Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Pathol ; 262(1): 61-75, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37796386

RESUMO

Pancreatic stellate cells (PSCs) are stromal cells in the pancreas that play an important role in pancreatic pathology. In chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC), PSCs are known to get activated to form myofibroblasts or cancer-associated fibroblasts (CAFs) that promote stromal fibroinflammatory reactions. However, previous studies on PSCs were mainly based on the findings obtained using ex vivo expanded PSCs, with few studies that addressed the significance of in situ tissue-resident PSCs using animal models. Their contributions to fibrotic reactions in CP and PDAC are also lesser-known. These limitations in our understanding of PSC biology have been attributed to the lack of specific molecular markers of PSCs. Herein, we established Meflin (Islr), a glycosylphosphatidylinositol-anchored membrane protein, as a PSC-specific marker in both mouse and human by using human pancreatic tissue samples and Meflin reporter mice. Meflin-positive (Meflin+ ) cells contain lipid droplets and express the conventional PSC marker Desmin in normal mouse pancreas, with some cells also positive for Gli1, the marker of pancreatic tissue-resident fibroblasts. Three-dimensional analysis of the cleared pancreas of Meflin reporter mice showed that Meflin+ PSCs have long and thin cytoplasmic protrusions, and are localised on the abluminal side of vessels in the normal pancreas. Lineage tracing experiments revealed that Meflin+ PSCs constitute one of the origins of fibroblasts and CAFs in CP and PDAC, respectively. In these diseases, Meflin+ PSC-derived fibroblasts showed a distinctive morphology and distribution from Meflin+ PSCs in the normal pancreas. Furthermore, we showed that the genetic depletion of Meflin+ PSCs accelerated fibrosis and attenuated epithelial regeneration and stromal R-spondin 3 expression, thereby implying that Meflin+ PSCs and their lineage cells may support tissue recovery and Wnt/R-spondin signalling after pancreatic injury and PDAC development. Together, these data indicate that Meflin may be a marker specific to tissue-resident PSCs and useful for studying their biology in both health and disease. © 2023 The Pathological Society of Great Britain and Ireland.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Pancreatite Crônica , Animais , Humanos , Camundongos , Carcinoma Ductal Pancreático/patologia , Fibrose , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Células Estreladas do Pâncreas/patologia , Pancreatite Crônica/genética , Pancreatite Crônica/metabolismo , Pancreatite Crônica/patologia , Regeneração
2.
Br J Cancer ; 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38849479

RESUMO

BACKGROUND: The proliferation of cancer-associated fibroblasts (CAFs) hampers drug delivery and anti-tumor immunity, inducing tumor resistance to immune checkpoint blockade (ICB) therapy. However, it has remained a challenge to develop therapeutics that specifically target or modulate CAFs. METHODS: We investigated the involvement of Meflin+ cancer-restraining CAFs (rCAFs) in ICB efficacy in patients with clear cell renal cell carcinoma (ccRCC) and urothelial carcinoma (UC). We examined the effects of Am80 (a synthetic retinoid) administration on CAF phenotype, the tumor immune microenvironment, and ICB efficacy in cancer mouse models. RESULTS: High infiltration of Meflin+ CAFs correlated with ICB efficacy in patients with ccRCC and UC. Meflin+ CAF induction by Am80 administration improved ICB efficacy in the mouse models of cancer. Am80 exerted this effect when administered prior to, but not concomitant with, ICB therapy in wild-type but not Meflin-deficient mice. Am80-mediated induction of Meflin+ CAFs was associated with increases in antibody delivery and M1-like tumor-associated macrophage (TAM) infiltration. Finally, we showed the role of Chemerin produced from CAFs after Am80 administration in the induction of M1-like TAMs. CONCLUSION: Our data suggested that Am80 administration prior to ICB therapy increases the number of Meflin+ rCAFs and ICB efficacy by inducing changes in TAM phenotype.

3.
Pathol Int ; 72(3): 161-175, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35020975

RESUMO

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.


Assuntos
Fibroblastos Associados a Câncer/fisiologia , Imunoglobulinas/análise , Proteínas de Membrana/análise , Neoplasias Pancreáticas/diagnóstico , Animais , Biomarcadores/análise , Fibroblastos Associados a Câncer/citologia , Fibroblastos Associados a Câncer/patologia , Modelos Animais de Doenças , Imunoglobulinas/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout/genética , Neoplasias Pancreáticas/patologia
4.
Cancer Sci ; 111(4): 1047-1057, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32060987

RESUMO

The roles of cancer-associated fibroblasts (CAF) in the progression of various types of cancers are well established. CAF promote cancer progression through pleiotropic mechanisms, including the secretion of soluble factors and extracellular matrix, physical interactions with cancer cells, and the regulation of angiogenesis, immunity and metabolism. Their contribution to therapeutic resistance is also well appreciated. Therefore, CAF have been considered as a therapeutic target in cancer. However, recent studies in autochthonous pancreatic cancer models suggest that specific subset(s) of CAF exhibit cancer-restraining roles, indicating that CAF are functionally and molecularly heterogeneous, which is supported by recent single-cell transcriptome analyses. While cancer-promoting CAF (pCAF) have been extensively studied, the nature and specific marker(s) of cancer-restraining CAF (rCAF) have remained uncharacterized. Interestingly, a recent study provided insight into the nature of rCAF and suggested that they may share molecular properties with pancreatic stellate cells (PSC) and mesenchymal stem/stromal cells (MSC). Complicating this finding is that PSC and MSC have been shown to promote the formation of a tumor-permissive and tumor-promoting environment in xenograft tumor models. However, these cells undergo significant transcriptional and epigenetic changes during ex vivo culture, which confounds the interpretation of experimental results based on the use of cultured cells. In this short review, we describe recent studies and hypotheses on the identity of rCAF and discuss their analogy to fibroblasts that suppress fibrosis in fibrotic diseases. Finally, we discuss how these findings can be exploited to develop novel anticancer therapies in the future.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Transformação Celular Neoplásica/genética , Neoplasias/genética , Neovascularização Patológica/genética , Animais , Fibroblastos Associados a Câncer/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Matriz Extracelular/genética , Xenoenxertos , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Neoplasias/patologia , Neovascularização Patológica/patologia , Células Estreladas do Pâncreas/metabolismo , Microambiente Tumoral/genética
5.
Biochem Biophys Res Commun ; 532(3): 406-413, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-32888647

RESUMO

The canonical Wnt signaling pathway plays a crucial role in embryonic development, tissue homeostasis and cancer progression. The binding of Wnt ligands to their cognate receptors, the Frizzled (Fzd) family of proteins, recruits Dishevelled segment polarity protein (Dvl) to the plasma membrane and induces its phosphorylation via casein kinase 1 (CK1), which leads to the activation of ß-catenin. Previous studies showed that Dishevelled-associating protein with a high frequency of leucine residues (Daple) is an important component of the Wnt signaling pathway and essential for Dvl phosphorylation. However, the mechanism by which Daple promotes CK1-mediated phosphorylation of Dvl is not fully understood. In this study, we found that Daple overexpression induced CK1ε-mediated Dvl2 phosphorylation at threonine 224 (Thr224). A Daple mutant (Daple ΔGCV) that lacks a carboxyl-terminal motif to associate with Dvl, retained the ability to interact with CK1ε, but did not induce Dvl phosphorylation, suggesting the importance of the Daple/Dvl/CK1ε trimeric protein complex. We further found that Thr224 phosphorylation of Dvl was required for full activation of ß-catenin transcriptional activity. Consistent with this, wild-type Daple promoted ß-catenin transcriptional activity, following dissociation of ß-catenin and axin. Finally, Wnt3a stimulation increased the membrane localization of Daple and its association with Dvl, and Daple knockdown attenuated Wnt3a-mediated ß-catenin transcriptional activity. Collectively, these data suggested a essential role of spatial Daple localization in CK1ε-mediated activation of Dvl in the canonical Wnt signaling pathway.


Assuntos
Caseína Quinase 1 épsilon/metabolismo , Proteínas Desgrenhadas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas dos Microfilamentos/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Proteínas de Transporte/metabolismo , Proteínas Desgrenhadas/química , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Células L , Camundongos , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/genética , Fosforilação , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
6.
Cancer Sci ; 109(1): 141-153, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29151270

RESUMO

Ganglioside GD2 is specifically expressed in small-cell lung cancer (SCLC) cells, leading to enhancement of malignant phenotypes, such as cell proliferation and migration. However, how GD2 promotes malignant phenotypes in SCLC cells is not well known. In this study, to reveal the mechanisms by which GD2 increases malignant phenotypes in SCLC cells, we used enzyme-mediated activation of radical sources combined with mass spectrometry in GD2+ SCLC cells. Consequently, we identified ASC amino acid transporter 2 (ASCT2), a major glutamine transporter, which coordinately works with GD2. We showed that ASCT2 was highly expressed in glycolipid-enriched microdomain/rafts in GD2+ SCLC cells, and colocalized with GD2 in both proximity ligation assay and immunocytostaining, and bound with GD2 in immunoprecipitation/TLC immunostaining. Malignant phenotypes of GD2+ SCLC cells were enhanced by glutamine uptake, and were suppressed by L-γ-glutamyl-p-nitroanilide, a specific inhibitor of ASCT2, through reduced phosphorylation of p70 S6K1 and S6. These results suggested that ASCT2 enhances glutamine uptake in glycolipid-enriched microdomain/rafts in GD2+ SCLC cells, leading to the enhancement of cell proliferation and migration through increased phosphorylation of the mTOR complex 1 signaling axis.


Assuntos
Sistema ASC de Transporte de Aminoácidos/metabolismo , Gangliosídeos/metabolismo , Neoplasias Pulmonares/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Carcinoma de Pequenas Células do Pulmão/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Glutamina/análogos & derivados , Glutamina/metabolismo , Glutamina/farmacologia , Humanos , Microdomínios da Membrana/metabolismo
7.
Cancer Sci ; 109(11): 3643-3656, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30194792

RESUMO

Pathological observations show that cancer cells frequently invade the surrounding stroma in collective groups rather than through single cell migration. Here, we studied the role of the actin-binding protein Girdin, a specific regulator of collective migration of neuroblasts in the brain, in collective cancer cell migration. We found that Girdin was essential for the collective migration of the skin cancer cell line A431 on collagen gels as well as their fibroblast-led collective invasion in an organotypic culture model. We provide evidence that Girdin binds to ß-catenin that plays important roles in the Wnt signaling pathway and in E-cadherin-mediated cell-cell adhesion. Girdin-depleted cells displayed scattering and impaired E-cadherin-specific cell-cell adhesion. Importantly, Girdin depletion led to impaired cytoskeletal association of the ß-catenin complex, which was accompanied by changes in the supracellular actin cytoskeletal organization of cancer cell cohorts on collagen gels. Although the underlying mechanism is unclear, this observation is consistent with the established role of the actin cytoskeletal system and cell-cell adhesion in the collective behavior of cells. Finally, we showed the correlation of the expression of Girdin with that of the components of the E-cadherin complex and the differentiation of human skin cancer. Collectively, our results suggest that Girdin is an important modulator of the collective behavior of cancer cells.


Assuntos
Citoesqueleto/metabolismo , Proteínas dos Microfilamentos/metabolismo , Neoplasias Cutâneas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , beta Catenina/metabolismo , Animais , Antígenos CD , Sítios de Ligação , Caderinas/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Células HeLa , Humanos , Camundongos , Proteínas dos Microfilamentos/química , Ligação Proteica , Proteínas de Transporte Vesicular/química , Via de Sinalização Wnt
8.
Pathol Res Pract ; 245: 154443, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37030166

RESUMO

Osteosarcoma, the most common primary malignant bone tumor, is defined by the formation of neoplastic osteoid and/or bone. This sarcoma is a highly heterogeneous disease with a wide range of patient outcomes. CD109 is a glycosylphosphatidylinositol-anchored glycoprotein that is highly expressed in various types of malignant tumors. We previously reported that CD109 is expressed in osteoblasts and osteoclasts in normal human tissues and plays a role in bone metabolism in vivo. While CD109 has been shown to promote various carcinomas through the downregulation of TGF-ß signaling, the role and mechanism of CD109 in sarcomas remain largely unknown. In this study, we investigated the molecular function of CD109 in sarcomas using osteosarcoma cell lines and tissue. Semi-quantitative immunohistochemical analysis using human osteosarcoma tissue revealed a significantly worse prognosis in the CD109-high group compared with the CD109-low group. We found no association between CD109 expression and TGF-ß signaling in osteosarcoma cells. However, enhancement of SMAD1/5/9 phosphorylation was observed in CD109 knockdown cells under bone morphogenetic protein-2 (BMP-2) stimulation. We also performed immunohistochemical analysis for phospho-SMAD1/5/9 using human osteosarcoma tissue and found a negative correlation between CD109 expression and SMAD1/5/9 phosphorylation. In vitro wound healing assay showed that osteosarcoma cell migration was significantly attenuated in CD109-knockdown cells compared with control cells in the presence of BMP. These results suggest that CD109 is a poor prognostic factor in osteosarcoma and affects tumor cell migration via BMP signaling.


Assuntos
Antígenos CD , Neoplasias Ósseas , Proteínas de Neoplasias , Osteossarcoma , Humanos , Antígenos CD/metabolismo , Proteínas Ligadas por GPI/metabolismo , Proteínas de Neoplasias/metabolismo , Processos Neoplásicos , Fator de Crescimento Transformador beta/metabolismo
9.
Nagoya J Med Sci ; 84(3): 484-496, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36237894

RESUMO

Cancer and fibrotic diseases are characterized by continuous inflammation, tissue wounds, and injuries. Cancer is a "wound that does not heal," and the uncontrolled proliferation of cancer cells disrupts normal tissue integrity and induces stromal fibroinflammatory reactions. Fibroblasts proliferate extensively in the stroma, playing a major role in the development of these diseases. There has been considerable evidence that fibroblasts contribute to fibrosis and tissue stiffening and promote disease progression via multiple mechanisms. However, recent emerging findings, mainly derived from single-cell transcriptomic analysis, indicated that fibroblasts are functionally heterogeneous, leading to the hypothesis that both disease-promoting and -restraining fibroblasts exist. We recently showed that a fibroblast population, defined by the expression of the glycosylphosphatidylinositol-anchored membrane protein Meflin may suppress but not promote fibrotic response and disease progression in cancer and fibrotic diseases. Although currently hypothetical, the primary function of Meflin-positive fibroblasts may be tissue repair after injury and cancer initiation occurred. This observation has led to the proposal of a potential therapy that converts the phenotype of fibroblasts from pro-tumor to anti-tumor. In this short review, we summarize our recent findings on the function of Meflin in the context of cancer and fibrotic diseases and discuss how we can utilize this knowledge on fibroblasts in translational medicine. We also discuss several aspects of the interpretation of survival analysis data, such as Kaplan-Meier analysis, to address the function of specific genes expressed in fibroblasts.


Assuntos
Glicosilfosfatidilinositóis , Neoplasias , Progressão da Doença , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Glicosilfosfatidilinositóis/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo
10.
Sci Rep ; 12(1): 5389, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35354870

RESUMO

Perivascular mesenchymal cells (PMCs), which include pericytes, give rise to myofibroblasts that contribute to chronic kidney disease progression. Several PMC markers have been identified; however, PMC heterogeneity and functions are not fully understood. Here, we describe a novel subset of renal PMCs that express Meflin, a glycosylphosphatidylinositol-anchored protein that was recently identified as a marker of fibroblasts essential for cardiac tissue repair. Tracing the lineage of Meflin+ PMCs, which are found in perivascular and periglomerular areas and exhibit renin-producing potential, showed that they detach from the vasculature and proliferate under disease conditions. Although the contribution of Meflin+ PMCs to conventional α-SMA+ myofibroblasts is low, they give rise to fibroblasts with heterogeneous α-SMA expression patterns. Genetic ablation of Meflin+ PMCs in a renal fibrosis mouse model revealed their essential role in collagen production. Consistent with this, human biopsy samples showed that progressive renal diseases exhibit high Meflin expression. Furthermore, Meflin overexpression in kidney fibroblasts promoted bone morphogenetic protein 7 signals and suppressed myofibroblastic differentiation, implicating the roles of Meflin in suppressing tissue fibrosis. These findings demonstrate that Meflin marks a PMC subset that is functionally distinct from classic pericytes and myofibroblasts, highlighting the importance of elucidating PMC heterogeneity.


Assuntos
Células-Tronco Mesenquimais , Miofibroblastos , Animais , Fibroblastos/metabolismo , Rim , Células-Tronco Mesenquimais/metabolismo , Camundongos , Miofibroblastos/metabolismo , Pericitos/metabolismo
11.
Oncogene ; 41(19): 2764-2777, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35414659

RESUMO

Previous therapeutic attempts to deplete cancer-associated fibroblasts (CAFs) or inhibit their proliferation in pancreatic ductal adenocarcinoma (PDAC) were not successful in mice or patients. Thus, CAFs may be tumor suppressive or heterogeneous, with distinct cancer-restraining and -promoting CAFs (rCAFs and pCAFs, respectively). Here, we showed that induced expression of the glycosylphosphatidylinositol-anchored protein Meflin, a rCAF-specific marker, in CAFs by genetic and pharmacological approaches improved the chemosensitivity of mouse PDAC. A chemical library screen identified Am80, a synthetic, nonnatural retinoid, as a reagent that effectively induced Meflin expression in CAFs. Am80 administration improved the sensitivity of PDAC to chemotherapeutics, accompanied by increases in tumor vessel area and intratumoral drug delivery. Mechanistically, Meflin was involved in the suppression of tissue stiffening by interacting with lysyl oxidase to inhibit its collagen crosslinking activity. These data suggested that modulation of CAF heterogeneity may represent a strategy for PDAC treatment.


Assuntos
Fibroblastos Associados a Câncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Fibroblastos Associados a Câncer/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fenótipo , Microambiente Tumoral , Neoplasias Pancreáticas
12.
iScience ; 23(6): 101213, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32535020

RESUMO

Despite common consensus about the importance of planar cell polarity (PCP) proteins in tissue orientation, little is known about the mechanisms used by PCP proteins to promote planar polarization of cytoskeletons within individual cells. One PCP protein Fzd6 asymmetrically localizes to the apical cell membrane of multi-ciliated ependymal cells lining the lateral ventricular (LV) wall on the side that contacts cerebrospinal fluid flow. Individual ependymal cells have planar polarized microtubules that connect ciliary basal bodies (BBs) with the cell cortex of the Fzd side to coordinate cilia orientation. Here, we report that cytoplasmic dynein is anchored to the cell cortex of the Fzd side via an adapter protein Daple that regulates microtubule dynamics. Asymmetric localization of cortical dynein generates a pulling force on dynamic microtubules connected to BBs, which in turn orients BBs toward the Fzd side. This is required for coordinated cilia orientation on the LV wall.

13.
Cancer Res ; 79(20): 5367-5381, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31439548

RESUMO

Cancer-associated fibroblasts (CAF) constitute a major component of the tumor microenvironment. Recent observations in genetically engineered mouse models and clinical studies have suggested that there may exist at least two functionally different populations of CAFs, that is, cancer-promoting CAFs (pCAF) and cancer-restraining CAFs (rCAF). Although various pCAF markers have been identified, the identity of rCAFs remains unknown because of the lack of rCAF-specific marker(s). In this study, we found that Meflin, a glycosylphosphatidylinositol-anchored protein that is a marker of mesenchymal stromal/stem cells and maintains their undifferentiated state, is expressed by pancreatic stellate cells that are a source of CAFs in pancreatic ductal adenocarcinoma (PDAC). In situ hybridization analysis of 71 human PDAC tissues revealed that the infiltration of Meflin-positive CAFs correlated with favorable patient outcome. Consistent herewith, Meflin deficiency led to significant tumor progression with poorly differentiated histology in a PDAC mouse model. Similarly, genetic ablation of Meflin-positive CAFs resulted in poor differentiation of tumors in a syngeneic transplantation model. Conversely, delivery of a Meflin-expressing lentivirus into the tumor stroma or overexpression of Meflin in CAFs suppressed the growth of xenograft tumors. Lineage tracing revealed that Meflin-positive cells gave rise to α-smooth muscle actin-positive CAFs that are positive or negative for Meflin, suggesting a mechanism for generating CAF heterogeneity. Meflin deficiency or low expression resulted in straightened stromal collagen fibers, which represent a signature for aggressive tumors, in mouse or human PDAC tissues, respectively. Together, the data suggest that Meflin is a marker of rCAFs that suppress PDAC progression. SIGNIFICANCE: Meflin marks and functionally contributes to a subset of cancer-associated fibroblasts that exert antitumoral effects.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5367/F1.large.jpg.


Assuntos
Carcinoma Ductal Pancreático/patologia , Fibroblastos/patologia , Imunoglobulinas/fisiologia , Neoplasias Pancreáticas/patologia , Animais , Biomarcadores Tumorais , Carcinogênese , Carcinoma Ductal Pancreático/química , Diferenciação Celular , Linhagem Celular Tumoral , Progressão da Doença , Fibroblastos/química , Regulação Neoplásica da Expressão Gênica , Genes Sintéticos , Xenoenxertos , Humanos , Imunoglobulinas/análise , Imunoglobulinas/deficiência , Imunoglobulinas/genética , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias Pancreáticas/química , Prognóstico , Proteínas Recombinantes de Fusão/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Vitamina D/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa