Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Int J Mol Sci ; 24(17)2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37686288

RESUMEN

The tumor microenvironment comprises multiple cell types, like cancer cells, endothelial cells, fibroblasts, and immune cells. In recent years, there have been massive research efforts focusing not only on cancer cells, but also on other cell types of the tumor microenvironment, thereby aiming to expand and determine novel treatment options. Fibroblasts represent a heterogenous cell family consisting of numerous subtypes, which can alter immune cell fractions, facilitate or inhibit tumor growth, build pre-metastatic niches, or stabilize vessels. These effects can be achieved through cell-cell interactions, which form the extracellular matrix, or via the secretion of cytokines or chemokines. The pro- or antitumorigenic fibroblast phenotypes show variability not only among different cancer entities, but also among intraindividual sites, including primary tumors or metastatic lesions. Commonly prescribed for arterial hypertension, the inhibitors of the renin-angiotensin system have recently been described as having an inhibitory effect on fibroblasts. This inhibition leads to modified immune cell fractions and increased tissue stiffness, thereby contributing to overcoming therapy resistance and ultimately inhibiting tumor growth. However, it is important to note that the inhibition of fibroblasts can also have the opposite effect, potentially resulting in increased tumor growth. We aim to summarize the latest state of research regarding fibroblast heterogeneity and its intricate impact on the tumor microenvironment and extracellular matrix. Specifically, we focus on highlighting recent advancements in the comprehension of intraindividual heterogeneity and therapy options within this context.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinogénesis , Neoplasias , Fibroblastos Asociados al Cáncer/clasificación , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/fisiología , Humanos , Microambiente Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Antihipertensivos/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología
2.
Pathol Int ; 72(3): 161-175, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35020975

RESUMEN

Cancer-associated fibroblasts (CAFs), a compartment of the tumor microenvironment, were previously thought to be a uniform cell population that promotes cancer progression. However, recent studies have shown that CAFs are heterogeneous and that there are at least two types of CAFs, that is, cancer-promoting and -restraining CAFs. We previously identified Meflin as a candidate marker of cancer-restraining CAFs (rCAFs) in pancreatic ductal adenocarcinoma (PDAC). The precise nature of rCAFs, however, has remained elusive owing to a lack of understanding of their comprehensive gene signatures. Here, we screened genes whose expression correlated with Meflin in single-cell transcriptomic analyses of human cancers. Among the identified genes, we identified matrix remodeling-associated protein 8 (MXRA8), which encodes a type I transmembrane protein with unknown molecular function. Analysis of MXRA8 expression in human PDAC samples showed that MXRA8 was differentially co-expressed with other CAF markers. Moreover, in patients with PDAC or syngeneic tumors developed in MXRA8-knockout mice, MXRA8 expression did not affect the roles of CAFs in cancer progression, and the biological importance of MXRA8+ CAFs is still unclear. Overall, we identified MXRA8 as a new CAF marker; further studies are needed to determine the relevance of this marker.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Inmunoglobulinas/análisis , Proteínas de la Membrana/análisis , Neoplasias Pancreáticas/diagnóstico , Animales , Biomarcadores/análisis , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/patología , Modelos Animales de Enfermedad , Inmunoglobulinas/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados/genética , Neoplasias Pancreáticas/patología
3.
BJU Int ; 129(1): 80-92, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34107167

RESUMEN

OBJECTIVES: To investigate the role of cancer-associated fibroblasts (CAFs) in clear cell renal cell carcinoma (ccRCC) with respect to tumour aggressiveness, metastasis development, and resistance to anti-angiogenic therapy (vascular endothelial growth factor receptor-tyrosine kinase inhibitors [VEGFR-TKI]). PATIENTS AND METHODS: Our study involved tissue samples from three distinct and independent cohorts of patients with ccRCC. The presence of CAFs and tumour lymphangiogenesis was investigated, respectively, by transcriptional signatures and then correlated with tumour development and prognosis. The effect of these CAFs on tumour cell migration and VEGFR-TKI resistance was analysed on co-cultures of ccRCC cells with CAFs. RESULTS: Results from our cohorts and from in silico investigations showed that VEGFR-TKI significantly increase the number of CAFs in tumours. In the same populations of patients with ccRCC, the proportion of intra-tumoral CAFs correlated to shorter disease-free and overall survival. The presence of CAFs was also correlated with lymphangiogenesis and lymph node metastasis. CAFs increased the migration and decreased the VEGFR-TKI-dependent cytotoxic effect of tumour cells. CONCLUSIONS: Our results show that VEGFR-TKI promote the development of CAFs, and CAFs favour tumour aggressiveness, metastatic dissemination, and resistance to treatment in ccRCC. CAFs could represent a new therapeutic target to fight resistance to treatment of ccRCC. Targeting CAF and immunotherapies combination are emerging as efficient treatments in many types of solid tumours. Our results highlight their relevance in ccRCC.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Carcinoma de Células Renales/patología , Resistencia a Antineoplásicos , Neoplasias Renales/patología , Neovascularización Patológica/patología , Actinas/genética , Adulto , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/fisiología , Capilares/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/secundario , Carcinoma de Células Renales/cirugía , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular , Supervivencia sin Enfermedad , Endopeptidasas/genética , Femenino , Humanos , Neoplasias Renales/genética , Neoplasias Renales/terapia , Linfangiogénesis , Metástasis Linfática , Masculino , Proteínas de la Membrana/genética , Ratones , Persona de Mediana Edad , Terapia Neoadyuvante , Neovascularización Patológica/tratamiento farmacológico , Nefrectomía , Estudios Retrospectivos , Sunitinib/metabolismo , Sunitinib/uso terapéutico , Tasa de Supervivencia , Transcriptoma
4.
Int J Oncol ; 60(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34913066

RESUMEN

The cancer­stromal interaction has been demonstrated to promote tumor progression, and cancer-associated fibroblasts (CAFs), which are the main components of stromal cells, have attracted attention as novel treatment targets. Chitinase 3-like 1 (CHI3L1) is a chitinase-like protein, which affects cell proliferation and angiogenesis. However, the mechanisms through which cells secrete CHI3L1 and through which CHI3L1 mediates tumor progression in the cancer microenvironment are still unclear. Accordingly, the present study assessed the secretion of CHI3L1 in the microenvironment of colorectal cancer and evaluated how CHI3L1 affects tumor angiogenesis. CAFs and normal fibroblasts (NFs) established from colorectal cancer tissue, and human colon cancer cell lines were evaluated using immunostaining, cytokine antibody array, RNA interference, reverse transcription-quantitative PCR (RT-qPCR), ELISA, western blotting and angiogenesis assays. The expression and secretion of CHI3L1 in CAFs were stronger than those in NFs and colorectal cancer cell lines. In addition, interleukin-13 receptor α2 (IL-13Rα2), a receptor for CHI3L1, was not expressed in colorectal cancer cell lines, but was expressed in fibroblasts, particularly CAFs. Furthermore, the expression and secretion of IL-8 in CAFs was stronger than that in NFs and cancer cell lines, and recombinant CHI3L1 addition increased IL-8 expression in CAFs, whereas knockdown of CHI3L1 suppressed IL-8 expression. Furthermore, IL-13Rα2 knockdown suppressed the enhancement of IL-8 expression induced by CHI3L1 treatment in CAFs. For vascular endothelial growth factor-A (VEGFA), similar results to IL-8 were observed in an ELISA for comparison of secretion between CAFs and NFs and for changes in secretion after CHI3L1 treatment in CAFs; however, no significant differences were observed for changes in expression after CHI3L1 treatment or IL-13Rα2 knockdown in CAFs assessed using RT-qPCR assays. Angiogenesis assays revealed that tube formation in vascular endothelial cells was suppressed by conditioned medium from CAFs with the addition of human CHI3L1 neutralizing antibodies compared with control IgG, and also suppressed by conditioned medium from CAFs transfected with CHI3L1, IL-8 or VEGFA small interfering RNA compared with negative control small interfering RNA. Overall, the present findings indicated that CHI3L1 secreted from CAFs acted on CAFs to increase the secretion of IL-8, thereby affecting tumor angiogenesis in colorectal cancer.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Fibroblastos Asociados al Cáncer/citología , Proteína 1 Similar a Quitinasa-3/biosíntesis , Neoplasias Colorrectales/sangre , Interleucina-8/biosíntesis , Anciano , Inductores de la Angiogénesis/efectos adversos , Western Blotting/métodos , Western Blotting/estadística & datos numéricos , Fibroblastos Asociados al Cáncer/fisiología , Línea Celular/citología , Línea Celular/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Proteína 1 Similar a Quitinasa-3/efectos adversos , Ensayo de Inmunoadsorción Enzimática/métodos , Ensayo de Inmunoadsorción Enzimática/estadística & datos numéricos , Humanos , Japón , Masculino
5.
Gastroenterology ; 162(3): 890-906, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34883119

RESUMEN

BACKGROUND & AIMS: Cancer-associated fibroblasts (CAFs) play an important role in colorectal cancer (CRC) progression and predict poor prognosis in CRC patients. However, the cellular origins of CAFs remain unknown, making it challenging to therapeutically target these cells. Here, we aimed to identify the origins and contribution of colorectal CAFs associated with poor prognosis. METHODS: To elucidate CAF origins, we used a colitis-associated CRC mouse model in 5 different fate-mapping mouse lines with 5-bromodeoxyuridine dosing. RNA sequencing of fluorescence-activated cell sorting-purified CRC CAFs was performed to identify a potential therapeutic target in CAFs. To examine the prognostic significance of the stromal target, CRC patient RNA sequencing data and tissue microarray were used. CRC organoids were injected into the colons of knockout mice to assess the mechanism by which the stromal gene contributes to colorectal tumorigenesis. RESULTS: Our lineage-tracing studies revealed that in CRC, many ACTA2+ CAFs emerge through proliferation from intestinal pericryptal leptin receptor (Lepr)+ cells. These Lepr-lineage CAFs, in turn, express melanoma cell adhesion molecule (MCAM), a CRC stroma-specific marker that we identified with the use of RNA sequencing. High MCAM expression induced by transforming growth factor ß was inversely associated with patient survival in human CRC. In mice, stromal Mcam knockout attenuated orthotopically injected colorectal tumoroid growth and improved survival through decreased tumor-associated macrophage recruitment. Mechanistically, fibroblast MCAM interacted with interleukin-1 receptor 1 to augment nuclear factor κB-IL34/CCL8 signaling that promotes macrophage chemotaxis. CONCLUSIONS: In colorectal carcinogenesis, pericryptal Lepr-lineage cells proliferate to generate MCAM+ CAFs that shape the tumor-promoting immune microenvironment. Preventing the expansion/differentiation of Lepr-lineage CAFs or inhibiting MCAM activity could be effective therapeutic approaches for CRC.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Fibroblastos Asociados al Cáncer/fisiología , Carcinogénesis/patología , Linaje de la Célula , Neoplasias Colorrectales/patología , Células Madre Mesenquimatosas/fisiología , Actinas/genética , Actinas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígeno CD146/genética , Antígeno CD146/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Diferenciación Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Intestinal/patología , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Organoides/patología , Organoides/fisiología , Pronóstico , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Análisis de Secuencia de ARN , Tasa de Supervivencia , Microambiente Tumoral
6.
Cell Death Dis ; 12(6): 586, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099638

RESUMEN

Hyaluronan (HA) cross-linking is a conformational state of HA, a covalent complex between HA and heavy chains (HCs) from inter-α-trypsin inhibitor (I-α-I) mediated by tumor necrosis factor-induced protein 6 (TSG6). Cross-linked HA has been identified as a protective factor in physiological and inflammatory conditions. However, the state of HA cross-linking in tumor microenvironment has not been fully elucidated. As a major constituent of the extracellular matrix (ECM), HA is mainly synthesized by cancer-associated fibroblasts (CAFs). Our study aimed to clarify the role of HA cross-linking in breast cancer malignancy. Compared to normal mammary gland tissues, cross-linked HA levels were significantly decreased in breast cancer and associated with tumor malignancy. When NFbs were activated into CAFs, the levels of cross-linked HA and TSG6 were both suppressed. Through upregulating TSG6, CAFs restored the high level of cross-linked HA and significantly inhibited breast cancer malignancy, whereas NFbs promoted the malignancy when the cross-linked HA level was reduced. Furthermore, the inhibitory role of HA cross-linking in tumor malignancy was directly verified using the synthesized HA-HC complex. Collectively, our study found that the deficiency of cross-linked HA induced breast cancer malignancy in a CAF-dependent manner, suggesting that recovering HA cross-linking may be a potential therapeutic strategy.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/fisiología , Ácido Hialurónico/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Moléculas de Adhesión Celular/metabolismo , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Humanos , Metástasis Linfática , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Microambiente Tumoral
7.
Life Sci Alliance ; 4(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33758075

RESUMEN

Citrate is important for lipid synthesis and epigenetic regulation in addition to ATP production. We have previously reported that cancer cells import extracellular citrate via the pmCiC transporter to support their metabolism. Here, we show for the first time that citrate is supplied to cancer by cancer-associated stroma (CAS) and also that citrate synthesis and release is one of the latter's major metabolic tasks. Citrate release from CAS is controlled by cancer cells through cross-cellular communication. The availability of citrate from CAS regulated the cytokine profile, metabolism and features of cellular invasion. Moreover, citrate released by CAS is involved in inducing cancer progression especially enhancing invasiveness and organ colonisation. In line with the in vitro observations, we show that depriving cancer cells of citrate using gluconate, a specific inhibitor of pmCiC, significantly reduced the growth and metastatic spread of human pancreatic cancer cells in vivo and muted stromal activation and angiogenesis. We conclude that citrate is supplied to tumour cells by CAS and citrate uptake plays a significant role in cancer metastatic progression.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Ácido Cítrico/metabolismo , Neoplasias Pancreáticas/metabolismo , Fibroblastos Asociados al Cáncer/fisiología , Línea Celular Tumoral , Epigénesis Genética , Humanos , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Neoplasias Pancreáticas/patología , Células del Estroma/metabolismo , Microambiente Tumoral/fisiología , Neoplasias Pancreáticas
8.
Mol Oncol ; 15(5): 1507-1527, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33682233

RESUMEN

Cancer-associated fibroblasts (CAFs) regulate cancer progression through the modulation of extracellular matrix (ECM) and cancer cell adhesion. While undergoing a series of phenotypic changes, CAFs control cancer-stroma interactions through integrin receptor signaling. Here, we isolated CAFs from patients with non-small-cell lung cancer (NSCLC) and examined their gene expression profiles. We identified collagen type XI α1 (COL11A1), integrin α11 (ITGA11), and the ITGA11 major ligand collagen type I α1 (COL1A1) among the 390 genes that were significantly enriched in NSCLC-associated CAFs. Increased ITGA11 expression in cancer stroma was correlated with a poor clinical outcome in patients with NSCLC. Increased expression of fibronectin and collagen type I induced ITGA11 expression in CAFs. The cellular migration of CAFs toward collagen type I and fibronectin was promoted via ERK1/2 signaling, independently of the fibronectin receptor integrin α5ß1. Additionally, ERK1/2 signaling induced ITGA11 and COL11A1 expression in cancer stroma. We, therefore, propose that targeting ITGA11 and COL11A1 expressing CAFs to block cancer-stroma interactions may serve as a novel, promising anti-tumor strategy.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Carcinoma de Pulmón de Células no Pequeñas/patología , Cadenas alfa de Integrinas/genética , Neoplasias Pulmonares/patología , Células A549 , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/genética , Estudios de Casos y Controles , Movimiento Celular/genética , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Cadenas alfa de Integrinas/metabolismo , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad , Unión Proteica , Regulación hacia Arriba/genética
9.
Cancer Med ; 10(7): 2509-2522, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33704935

RESUMEN

Prostate cancer-associated fibroblasts (prostate CAFs) are essential components of the tumor microenvironment and can promote tumor progression through their immunosuppressive functions. MPSSS, a novel polysaccharide purified from Lentinus edodes, has been reported to have anti-tumor activity. MPSSS could also inhibit the immunosuppressive function of prostate CAFs, which has been demonstrated through that the secretome of MPSSS-treated prostate CAFs could inhibit the proliferation of T cells. However, how the secretome of MPSSS-treated prostate CAFs influence prostate cancer progression is still unclear. Interestingly, we found that the low molecular weight (3-100kD) secretome of prostate CAFs (lmwCAFS) could promote the growth of PC-3 cells, while that of MPSSS-treated prostate CAFs (MT-lmwCAFS) could inhibit their growth. We carried out comparative secretomic analysis of lmwCAFS and MT-lmwCAFS to identify functional molecules that inhibit the growth of PC-3 cells, and proteomic analysis of lmwCAFS-treated PC-3 cells and MT-lmwCAFS-treated PC-3 cells to investigate the underlying molecular mechanism. These analyses suggest that TGF-ß3 from MT-lmwCAFS may inhibit the growth of PC-3 cells. The validated experiments revealed that TGF-ß3 from MT-lmwCAFS activated p21 expression in PC-3 cells by regulating the FoxO pathway thereby inducing G0/G1 cell cycle arrest of PC-3 cells. Overall, our data demonstrated that MPSSS reversed the ability of prostate CAFs to suppress the cell viability of PC-3 cells, which might provide a potential therapeutic strategy to prevent prostate cancer progression.


Asunto(s)
Fibroblastos Asociados al Cáncer/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Polisacáridos Fúngicos/farmacología , Neoplasias de la Próstata/patología , Proteómica , Factor de Crecimiento Transformador beta3/farmacología , Actinas/metabolismo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Fibroblastos Asociados al Cáncer/química , Fibroblastos Asociados al Cáncer/fisiología , Puntos de Control del Ciclo Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Progresión de la Enfermedad , Vesículas Extracelulares/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Peso Molecular , Células PC-3 , Neoplasias de la Próstata/terapia , Hongos Shiitake/química , Microambiente Tumoral/inmunología
10.
Arch Immunol Ther Exp (Warsz) ; 69(1): 2, 2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33630157

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive, treatment-resistant cancer. Five-year survival rate is about 9%, one of the lowest among all solid tumors. Such a poor outcome is partly due to the limited knowledge of tumor biology, and the resulting lack of effective treatment options and robust predictive biomarkers. The leukemia inhibitory factor (LIF) has recently emerged as a potential biomarker and therapeutic target for PDAC. Accumulating evidence has suggested that LIF plays a role in supporting cancer evolution as a regulator of cell differentiation, renewal and survival. Interestingly, it can be detected in the serum of PDAC patients at higher concentrations than healthy individuals, this supporting its potential value as diagnostic biomarker. Furthermore, preliminary data indicate that testing for LIF serum concentration or tissue expression may help with treatment response monitoring and prognostication. Finally, studies in PDAC mouse models have also shown that LIF may be a valuable therapeutic target, and first-in-human clinical trial is currently ongoing. This article aims to review the available data on the role of LIF in PDAC promotion, and to discuss the evidence supporting its potential role as a biomarker and target of effective anti-cancer therapy in this setting.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Factor Inhibidor de Leucemia/fisiología , Neoplasias Pancreáticas/patología , Biomarcadores de Tumor/análisis , Fibroblastos Asociados al Cáncer/fisiología , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/terapia , Resistencia a Antineoplásicos , Humanos , Tolerancia Inmunológica , Factor Inhibidor de Leucemia/análisis , Invasividad Neoplásica , Células Madre Neoplásicas/fisiología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/terapia , Microambiente Tumoral
11.
Cancer Res ; 81(7): 1639-1653, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33547159

RESUMEN

Gastric cancer is the third most lethal cancer worldwide, and evaluation of the genomic status of gastric cancer cells has not translated into effective prognostic or therapeutic strategies. We therefore hypothesize that outcomes may depend on the tumor microenvironment (TME), in particular, cancer-associated fibroblasts (CAF). However, very little is known about the role of CAFs in gastric cancer. To address this, we mapped the transcriptional landscape of human gastric cancer stroma by microdissection and RNA sequencing of CAFs from patients with gastric cancer. A stromal gene signature was associated with poor disease outcome, and the transcription factor heat shock factor 1 (HSF1) regulated the signature. HSF1 upregulated inhibin subunit beta A and thrombospondin 2, which were secreted in CAF-derived extracellular vesicles to the TME to promote cancer. Together, our work provides the first transcriptional map of human gastric cancer stroma and highlights HSF1 and its transcriptional targets as potential diagnostic and therapeutic targets in the genomically stable tumor microenvironment. SIGNIFICANCE: This study shows how HSF1 regulates a stromal transcriptional program associated with aggressive gastric cancer and identifies multiple proteins within this program as candidates for therapeutic intervention. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/7/1639/F1.large.jpg.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Vesículas Extracelulares/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Neoplasias Gástricas/patología , Animales , Fibroblastos Asociados al Cáncer/patología , Células Cultivadas , Estudios de Cohortes , Progresión de la Enfermedad , Vesículas Extracelulares/patología , Factores de Transcripción del Choque Térmico/genética , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Transgénicos , Invasividad Neoplásica , Fenotipo , Pronóstico , Vías Secretoras/fisiología , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidad , Análisis de Supervivencia , Microambiente Tumoral/fisiología
12.
Methods Mol Biol ; 2224: 99-111, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606209

RESUMEN

The tumor microenvironment (TME) contains stromal cells in a complex interaction with cancer cells. This relationship has become better understood with the use of fluorescent proteins for in vivo imaging, originally developed by our laboratories. Spectrally-distinct fluorescent proteins can be used for color-coded imaging of the complex interaction of the tumor microenvironment in the living state using cancer cells expressing a fluorescent protein of one color and host mice expressing another-color fluorescent protein. Cancer cells engineered in vitro to express a fluorescent protein were orthotopically implanted into transgenic mice expressing a fluorescent protein of a different color. Confocal microscopy was then used for color-coded imaging of the TME. Color-coded imaging of the TME has enabled us to discover that stromal cells are necessary for metastasis. Patient-derived orthotopic xenograft (PDOX) tumors were labeled by first passaging them orthotopically through transgenic nude mice expressing either green, red, or cyan fluorescent protein in order to label the stromal cells of the tumor (Yang et al., Cancer Res 64:8651-8656, 2004; Yang et al. J Cell Biochem 106: 279-284, 2009). The colored stromal cells become stably associated with the PDOX tumors through multiple passages in transgenic colored nude mice or non-colored nude mice. The fluorescent protein-expressing stromal cells included cancer-associated fibroblasts and tumor-associated macrophages. Color-coded imaging enabled the visualization of apparent fusion of cancer and stromal cells. Color-coded imaging is a powerful tool visualizing the interaction of cancer and stromal cells during cancer progression and treatment.


Asunto(s)
Comunicación Celular/fisiología , Microscopía Confocal/métodos , Neoplasias Pancreáticas/patología , Células del Estroma/fisiología , Microambiente Tumoral/fisiología , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/fisiología , Línea Celular Tumoral , Color , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Ratones Transgénicos , Neoplasias Pancreáticas/metabolismo , Células del Estroma/metabolismo
13.
Methods Mol Biol ; 2179: 243-256, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32939725

RESUMEN

In many solid tumors, collective cell invasion prevails over single-cell dissemination strategies. Collective modes of invasion often display specific front/rear cellular organization, where invasive leader cells arise from cancer cell populations or the tumor stroma. Collective invasion involves coordinated cellular movements which require tight mechanical crosstalk through specific combinations of cell-cell interactions and cell-matrix adhesions. Cancer Associated Fibroblasts (CAFs) have been recently reported to drive the dissemination of epithelial cancer cells through ECM remodeling and direct intercellular contact. However, the cooperation between tumor and stromal cells remains poorly understood. Here we present a simple spheroid invasion assay to assess the role of CAFs in the collective migration of epithelial tumor cells. This method enables the characterization of 3D spheroid invasion patterns through live cell fluorescent labeling combined with spinning disc microscopy. When embedded in extracellular matrix, the invasive strands of spheroids can be tracked and leader/follower organization of CAFs and cancer cells can be quantified.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Movimiento Celular , Rastreo Celular/métodos , Imagenología Tridimensional/métodos , Esferoides Celulares/fisiología , Fibroblastos Asociados al Cáncer/citología , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Rastreo Celular/instrumentación , Matriz Extracelular/química , Humanos , Imagenología Tridimensional/instrumentación , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Esferoides Celulares/citología , Células Tumorales Cultivadas
15.
Gastroenterology ; 160(1): 346-361.e24, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33007300

RESUMEN

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibroblast-rich desmoplastic stroma. Cancer-associated fibroblasts (CAFs) have been shown to display a high degree of interconvertible states including quiescent, inflammatory, and myofibroblastic phenotypes; however, the mechanisms by which this plasticity is achieved are poorly understood. Here, we aim to elucidate the role of CAF plasticity and its impact on PDAC biology. METHODS: To investigate the role of mesenchymal plasticity in PDAC progression, we generated a PDAC mouse model in which CAF plasticity is modulated by genetic depletion of the transcription factor Prrx1. Primary pancreatic fibroblasts from this mouse model were further characterized by functional in vitro assays. To characterize the impact of CAFs on tumor differentiation and response to chemotherapy, various coculture experiments were performed. In vivo, tumors were characterized by morphology, extracellular matrix composition, and tumor dissemination and metastasis. RESULTS: Our in vivo findings showed that Prrx1-deficient CAFs remain constitutively activated. Importantly, this CAF phenotype determines tumor differentiation and disrupts systemic tumor dissemination. Mechanistically, coculture experiments of tumor organoids and CAFs showed that CAFs shape the epithelial-to-mesenchymal phenotype and confer gemcitabine resistance of PDAC cells induced by CAF-derived hepatocyte growth factor. Furthermore, gene expression analysis showed that patients with pancreatic cancer with high stromal expression of Prrx1 display the squamous, most aggressive, subtype of PDAC. CONCLUSIONS: Here, we define that the Prrx1 transcription factor is critical for tuning CAF activation, allowing a dynamic switch between a dormant and an activated state. This work shows that Prrx1-mediated CAF plasticity has significant impact on PDAC biology and therapeutic resistance.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Carcinoma Ductal Pancreático/etiología , Carcinoma Ductal Pancreático/patología , Proteínas de Homeodominio/fisiología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/patología , Animales , Plasticidad de la Célula/fisiología , Modelos Animales de Enfermedad , Ratones
16.
Curr Radiopharm ; 14(4): 374-393, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33357190

RESUMEN

The tumor microenvironment is a dynamic ecosystem where malignant cells interact with the stromal cells sustaining and promoting tumor growth and metastasis. Cancer-associated fibroblasts (CAFs) are the major component of tumor stroma. CAFs control key tumorigenic activities by participating in immune evasion and suppression, extracellular matrix remodeling, neo-angiogenesis, and drug resistance. Therefore, targeting CAFs emerges as an attractive anti-cancer strategy. This review summarized recent advancements in targeting CAFs with diagnostic and therapeutic radiopharmaceuticals using clinically-promising biomarkers. The efforts to improve clinical outcomes via the application of new radiotheranostic compounds are discussed in the context of radionuclide, the pharmacophore, and, more generally, in terms of biomarker specificity and expression across different cancers and CAF phenotypes.


Asunto(s)
Fibroblastos Asociados al Cáncer/efectos de la radiación , Medicina de Precisión , Radiofármacos/farmacología , Microambiente Tumoral , Fibroblastos Asociados al Cáncer/fisiología , Humanos , Fenotipo
17.
Semin Immunol ; 48: 101417, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-33077325

RESUMEN

In tumors, Cancer-Associated Fibroblasts (CAFs) constitute the most prominent component of the tumor microenvironment (TME). CAFs are heterogeneous and composed of different CAF subsets exerting distinct functions in tumors. Specific CAF subpopulations actively influence various aspects of tumor growth, including cancer cell survival and proliferation, angiogenesis, extracellular matrix (ECM) remodeling, metastatic spread and chemoresistance. During the past decade, some CAF subsets have also been shown to modulate anti-tumor immune response. Indeed, they can increase the content in regulatory T lymphocytes and inhibit the activity of effector and cytotoxic immune cells. These functions are mainly controlled by their constitutive secretion of cytokines, chemokines, growth factors and ECM proteins, either directly in the surrounding extracellular space or through micro-vesicles. Some CAFs also express key regulators of immune checkpoints. The different roles played by CAFs, both as immunosuppressor or as physical support for tumor cell progression, set them as promising targets for anti-tumor therapies. In this review, we describe the main current knowledge on CAFs heterogeneity and immunosuppressive microenvironment, as well as their potential therapeutic implications.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Fibroblastos/fisiología , Macrófagos/inmunología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Tolerancia Inmunológica , Neoplasias/terapia , Microambiente Tumoral
18.
Crit Rev Oncog ; 25(1): 1-9, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32865906

RESUMEN

The tumor microenvironment (TME) favors the complex interaction of tumor cells with stromal cells that are recognized to be the regulators of hallmarks of liver cancer growth and metastasis. The most common components of TME include cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and tumor-associated extracellular matrix (ECM) are involved in facilitating the enhancement of liver cancer and can be exploited as potential targets. In addition, cancer stem cells (CSCs) that are known to regulate tumor initiation and progression are present in the TME. All these accumulated factors of the TME represent the driving force for liver cancer progression. This review is focused on the functions of each of the above-mentioned components of the TME and their roles as potential key players in targeting liver cancer.


Asunto(s)
Neoplasias Hepáticas/etiología , Microambiente Tumoral/fisiología , Animales , Fibroblastos Asociados al Cáncer/fisiología , Progresión de la Enfermedad , Exosomas/fisiología , Matriz Extracelular/fisiología , Humanos , Neoplasias Hepáticas/inmunología , Metástasis de la Neoplasia , Macrófagos Asociados a Tumores/fisiología
19.
Cancer Lett ; 492: 71-83, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32860852

RESUMEN

The blood vessel growth inhibitor bevacizumab targets vascular endothelial growth factor (VEGF), a crucial regulator of angiogenesis. Recently, small extracellular vesicles (sEVs) have been demonstrated to be important vehicles in the transport of growth factors to target cells. In this study, we isolated primary carcinoma-associated fibroblasts (CAFs) from four human oral squamous cell carcinoma (OSCC) specimens. Compared with other non-extracellular vesicle components, CAF-derived sEVs were found to be the main regulators of angiogenesis. The ability of CAF sEVs to activate VEGF receptor 2 (VEGFR2) signaling in human umbilical vein endothelial cells (HUVEC) was dependent on the association between sEVs and VEGF. In addition, sEV-bound VEGF secreted by CAFs further activated VEGFR2 signaling in HUVEC in a bevacizumab-resistant manner. VEGF was found to interact with heparan sulfate proteoglycans on the CAF sEV surface and could be released by heparinase I/III. The bioactivity of the dissociated VEGF was retained in vitro and in vivo and could be neutralized by bevacizumab. These findings suggest that the combined use of heparinase and bevacizumab might inhibit angiogenesis in patients with high levels of sEV-bound VEGF.


Asunto(s)
Bevacizumab/uso terapéutico , Fibroblastos Asociados al Cáncer/fisiología , Vesículas Extracelulares/fisiología , Neoplasias de la Boca/irrigación sanguínea , Neovascularización Patológica/etiología , Carcinoma de Células Escamosas de Cabeza y Cuello/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/fisiología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Liasa de Heparina/farmacología , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología
20.
Br J Cancer ; 123(9): 1353-1355, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32830198
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...