Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell Commun Signal ; 22(1): 15, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38183060

RESUMEN

BACKGROUND: The dynamic interaction between cancer cells and tumour-associated macrophages (TAMs) in the hypoxic tumour microenvironment (TME) is an active barrier to the effector arm of the antitumour immune response. Cancer-secreted exosomes are emerging mediators of this cancer-stromal cross-talk in the TME; however, the mechanisms underlying this interaction remain unclear. METHODS: Exosomes were isolated with ExoQuick exosome precipitation solution. The polarizing effect of TAMs was evaluated by flow cytometry, western blot analysis, immunofluorescence staining and in vitro phagocytosis assays. Clinical cervical cancer specimens and an in vivo xenograft model were also employed. RESULTS: Our previous study showed that hypoxia increased the expression of ZEB1 in cervical squamous cell carcinoma (CSCC) cells, which resulted in increased infiltration of TAMs. Here, we found that hypoxia-induced ZEB1 expression is closely correlated with CD47-SIRPα axis activity in CSCC, which enables cancer cells to evade phagocytosis by macrophages and promotes tumour progression. ZEB1 was found to directly activate the transcription of the CD47 gene in hypoxic CSCC cells. We further showed that endogenous ZEB1 was characteristically enriched in hypoxic CSCC cell-derived exosomes and transferred into macrophages via these exosomes to promote SIRPα+ TAM polarization. Intriguingly, exosomal ZEB1 retained transcriptional activity and reprogrammed SIRPα+ TAMs via activation of the STAT3 signalling pathway in vitro and in vivo. STAT3 inhibition reduced the polarizing effect induced by exosomal ZEB1. Knockdown of ZEB1 increased the phagocytosis of CSCC cells by macrophages via decreasing CD47 and SIRPα expression. CONCLUSIONS: Our results suggest that hypoxia-induced ZEB1 promotes immune evasion in CSCC by strengthening the CD47-SIRPα axis. ZEB1-targeted therapy in combination with CD47-SIRPα checkpoint immunotherapy may improve the outcomes of CSCC patients in part by disinhibiting innate immunity.


Asunto(s)
Carcinoma de Células Escamosas , Escape del Tumor , Neoplasias del Cuello Uterino , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Femenino , Humanos , Antígeno CD47 , Exosomas , Evasión Inmune , Microambiente Tumoral , Neoplasias del Cuello Uterino/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
5.
J Exp Clin Cancer Res ; 40(1): 87, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33648530

RESUMEN

BACKGROUND: Irradiation has emerged as a valid tool for nasopharyngeal carcinoma (NPC) in situ treatment; however, NPC derived from tissues treated with irradiation is a main cause cancer-related death. The purpose of this study is to uncover the underlying mechanism regarding tumor growth after irradiation and provided potential therapeutic strategy. METHODS: Fibroblasts were extracted from fresh NPC tissue and normal nasopharyngeal mucosa. Immunohistochemistry was conducted to measure the expression of α-SMA and FAP. Cytokines were detected by protein array chip and identified by real-time PCR. CCK-8 assay was used to detect cell proliferation. Radiation-resistant (IRR) 5-8F cell line was established and colony assay was performed to evaluate tumor cell growth after irradiation. Signaling pathways were acquired via gene set enrichment analysis (GSEA). Comet assay and γ-H2AX foci assay were used to measure DNA damage level. Protein expression was detected by western blot assay. In vivo experiment was performed subcutaneously. RESULTS: We found that radiation-resistant NPC tissues were constantly infiltrated with a greater number of cancer-associated fibroblasts (CAFs) compared to radiosensitive NPC tissues. Further research revealed that CAFs induced the formation of radioresistance and promoted NPC cell survival following irradiation via the IL-8/NF-κB pathway to reduce irradiation-induced DNA damage. Treatment with Tranilast, a CAF inhibitor, restricted the survival of CAF-induced NPC cells and attenuated the of radioresistance properties. CONCLUSIONS: Together, these data demonstrate that CAFs can promote the survival of irradiated NPC cells via the NF-κB pathway and induce radioresistance that can be interrupted by Tranilast, suggesting the potential value of Tranilast in sensitizing NPC cells to irradiation.


Asunto(s)
Fibroblastos/metabolismo , FN-kappa B/metabolismo , Carcinoma Nasofaríngeo/genética , Animales , Humanos , Masculino , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo/mortalidad , Carcinoma Nasofaríngeo/patología , Análisis de Supervivencia
7.
Angiogenesis ; 24(3): 549-565, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33484377

RESUMEN

Lymphatic remodelling in the hypoxic tumour microenvironment (TME) is critically involved in the metastasis of cervical squamous cell carcinoma (CSCC); however, its underlying mechanisms remain unclear. Here, we uncovered a novel lymphatic pattern in the hypoxic TME, wherein lymphatic vessels (LVs) are encapsulated by tumour-associated macrophages (TAMs) to form an interconnected network. We describe these aggregates as LVEM (LVs encapsulated by TAMs) considering their advantageous metastatic capacity and active involvement in early lymph node metastasis (LNM). Mechanistic investigations revealed that interleukin-10 (IL-10) derived from hypoxic TAMs adjacent to LVs was a prerequisite for lymphangiogenesis and LVEM formation through its induction of Sp1 upregulation in lymphatic endothelial cells (LECs). Interestingly, Sp1high LECs promoted the transactivation of C-C motif chemokine ligand 1 (CCL1) to facilitate TAM and tumour cell recruitment, thereby forming a positive feedback loop to strengthen the LVEM formation. Knockdown of Sp1 or blockage of CCL1 abrogated LVEM and consequently attenuated LNM. Notably, CSCCnon-LNM is largely devoid of hypoxic TAMs and the resultant LVEM, which might explain its metastatic delay. These findings identify a novel and efficient metastasis-promoting lymphatic pattern in the hypoxic TME, which might provide new targets for anti-metastasis therapy and prognostic assessment.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Adulto , Animales , Hipoxia de la Célula , Femenino , Humanos , Vasos Linfáticos/patología , Ratones , Metástasis de la Neoplasia , Células RAW 264.7 , Células THP-1 , Macrófagos Asociados a Tumores/patología , Neoplasias del Cuello Uterino/patología
8.
Mol Ther ; 29(4): 1512-1528, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33388421

RESUMEN

Cancer-associated lymphatic endothelial cells (LECs) are an active barrier to the effector arm of the anti-tumor immune response; however, it remains unclear how LECs become immunosuppressive in the tumor microenvironment (TME). Exosomal microRNAs (miRNAs) have recently been implicated in intercellular crosstalk within the TME. Here, we report a mechanistic model via which cervical cancer-secreted, exosome-encapsulated microRNA (miR)-1468-5p promotes lymphatic PD-L1 upregulation and lymphangiogenesis to impair T cell immunity. Subsequently, exosomal miR-1468-5p epigenetically activates the JAK2/STAT3 pathway in LECs by directly targeting homeobox containing 1 (HMBOX1) in the SOCS1 promoter, activating an immunosuppressive program that allows cancer cells to escape anti-cancer immunity. Furthermore, clinical data reveal that high serum exosomal miR-1468-5p levels correlate with TME immunosuppressive status and poor prognosis in cervical cancer (CCa) patients. Taken together, our results suggest that cancer-secreted exosomal miR-1468-5p instructs LECs to form an integrated immunosuppressive TME component and may be a prognostic biomarker and therapeutic target for CCa.


Asunto(s)
Células Endoteliales/inmunología , Proteínas de Homeodominio/genética , MicroARNs/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética , Neoplasias del Cuello Uterino/terapia , Antígeno B7-H1/genética , Reprogramación Celular/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Exosomas/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunidad/genética , Inmunidad/inmunología , Terapia de Inmunosupresión/métodos , Janus Quinasa 2/genética , Linfangiogénesis/genética , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Regiones Promotoras Genéticas/genética , Factor de Transcripción STAT3/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Escape del Tumor/genética , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/inmunología
9.
Mol Oncol ; 15(1): 210-227, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33124726

RESUMEN

Lymph node metastasis (LNM), a critical prognostic determinant in cancer patients, is critically influenced by the presence of numerous heterogeneous cancer-associated fibroblasts (CAFs) in the tumor microenvironment. However, the phenotypes and characteristics of the various pro-metastatic CAF subsets in cervical squamous cell carcinoma (CSCC) remain unknown. Here, we describe a CAF subpopulation with elevated periostin expression (periostin+ CAFs), located in the primary tumor sites and metastatic lymph nodes, that positively correlated with LNM and poor survival in CSCC patients. Mechanistically, periostin+ CAFs impaired lymphatic endothelial barriers by activating the integrin-FAK/Src-VE-cadherin signaling pathway in lymphatic endothelial cells and consequently enhanced metastatic dissemination. In contrast, inhibition of the FAK/Src signaling pathway alleviated periostin-induced lymphatic endothelial barrier dysfunction and its related effects. Notably, periostin- CAFs were incapable of impairing endothelial barrier integrity, which may explain the occurrence of CAF-enriched cases without LNM. In conclusion, we identified a specific periostin+ CAF subset that promotes LNM in CSCC, mainly by impairing the lymphatic endothelial barriers, thus providing the basis for potential stromal fibroblast-targeted interventions that block CAF-dependent metastasis.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/patología , Metástasis Linfática/patología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Adulto , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Regulación hacia Abajo , Células Endoteliales/metabolismo , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Integrinas/metabolismo , Persona de Mediana Edad , Análisis de Supervivencia , Familia-src Quinasas/metabolismo
10.
Cell Death Dis ; 10(7): 508, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31263103

RESUMEN

The accumulation of tumour-associated macrophages (TAMs) in the hypoxic tumour microenvironment (TME) is associated with malignant progression in cancer. However, the mechanisms by which the hypoxic TME facilitates TAM infiltration are not fully understood. This study showed that high ZEB1 expression in hypoxic cervical cancer cell islets was positively correlated with CD163+ TAM accumulation. ZEB1 in hypoxic cancer cells promoted the migration of TAMs in vitro and altered the expression of multiple chemokines, especially CCL8. Mechanistically, hypoxia-induced ZEB1 activated the transcription of CCL8, which attracted macrophages via the CCR2-NF-κB pathway. Furthermore, ZEB1 and CCL8 were independent prognostic factors in cervical cancer patients based on The Cancer Genome Atlas (TCGA) data analysis. In conclusion, hypoxia-induced ZEB1 exerts unexpected functions in cancer progression by fostering a prometastatic environment through increased CCL8 secretion and TAM recruitment; thus, ZEB1 may serve as a candidate biomarker of tumour progression and provide a potential target for disrupting hypoxia-mediated TME remodelling.


Asunto(s)
Quimiocina CCL8/metabolismo , Hipoxia/metabolismo , Macrófagos/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Adulto , Western Blotting , Línea Celular Tumoral , Quimiocina CCL8/genética , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Persona de Mediana Edad , FN-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Microambiente Tumoral/genética , Microambiente Tumoral/fisiología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA