Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Hepatology ; 74(3): 1578-1594, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33817801

RESUMEN

BACKGROUND AND AIMS: Studies of the identity and pathophysiology of fibrogenic HSCs have been hampered by a lack of genetic tools that permit specific and inducible fate-mapping of these cells in vivo. Here, by single-cell RNA sequencing of nonparenchymal cells from mouse liver, we identified transcription factor 21 (Tcf21) as a unique marker that restricted its expression to quiescent HSCs. APPROACH AND RESULTS: Tracing Tcf21+ cells by Tcf21-CreER (Cre-Estrogen Receptor fusion protein under the control of Tcf21 gene promoter) targeted ~10% of all HSCs, most of which were located at periportal and pericentral zones. These HSCs were quiescent under steady state but became activated on injuries, generating 62%-67% of all myofibroblasts in fibrotic livers and ~85% of all cancer-associated fibroblasts (CAFs) in liver tumors. Conditional deletion of Transforming Growth Factor Beta Receptor 2 (Tgfbr2) by Tcf21-CreER blocked HSC activation, compromised liver fibrosis, and inhibited liver tumor progression. CONCLUSIONS: In conclusion, Tcf21-CreER-targeted perivenous stellate cells are the main source of myofibroblasts and CAFs in chronically injured livers. TGF-ß signaling links HSC activation to liver fibrosis and tumorigenesis.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Células Estrelladas Hepáticas/citología , Cirrosis Hepática Experimental/patología , Hepatopatías/patología , Neoplasias Hepáticas Experimentales/patología , Miofibroblastos/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Conductos Biliares/cirugía , Tetracloruro de Carbono/toxicidad , Linaje de la Célula , Colestasis , Enfermedad Crónica , Células Estrelladas Hepáticas/metabolismo , Venas Hepáticas/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática Experimental/metabolismo , Hepatopatías/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/metabolismo , Ratones , Miofibroblastos/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual
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.
Cancer Sci ; 112(3): 1251-1261, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33393151

RESUMEN

Asporin (ASPN), a small leucine-rich proteoglycan expressed predominantly by cancer associated fibroblasts (CAFs), plays a pivotal role in tumor progression. ASPN is also expressed by some cancer cells, but its biological significance is unclear. Here, we investigated the effects of ASPN expression in gastric cancer cells. Overexpression of ASPN in 2 gastric cancer cell lines, HSC-43 and 44As3, led to increased migration and invasion capacity, accompanied by induction of CD44 expression and activation of Rac1 and MMP9. ASPN expression increased resistance of HSC-43 cells to oxidative stress by reducing the amount of mitochondrial reactive oxygen species. ASPN induced expression of the transcription factor HIF1α and upregulated lactate dehydrogenase A (LDHA) and PDH-E1α, suggesting that ASPN reprograms HSC-43 cells to undergo anaerobic glycolysis and suppresses ROS generation in mitochondria, which has been observed in another cell line HSC-44PE. By contrast, 44As3 cells expressed high levels of HIF1α in response to oxidant stress and escaped apoptosis regardless of ASPN expression. Examination of xenografts in the gastric wall of ASPN-/- mice revealed that growth of HSC-43 tumors with increased micro blood vessel density was significantly accelerated by ASPN; however, ASPN increased the invasion depth of both HSC-43 and 44As3 tumors. These results suggest that ASPN has 2 distinct effects on cancer cells: HIF1α-mediated resistance to oxidative stress via reprogramming of glucose metabolism, and activation of CD44-Rac1 and MMP9 to promote cell migration and invasion. Therefore, ASPN may be a new therapeutic target in tumor fibroblasts and cancer cells in some gastric carcinomas.


Asunto(s)
Carcinoma/patología , Proteínas de la Matriz Extracelular/metabolismo , Neoplasias Gástricas/patología , Animales , Apoptosis , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/patología , Carcinoma/cirugía , Línea Celular Tumoral , Movimiento Celular , Proteínas de la Matriz Extracelular/genética , Gastrectomía , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/patología , Invasividad Neoplásica/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Estómago/patología , Estómago/cirugía , Neoplasias Gástricas/cirugía , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína de Unión al GTP rac1/metabolismo
4.
Cancer Sci ; 112(10): 4037-4049, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34309966

RESUMEN

Immunotherapy with immune-checkpoint therapy has recently been used to treat oral squamous cell carcinomas (OSCCs). However, improvements in current immunotherapy are expected because response rates are limited. Transforming growth factor-ß (TGF-ß) creates an immunosuppressive tumor microenvironment (TME) by inducing the production of regulatory T-cells (Tregs) and cancer-associated fibroblasts and inhibiting the function of cytotoxic T-lymphocytes (CTLs) and natural killer cells. TGF-ß may be an important target in the development of novel cancer immunotherapies. In this study, we investigated the suppressive effect of TGF-ß on CTL function in vitro using OSCC cell lines and their specific CTLs. Moreover, TGFB1 mRNA expression and T-cell infiltration in 25 OSCC tissues were examined by in situ hybridization and multifluorescence immunohistochemistry. We found that TGF-ß suppressed the function of antigen-specific CTLs in the priming and effector phases in vitro. Additionally, TGF-ß inhibitor effectively restored the CTL function, and TGFB1 mRNA was primarily expressed in the tumor invasive front. Interestingly, we found a significant negative correlation between TGFB1 mRNA expression and the CD8+ T-cell/Treg ratio and between TGFB1 mRNA expression and the Ki-67 expression in CD8+ T-cells, indicating that TGF-ß also suppressed the function of CTLs in situ. Our findings suggest that the regulation of TGF-ß function restores the immunosuppressive TME to active status and is important for developing new immunotherapeutic strategies, such as a combination of immune-checkpoint inhibitors and TGF-ß inhibitors, for OSCCs.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva/métodos , Neoplasias de la Boca/terapia , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Linfocitos T Citotóxicos/efectos de los fármacos , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/inmunología , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Interferón gamma/análisis , Interferón gamma/metabolismo , Antígeno Ki-67/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/metabolismo , ARN Mensajero/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Sales de Tetrazolio/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
5.
Mol Biol Rep ; 48(2): 1773-1786, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33492572

RESUMEN

Oral cancer has been a major problem all across the globe, majorly in the developing countries. With a growing emphasis in the field of cancer research, the contribution of the tumour microenvironment has been gaining a lot of importance in identifying the role of components other than the tumour cells that cause the development of cancer, thus changing the outlook. The review will shed light on the studies that describe the role of microenvironment, its components as well as summarize the studies related to their mechanism in the progression of oral cancer. The literature for the review was derived mainly from Google Scholar and PubMed, in particular concentrating on the most recent papers published in 2019 and 2020, by using the keywords "Cancer, Oral Cancer, Metastasis, OSCC, Tumour microenvironment, CAFs, ECM, Cytokines, Hypoxia, Therapeutics targeting the microenvironment". The study provides insight into the world of micro-environmental regulation of oral cancer, the mechanism by which they interact and how to exploit it as a potential therapeutic haven for treating the disease. The components Cancer-Associated Fibroblasts (CAFs), Tumour-associated Macrophages (TAMs), Tumour-associated neutrophils (TANs), Hypoxic environment, myeloid-derived stem cells (MDSCs) and T regulatory (Tregs) cells and underlying mechanisms that control them will be the targets of study to understand the microenvironment.


Asunto(s)
Citocinas/metabolismo , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Boca/metabolismo , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/metabolismo , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Hipoxia de la Célula , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/inmunología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Matriz Extracelular/inmunología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Macrófagos Asociados a Tumores/citología
6.
Int J Mol Sci ; 22(20)2021 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-34681633

RESUMEN

Fibroblasts constitute a ubiquitous mesenchymal cell type and produce the extracellular matrix (ECM) of connective tissue, thereby providing the structural basis of various organs. Fibroblasts display differential transcriptional patterns unique to the organ of their origin and they can be activated by common stimuli such as transforming growth factor-ß (TGF-ß) and platelet-derived growth factor (PDGF) signaling. Cancer-associated fibroblasts (CAFs) reside in the cancer tissue and contribute to cancer progression by influencing cancer cell growth, invasion, angiogenesis and tumor immunity. CAFs impact on the tumor microenvironment by remodeling the ECM and secreting soluble factors such as chemokines and growth factors. Differential expression patterns of molecular markers suggest heterogeneous features of CAFs in terms of their function, pathogenic role and cellular origin. Recent studies elucidated the bimodal action of CAFs on cancer progression and suggest a subgroup of CAFs with tumor-suppressive effects. This review attempts to describe cellular features of colorectal CAFs with an emphasis on their heterogeneity and functional diversity.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias Colorrectales/patología , Fibroblastos Asociados al Cáncer/citología , Neoplasias Colorrectales/metabolismo , Matriz Extracelular/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/metabolismo , Especificidad de Órganos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
7.
Int J Mol Sci ; 22(20)2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34681692

RESUMEN

Osteosarcoma (OS) is an aggressive bone tumor that mainly affects children and adolescents. OS has a strong tendency to relapse and metastasize, resulting in poor prognosis and survival. The high heterogeneity and genetic complexity of OS make it challenging to identify new therapeutic targets. Mesenchymal stem cells (MSCs) are multipotent stem cells that can differentiate into adipocytes, osteoblasts, or chondroblasts. OS is thought to originate at some stage in the differentiation process of MSC to pre-osteoblast or from osteoblast precursors. MSCs contribute to OS progression by interacting with tumor cells via paracrine signaling and affect tumor cell proliferation, invasion, angiogenesis, immune response, and metastasis. Extracellular vesicles (EVs), secreted by OS cells and MSCs in the tumor microenvironment, are crucial mediators of intercellular communication, driving OS progression by transferring miRNAs/RNA and proteins to other cells. MSC-derived EVs have both pro-tumor and anti-tumor effects on OS progression. MSC-EVs can be also engineered to deliver anti-tumor cargo to the tumor site, which offers potential applications in MSC-EV-based OS treatment. In this review, we highlight the role of MSCs in OS, with a focus on EV-mediated communication between OS cells and MSCs and their role in OS pathogenesis and therapy.


Asunto(s)
Neoplasias Óseas/patología , Vesículas Extracelulares/metabolismo , Osteosarcoma/patología , Neoplasias Óseas/terapia , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Comunicación Celular , Vesículas Extracelulares/trasplante , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Metástasis de la Neoplasia , Osteosarcoma/terapia , Microambiente Tumoral
8.
Cell Mol Biol (Noisy-le-grand) ; 66(2): 36-40, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415924

RESUMEN

Hepatocellular carcinoma is known to be a common predominant cancer in adults, especially in eastern countries. Immune response and cancer-associated fibroblasts (CAFs) have significant influences on tumor development. However, the interaction between CAFs and immunotherapy is unclear in hepatocellular carcinoma. We measured the number of activated fibroblasts in hepatocellular carcinoma samples and samples taken from normal liver tissues. A total of 20 patients' fresh hepatocellular carcinoma and normal tissues which were surrounding the tumor were obtained from the surgery and used for evaluating alpha-SMA expression. We investigated the effects of CAFs in anti-tumor immunity in hepatocellular carcinoma animal model. The effects of CAFs in inducing anti-PD-1 treatment resistance were also measured in a preclinical animal model. Activated fibroblasts were highly accumulated in hepatocellular carcinoma tissues but not in surrounding normal tissues. CAFs showed a significant tumor-promoting effect in an immunocompetent model. The infiltration and function of some immune cells like myeloid-derived suppressive cells and T-cells were increased by CAFs. CAFs also reduced the number and activation of tumor-infiltrating cytotoxic T-cell in tumor tissue. In the treatment model, tumors with a higher amount of CAFs had been insensitive to therapy with anti-PD-1. CAFs are potent inducers of immunosuppression in hepatocellular carcinoma. Depleting CAFs rescued the antitumor immunity in the hepatocellular model and could be a novel treatment to combine with the existing immunotherapy.


Asunto(s)
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Actinas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/trasplante , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Citocinas/metabolismo , Dasatinib/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Inmunoterapia , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
9.
Mol Cancer ; 18(1): 68, 2019 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-30927911

RESUMEN

BACKGROUND: Although the tumor stroma in solid tumors like gastric cancer (GC) plays a crucial role in chemo-resistance, specific targets to inhibit the interaction between the stromal and cancer cells have not yet been utilized in clinical practice. The present study aims to determine whether cancer-associated fibroblasts (CAFs), a major component of the tumor stroma, confer chemotherapeutic resistance to GC cells, and to discover potential targets to improve chemo-response in GC. METHODS: To identify CAF-specific proteins and signal transduction pathways affecting chemo-resistance in GC cells, secretome and transcriptome analyses were performed. We evaluated the inhibiting effect of CAF-specific protein in in vivo and in vitro models and investigated the expression of CAF-specific protein in human GC tissues. RESULTS: Secretome and transcriptome data revealed that interleukin-6 (IL-6) is a CAF-specific secretory protein that protects GC cells via paracrine signaling. Furthermore, CAF-induced activation of the Janus kinase 1-signal transducer and activator of transcription 3 signal transduction pathway confers chemo-resistance in GC cells. CAF-mediated inhibition of chemotherapy-induced apoptosis was abrogated by the anti-IL-6 receptor monoclonal antibody tocilizumab in various experimental models. Clinical data revealed that IL-6 was prominently expressed in the stromal portion of GC tissues, and IL-6 upregulation in GC tissues was correlated with poor responsiveness to chemotherapy. CONCLUSIONS: Our data provide plausible evidence for crosstalk between GC cells and CAFs, wherein IL-6 is a key contributor to chemoresistance. These findings suggest the potential therapeutic application of IL-6 inhibitors to enhance the responsiveness to chemotherapy in GC.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Fluorouracilo/administración & dosificación , Interleucina-6/genética , ARN Interferente Pequeño/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Animales , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Resistencia a Antineoplásicos/efectos de los fármacos , Fluorouracilo/farmacología , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Ratones , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Int J Cancer ; 145(11): 3064-3077, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31032902

RESUMEN

Myofibroblasts are a population of highly contractile fibroblasts that express and require the activity of the transcription factor Snail1. Cancer-associated fibroblasts (CAFs) correlate with low survival of cancer patients when present in the stroma of primary tumors. Remarkably, the presence of myofibroblastic CAFs (which express Snail1) creates mechanical properties in the tumor microenvironment that support metastasis. However, therapeutic blockage of fibroblast activity in patients with cancer is a double-edged sword, as normal fibroblast activities often restrict tumor cell invasion. We used fibroblasts depleted of Snail1 or protein arginine methyltransferases 1 and 4 (PRMT1/-4) to identify specific epigenetic modifications induced by TGFß/Snail1. Furthermore, we analyzed the in vivo efficiency of methyltransferase inhibitors using mouse models of wound healing and metastasis, as well as fibroblasts isolated from patients with idiopathic pulmonary fibrosis (IPF). Mechanistically, TGFß-induced Snail1 promotes the epigenetic mark of asymmetrically dimethylated arginine. Critically, we found that inhibitors of methyltransferases prevent myofibroblast activity (but not regular fibroblast activity) in the extracellular matrix, both in cell culture and in vivo. In a mouse breast cancer model, the inhibitor sinefungin reduces both the myofibroblast activity in the tumor stroma and the metastatic burden in the lung. Two distinct inhibitors effectively blocked the exacerbated myofibroblast activity of patient-derived IPF fibroblasts. Our data reveal epigenetic regulation of myofibroblast transdifferentiation in both wound healing and in disease (fibrosis and breast cancer). Thus, methyltransferase inhibitors are good candidates as therapeutic reagents for these diseases.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Metiltransferasas/antagonistas & inhibidores , Miofibroblastos/efectos de los fármacos , Factores de Transcripción de la Familia Snail/genética , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Neoplasias de la Mama/enzimología , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Transdiferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Eliminación de Gen , Humanos , Fibrosis Pulmonar Idiopática/enzimología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Metiltransferasas/genética , Ratones , Miofibroblastos/citología , Miofibroblastos/enzimología , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Sci ; 110(5): 1609-1620, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30868675

RESUMEN

Cancer tissues consist of cancer cells, surrounding stromal cells and the extracellular matrix. Cancer-associated fibroblasts (CAF) are one of the key components of stromal cells. CAF have a great impact on the behavior of cancer cells, including proliferation, invasion, metastasis and chemoresistance in many ways. However, the underlying mechanism had not been fully elucidated. In this study, we investigated the role of CAF in cisplatin resistance of lung cancer cells. By using conditioned medium from CAF (CAF-CM), we found that CAF decreased the sensitivity of lung cancer cells to cisplatin. RNA sequencing results showed that CAF expressed a higher level of Annexin A3 (ANXA3) than normal fibroblasts (NF), and CAF-CM incubation increased the ANXA3 level in lung cancer cells. Overexpression of ANXA3 in lung cancer cells increased cisplatin resistance and activated c-jun N-terminal kinase (JNK), whereas knockdown of ANXA3 increased cisplatin sensitivity. Further study showed that CAF-CM enhanced cisplatin resistance by inhibiting cisplatin-induced apoptosis, determined by repression of caspase-3 and caspase-8, through activation of the ANXA3/JNK pathway. Conversely, suppression of JNK activation by specific inhibitor retarded the effect of CAF-CM and ANXA3 on cisplatin sensitivity. Taken together, our study demonstrated that CAF potentiated chemoresistance of lung cancer cells through a novel ANXA3/JNK pathway both in vitro and in vivo, suggesting ANXA3 could be a potential therapeutic target for the treatment of chemoresistant cancer.


Asunto(s)
Anexina A3/genética , Fibroblastos Asociados al Cáncer/citología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Neoplasias Pulmonares/patología , Células A549 , Animales , Anexina A3/metabolismo , Apoptosis/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Análisis de Secuencia de ARN/métodos , Células Tumorales Cultivadas , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Invest ; 37(3): 134-143, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30961403

RESUMEN

Cancer-associated fibroblasts (CAFs) play an important role in the development and progression of cancer by inducing epithelial-mesenchymal transition (EMT). In this study, we investigated the role of CAFs in endometrial cancer (EC) cells. We found that the pituitary tumor transforming gene (PTTG) expression was significantly increased in EC cell lines compared to normal human endometrial epithelial cells. Furthermore, CAFs could induce PTTG over-expression and increase EC cell invasion and migration in vitro. In addition, CAFs also induced EMT in EC cells. This study demonstrated that CAFs induced EMT in endometrial cancer cells by regulating PTTG.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias Endometriales/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Securina/genética , Fibroblastos Asociados al Cáncer/citología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Técnicas de Cocultivo , Neoplasias Endometriales/patología , Endometrio/metabolismo , Endometrio/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Invasividad Neoplásica
13.
Angew Chem Int Ed Engl ; 58(43): 15287-15294, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31402550

RESUMEN

Tumor complexity makes the development of highly sensitive tumor imaging probes an arduous task. Here, we construct a peptide-based near-infrared probe that is responsive to fibroblast activation protein-α (FAP-α), and specifically forms nanofibers on the surface of cancer-associated fibroblasts (CAFs) in situ. The assembly/aggregation-induced retention (AIR) effect results in enhanced accumulation and retention of the probe around the tumor, resulting in a 5.5-fold signal enhancement in the tumor 48 h after administration compared to that of a control molecule that does not aggregate. The probe provides a prolonged detectable window of 48 h for tumor diagnosis. The selective assembly of the probe results in a signal intensity over four- and fivefold higher in tumor than in the liver and kidney, respectively. With enhanced tumor imaging capability, this probe can visualize small tumors around 2 mm in diameter.


Asunto(s)
Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Nanofibras/química , Péptidos/metabolismo , Serina Endopeptidasas/metabolismo , Espectroscopía Infrarroja Corta/métodos , Secuencia de Aminoácidos , Animales , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Endopeptidasas , Colorantes Fluorescentes/química , Gelatinasas/química , Humanos , Proteínas de la Membrana/química , Ratones , Ratones Desnudos , Microscopía Electrónica de Rastreo , Neoplasias/diagnóstico por imagen , Péptidos/síntesis química , Péptidos/química , Serina Endopeptidasas/química , Distribución Tisular , Trasplante Heterólogo
14.
Mol Cancer ; 17(1): 114, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30075793

RESUMEN

Exosome production from cancer-associated fibroblasts seems to be an important driver of tumor progression. We report the first in-depth biotype characterization of ncRNAs, analyzed by Next Generation Sequencing and Bioinformatics, expressed in established primary human normal and cancer-associated fibroblasts (CAFs) from cancer and normal mucosa tissues from 9 colorectal cancer patients, and/or packaged in their derived exosomes. Differential representation and enrichment analyses based on these ncRNAs revealed a significant number of differences between the ncRNA content of exosomes and the expression patterns of the normal and cancer-associated fibroblast cells. ncRNA regulatory elements are specifically packaged in CAF-derived exosomes, supporting a specific cross-talk between CAFs and colon cancer cells and/or other stromal cells, mediated by exosomes. These sncRNAs are potential biomarkers present in cancer-associated fibroblast-derived exosomes, which should thereby contribute to developing new non-invasive diagnostic, prognostic and predictive methods for clinical applications in management of cancer patients.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Neoplasias Colorrectales/genética , Exosomas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ARN no Traducido/genética , Biomarcadores de Tumor/genética , Fibroblastos Asociados al Cáncer/química , Movimiento Celular , Proliferación Celular , Células Cultivadas , Fibroblastos/química , Fibroblastos/citología , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , Análisis de Secuencia de ARN , Microambiente Tumoral
15.
Int J Cancer ; 143(2): 383-395, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29441570

RESUMEN

Carcinoma-associated fibroblasts (CAFs) play a key onco-supportive role during prostate cancer (PCa) development and progression. We previously reported that the reactive oxygen species (ROS)-producing enzyme NADPH oxidase 4 (Nox4) is essential for TGFß1-mediated activation of primary prostate human fibroblasts to a CAF-like phenotype. This study aimed to further investigate the functional relevance of prostatic Nox4 and determine whether pharmacological inhibition of stromal Nox4 abrogates paracrine-mediated PCa-relevant processes. RNA in situ hybridization revealed significantly elevated Nox4 mRNA levels predominantly in the peri-tumoral stroma of clinical PCa with intense stromal Nox4 staining adjacent to tumor foci expressing abundant TGFß protein levels. At pharmacologically relevant concentrations, the Nox1/Nox4 inhibitor GKT137831 attenuated ROS production, CAF-associated marker expression and migration of TGFß1-activated but not nonactivated primary human prostate fibroblasts. Similar effects were obtained upon shRNA-mediated silencing of Nox4 but not Nox1 indicating that GKT137831 primarily abrogates TGFß1-driven fibroblast activation via Nox4 inhibition. Moreover, inhibiting stromal Nox4 abrogated the enhanced proliferation and migration of PCa cell lines induced by TGFß1-activated prostate fibroblast conditioned media. These effects were not restricted to recombinant TGFß1 as conditioned media from PCa cell lines endogenously secreting high TGFß1 levels induced fibroblast activation in a stromal Nox4- and TGFß receptor-dependent manner. Importantly, GKT137831 also attenuated PCa cell-driven fibroblast activation. Collectively, these findings suggest the TGFß-Nox4 signaling axis is a key interface to dysregulated reciprocal stromal-epithelial interactions in PCa pathophysiology and provide a strong rationale for further investigating the applicability of Nox4 inhibition as a stromal-targeted approach to complement current PCa treatment modalities.


Asunto(s)
Fibroblastos Asociados al Cáncer/efectos de los fármacos , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , Neoplasias de la Próstata/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Humanos , Masculino , Estrés Oxidativo/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Pirazolonas , Piridonas , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Células del Estroma/citología , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
16.
Gastroenterology ; 152(5): 964-979, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28111227

RESUMEN

Mesenchymal cells in the intestine comprise a variety of cell types of diverse origins, functions, and molecular markers. They provide mechanical and structural support and have important functions during intestinal organogenesis, morphogenesis, and homeostasis. Recent studies of the human transcriptome have revealed their importance in the development of colorectal cancer, and studies from animal models have provided evidence for their roles in the pathogenesis of colitis-associated cancer and sporadic colorectal cancer. Mesenchymal cells in tumors, called cancer-associated fibroblasts, arise via activation of resident mesenchymal cell populations and the recruitment of bone marrow-derived mesenchymal stem cells and fibrocytes. Cancer-associated fibroblasts have a variety of activities that promote colon tumor development and progression; these include regulation of intestinal inflammation, epithelial proliferation, stem cell maintenance, angiogenesis, extracellular matrix remodeling, and metastasis. We review the intestinal mesenchymal cell-specific pathways that regulate these processes, with a focus on their roles in mediating interactions between inflammation and carcinogenesis. We also discuss how increasing our understanding of intestinal mesenchymal cell biology and function could lead to new strategies to identify and treat colitis-associated cancers.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Colitis , Neoplasias del Colon , Intestinos/citología , Células Madre Mesenquimatosas/citología , Carcinogénesis , Diferenciación Celular , Proliferación Celular , Matriz Extracelular , Fibroblastos/citología , Humanos , Inflamación , Miocitos del Músculo Liso/citología , Miofibroblastos/citología , Neovascularización Patológica , Pericitos/citología , Microambiente Tumoral
17.
Cell Physiol Biochem ; 49(3): 869-883, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30184548

RESUMEN

BACKGROUND/AIMS: The malignant biological behavior of gastric cancer(GC) is not only determined by cancer cells alone, but also closely regulated by the microenvironment. Fibroblasts represent a large proportion of the components in the tumor microenvironment, and they promote the development of disease. Currently, accumulating evidence suggests that exosomes can function as intercellular transport systems to relay their contents, especially microRNAs(miRNAs). METHODS: First, we detected the highly-expressed level of miR-27a in exosomes isolated from gastric cancer cells by qRT-PCR. MiR-27a -over-expressed models in vitro and in vivo were established to investigate the transformation of cancer-associated fibroblasts observed by Western blotting, and the malignant behavior of gastric cancer cells using the methods CCK8 and Transwell. Moreover, the downregulation of CSRP2 in fibroblasts was used to evaluate the promotion of malignancy of gastric cancer using the methods CCK8 and Transwell. RESULTS: In this study, we found a marked high level of miR-27a in exosomes derived from GC cells. miR-27a was found to function an oncogene that not only induced the reprogramming of fibroblasts into cancer-associated fibroblasts(CAFs), but also promoted the proliferation, motility and metastasis of cancer cells in vitro and in vivo. Conversely, CAFs with over-expression of miR-27a could pleiotropically increase the malignant behavior of the GC cells. For the first time, we revealed that CSRP2 is a downstream target of miR-27a. CSRP2 downregulation could increase the proliferation and motility of GC cells. CONCLUSION: Thus, this report indicates that miR-27a in exosomes derived from GC cells has a crucial impact on the microenvironment and may be used as a potential therapeutic target in the treatment of GC.


Asunto(s)
Exosomas/metabolismo , MicroARNs/metabolismo , Animales , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica , Regulación hacia Abajo , Exosomas/genética , Femenino , Humanos , Proteínas con Dominio LIM/antagonistas & inhibidores , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
18.
Cell Physiol Biochem ; 51(1): 315-336, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30453281

RESUMEN

BACKGROUND/AIMS: Gap junctions, which are assembled by connexins, can directly connect the cytoplasm of adjacent cells and enable gap junctional intercellular communication (GJIC) as well as metabolic coupling between neighboring cells. Here, we investigated the role of connexin 43 (Cx43) and its derived GJIC in the interplay between non-small cell lung cancer (NSCLC) cells and cancer-associated fibroblasts (CAFs). METHODS: CAFs and NSCLC cells were co-cultured with direct contact and separated using flow cytometry. Glucose uptake, lactate production, and the expression and activity of PKM-2 and LDH-A in sorted CAFs were measured by a colorimetric assay, western blotting, and enzyme-linked immunosorbent assay (ELISA). Meanwhile, E-cadherin and N-cadherin expression and the migration and invasion of sorted NSCLC cells were detected by western blotting, wound width, and Transwell assays. Pyruvate, acetyl-CoA, and citric acid levels, ATP levels, and LDH-B and α-KG activity in sorted NSCLC cells were determined by a colorimetric or fluorometric assay and ELISA, respectively. Functional GJIC between cells and the subcellular location of connexins were detected by a "Parachute" assay and immunofluorescence. Levels of α-SMA, Cx43, and LDH-B in tissue from patients with NSCLC were determined by immunohistochemistry. RESULTS: Cx43 accumulated in the plasma membrane, which favored the assembly of asymmetric unidirectional GJIC from CAFs to NSCLC cells. CAFs underwent increased aerobic glycolysis and promoted the epithelial-mesenchymal transition, migration, and invasion of NSCLC cells. In contrast, NSCLC cells experienced enhanced oxidative phosphorylation upon CAF stimulation, with an increase in ATP generation and thereby activation of the PI3K/Akt and MAPK/ERK pathways. Metabolic coupling between CAFs and NSCLC cells was under the strict control of Cx43-formed unidirectional GJIC. Patients with high tri-expression of α-SMA, Cx43, and LDH-B had the shortest overall survival and relapse-free survival compared with those with individual overexpression or high bi-expression. CONCLUSION: Cx43-formed unidirectional GJIC plays a critical role in mediating close metabolic cooperation between CAFs and NSCLC cells to support the malignant progression of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Neoplasias Pulmonares/patología , Adenosina Trifosfato/metabolismo , Cadherinas/metabolismo , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Línea Celular Tumoral , Movimiento Celular , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Transición Epitelial-Mesenquimal , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal
19.
Helicobacter ; 23(6): e12538, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30246423

RESUMEN

BACKGROUND: Major human gastrointestinal pathogen Helicobacter pylori (H. pylori) colonizes the gastric mucosa causing inflammation and severe complications including cancer, but the involvement of fibroblasts in the pathogenesis of these disorders in H. pylori-infected stomach has been little studied. Normal stroma contains few fibroblasts, especially myofibroblasts. Their number rapidly increases in the reactive stroma surrounding inflammatory region and neoplastic tissue; however, the interaction between H. pylori and fibroblasts remains unknown. We determined the effect of coincubation of normal rat gastric fibroblasts with alive H. pylori (cagA+vacA+) and H. pylori (cagA-vacA-) strains on the differentiation of these fibroblasts into cells possessing characteristics of cancer-associated fibroblasts (CAFs) able to induce epithelial-mesenchymal transition (EMT) of normal rat gastric epithelial cells (RGM-1). MATERIALS AND METHODS: The panel of CAFs markers mRNA was analyzed in H. pylori (cagA+vacA+)-infected fibroblasts by RT-PCR. After insert coculture of differentiated fibroblasts with RGM-1 cells from 24 up to 48, 72, and 96 hours, the mRNA expression for EMT-associated genes was analyzed by RT-PCR. RESULTS: The mRNA expression for CAFs markers was significantly increased after 72 hours of infection with H. pylori (cagA+vacA+) but not H. pylori (cagA-vacA-) strain. Following coculture with CAFs, RGM-1 cells showed significant decrease in E-cadherin mRNA, and the parallel increase in the expression of Twist and Snail transcription factors mRNA was observed along with the overexpression of mRNAs for TGFßR, HGFR, FGFR, N-cadherin, vimentin, α-SMA, VEGF, and integrin-ß1. CONCLUSION: Helicobacter pylori (cagA+vacA+) strain induces differentiation of normal fibroblasts into CAFs, likely to initiate the EMT process in RGM-1 epithelial cell line.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Infecciones por Helicobacter/microbiología , Humanos , ARN Mensajero/genética , Ratas , Estómago/citología
20.
J Pathol ; 241(3): 313-315, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27753093

RESUMEN

Many non-tumour host cells such as inflammatory cells, fibroblasts, and endothelial cells are present in the tumour microenvironment and affect the malignant potential and chemo-resistance of the tumour cells. Macrophages and fibroblasts are the main components of infiltrating stromal cells and are referred to as tumour-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs), respectively. TAMs and CAFs are reported to be involved in tumour progression, although their functions change to those of an anti-tumour phenotype under specific conditions. Notably, recent work published in The Journal of Pathology by Hashimoto and colleagues provided critical evidence indicating the significance of collaboration between TAMs and CAFs for tumour progression. They showed that cell-cell interaction between these two cell types induced recruitment and activation of each other, and that the combined activities of these cells were involved in neuroblastoma progression. Although many research groups are now interested in the significance of stromal cells such as CAFs and TAMs for tumour progression, only a few studies have been published describing the cell-cell interactions of these cells. Cell-cell interactions of stromal cells potentially play important roles in tumour progression and should be a focus for further oncology research. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Comunicación Celular/fisiología , Macrófagos/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/fisiología , Progresión de la Enfermedad , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA