upon TNFSF15 interaction with the latter. In addition, a protein complex consisting of VEGFR2, DR3, and SHP-1 is formed in response to the effects of TNFSF15 and VEGF on endothelial cells. It is plausible that this protein complex provides a structural basis for the molecular mechanism in which TNFSF15 induces the inhibition of VEGF-stimulated vascular hyperpermeability.-Yang, G.-L., Zhao, Z., Qin, T.-T., Wang, D., Chen, L., Xiang, R., Xi, Z., Jiang, R., Zhang, Z.-S., Zhang, J., Li. L.-Y. TNFSF15 inhibits VEGF-stimulated vascular hyperpermeability by inducing VEGFR2 dephosphorylation.


Asunto(s)
Células Endoteliales/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Humanos , Permeabilidad , Fosforilación , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa
19.
Int J Biochem Cell Biol ; 55: 109-18, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25161149

RESUMEN

Tumor necrosis factor superfamily 15 (TNFSF15) suppresses angiogenesis by specifically inducing apoptosis in proliferating endothelial cells. Death receptor 3 (DR3), a member of the TNF receptor superfamily (TNFRSF25), has been identified as a receptor for TNFSF15 to activate T cells. It is unclear, however, whether DR3 mediates TNFSF15 activity on endothelial cells. Here we show that siRNA-mediated knockdown of DR3 in an in vivo Matrigel angiogenesis assay, or in adult bovine aortic endothelial (ABAE) cell cultures, leads to resistance of endothelial cells to TNFSF15-induced apoptosis. Interestingly, DR3-depleted cells also exhibited markedly diminished responsiveness to TNFα cytotoxicity, even though DR3 is not a receptor for TNFα. Treatment of the cells with either TNFSF15 siRNA or a TNFSF15-neutralizing antibody, 4-3H, also results in a significant inhibition of TNFα-induced apoptosis. Mechanistically, DR3 siRNA treatment gives rise to an increase of ERK1/2 MAPK activity, and up-regulation of the anti-apoptotic proteins c-FLIP and Bcl-2, thus strengthening apoptosis-resisting potential in the cells. These findings indicate that DR3 mediates TNFSF15-induced endothelial cell apoptosis, and that up-regulation of TNFSF15 expression stimulated by TNFα is partly but significantly responsible for TNFα-induced apoptosis in endothelial cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasas/metabolismo , Bovinos , Línea Celular , Doxorrubicina/farmacología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Expresión Génica , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
20.
Cancer Res ; 74(10): 2719-30, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24648344

RESUMEN

Intermittent oxygen deficiency in cancers promotes prolonged inflammation, continuous angiogenesis, and increased drug resistance. Hypoxia-inducible factor-1 (HIF1) has a pivotal role in the regulation of cellular responses to oxygen deficiency. The α-subunit of HIF1 (HIF1α) is degraded in normoxia but stabilized in hypoxia. However, the molecular mechanism that controls oxygen-independent degradation of HIF1α has remained elusive. Human rhomboid family-1 (RHBDF1) is a member of a large family of nonprotease rhomboids whose function is basically unknown. We report here that RHBDF1 expression in breast cancer is highly elevated and is strongly correlated with escalated disease progression, metastasis, poor prognosis, and poor response to chemotherapy. We show that RHBDF1 interaction with the receptor of activated protein-C kinase-1 (RACK1) in breast cancer cells prevents RACK1-assisted, oxygen-independent HIF1α degradation. In addition, we show that the HIF1α-stabilizing activity of RHBDF1 diminishes when the phosphorylation of a tyrosine residue on the RHBDF1 molecule is inhibited. These findings are consistent with the view that RHBDF1 is a critical component of a molecular switch that regulates HIF1α stability in cancer cells in hypoxia and that RHBDF1 is of potential value as a new target for cancer treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Receptores ErbB/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Progresión de la Enfermedad , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Femenino , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteínas de Unión al GTP/metabolismo , Silenciador del Gen , Células HEK293 , Humanos , Células MCF-7 , Proteínas de la Membrana , Persona de Mediana Edad , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Oxígeno/metabolismo , Fosforilación , Receptores de Cinasa C Activada , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA