Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 173(5): 1135-1149.e15, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29754817

RESUMEN

A primary cause of disease progression in type 2 diabetes (T2D) is ß cell dysfunction due to inflammatory stress and insulin resistance. However, preventing ß cell exhaustion under diabetic conditions is a major therapeutic challenge. Here, we identify the vitamin D receptor (VDR) as a key modulator of inflammation and ß cell survival. Alternative recognition of an acetylated lysine in VDR by bromodomain proteins BRD7 and BRD9 directs association to PBAF and BAF chromatin remodeling complexes, respectively. Mechanistically, ligand promotes VDR association with PBAF to effect genome-wide changes in chromatin accessibility and enhancer landscape, resulting in an anti-inflammatory response. Importantly, pharmacological inhibition of BRD9 promotes PBAF-VDR association to restore ß cell function and ameliorate hyperglycemia in murine T2D models. These studies reveal an unrecognized VDR-dependent transcriptional program underpinning ß cell survival and identifies the VDR:PBAF/BAF association as a potential therapeutic target for T2D.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Receptores de Calcitriol/metabolismo , Factores de Transcripción/metabolismo , Vitamina D/farmacología , Animales , Calcitriol/análogos & derivados , Calcitriol/farmacología , Ensamble y Desensamble de Cromatina , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Humanos , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Mutagénesis Sitio-Dirigida , Fosforilación Oxidativa/efectos de los fármacos , Unión Proteica , Interferencia de ARN , ARN Guía de Kinetoplastida/genética , ARN Interferente Pequeño/metabolismo , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos
2.
Annu Rev Cell Dev Biol ; 34: 333-355, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30028641

RESUMEN

Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Inflamación/genética , Neoplasias/genética , Células Estrelladas Pancreáticas/metabolismo , Transdiferenciación Celular/genética , Células Estrelladas Hepáticas/patología , Humanos , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Neoplasias/patología , Páncreas/lesiones , Páncreas/metabolismo , Páncreas/patología , Células Estrelladas Pancreáticas/patología , Microambiente Tumoral/genética , Cicatrización de Heridas
3.
Cell ; 159(1): 80-93, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25259922

RESUMEN

The poor clinical outcome in pancreatic ductal adenocarcinoma (PDA) is attributed to intrinsic chemoresistance and a growth-permissive tumor microenvironment. Conversion of quiescent to activated pancreatic stellate cells (PSCs) drives the severe stromal reaction that characterizes PDA. Here, we reveal that the vitamin D receptor (VDR) is expressed in stroma from human pancreatic tumors and that treatment with the VDR ligand calcipotriol markedly reduced markers of inflammation and fibrosis in pancreatitis and human tumor stroma. We show that VDR acts as a master transcriptional regulator of PSCs to reprise the quiescent state, resulting in induced stromal remodeling, increased intratumoral gemcitabine, reduced tumor volume, and a 57% increase in survival compared to chemotherapy alone. This work describes a molecular strategy through which transcriptional reprogramming of tumor stroma enables chemotherapeutic response and suggests vitamin D priming as an adjunct in PDA therapy. PAPERFLICK:


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Calcitriol/análogos & derivados , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Receptores de Calcitriol/metabolismo , Adenocarcinoma/patología , Animales , Calcitriol/farmacología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neoplasias Pancreáticas/patología , Pancreatitis/tratamiento farmacológico , Pancreatitis/prevención & control , Transducción de Señal , Células del Estroma/patología
4.
Cell ; 153(3): 601-13, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23622244

RESUMEN

Liver fibrosis is a reversible wound-healing response involving TGFß1/SMAD activation of hepatic stellate cells (HSCs). It results from excessive deposition of extracellular matrix components and can lead to impairment of liver function. Here, we show that vitamin D receptor (VDR) ligands inhibit HSC activation by TGFß1 and abrogate liver fibrosis, whereas Vdr knockout mice spontaneously develop hepatic fibrosis. Mechanistically, we show that TGFß1 signaling causes a redistribution of genome-wide VDR-binding sites (VDR cistrome) in HSCs and facilitates VDR binding at SMAD3 profibrotic target genes via TGFß1-dependent chromatin remodeling. In the presence of VDR ligands, VDR binding to the coregulated genes reduces SMAD3 occupancy at these sites, inhibiting fibrosis. These results reveal an intersecting VDR/SMAD genomic circuit that regulates hepatic fibrogenesis and define a role for VDR as an endocrine checkpoint to modulate the wound-healing response in liver. Furthermore, the findings suggest VDR ligands as a potential therapy for liver fibrosis.


Asunto(s)
Redes Reguladoras de Genes , Hígado/metabolismo , Hígado/patología , Receptores de Calcitriol/metabolismo , Transducción de Señal , Animales , Calcitriol/análogos & derivados , Fibrosis/prevención & control , Estudio de Asociación del Genoma Completo , Células Estrelladas Hepáticas , Hígado/lesiones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Receptores de Calcitriol/agonistas , Proteína smad3/metabolismo , Transcriptoma , Factor de Crecimiento Transformador beta1/metabolismo
5.
Nature ; 536(7617): 479-83, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27509858

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease characterized by an intense fibrotic stromal response and deregulated metabolism. The role of the stroma in PDAC biology is complex and it has been shown to play critical roles that differ depending on the biological context. The stromal reaction also impairs the vasculature, leading to a highly hypoxic, nutrient-poor environment. As such, these tumours must alter how they capture and use nutrients to support their metabolic needs. Here we show that stroma-associated pancreatic stellate cells (PSCs) are critical for PDAC metabolism through the secretion of non-essential amino acids (NEAA). Specifically, we uncover a previously undescribed role for alanine, which outcompetes glucose and glutamine-derived carbon in PDAC to fuel the tricarboxylic acid (TCA) cycle, and thus NEAA and lipid biosynthesis. This shift in fuel source decreases the tumour's dependence on glucose and serum-derived nutrients, which are limited in the pancreatic tumour microenvironment. Moreover, we demonstrate that alanine secretion by PSCs is dependent on PSC autophagy, a process that is stimulated by cancer cells. Thus, our results demonstrate a novel metabolic interaction between PSCs and cancer cells, in which PSC-derived alanine acts as an alternative carbon source. This finding highlights a previously unappreciated metabolic network within pancreatic tumours in which diverse fuel sources are used to promote growth in an austere tumour microenvironment.


Asunto(s)
Alanina/metabolismo , Autofagia , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/citología , Células Estrelladas Pancreáticas/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Vías Biosintéticas , Carbono/metabolismo , Carcinoma Ductal Pancreático/patología , Ciclo del Ácido Cítrico , Femenino , Glucosa/metabolismo , Xenoinjertos , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias Pancreáticas/patología , Microambiente Tumoral/fisiología
6.
Proc Natl Acad Sci U S A ; 114(5): 1129-1134, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096419

RESUMEN

A fibroinflammatory stromal reaction cooperates with oncogenic signaling to influence pancreatic ductal adenocarcinoma (PDAC) initiation, progression, and therapeutic outcome, yet the mechanistic underpinning of this crosstalk remains poorly understood. Here we show that stromal cues elicit an adaptive response in the cancer cell including the rapid mobilization of a transcriptional network implicated in accelerated growth, along with anabolic changes of an altered metabolome. The close overlap of stroma-induced changes in vitro with those previously shown to be regulated by oncogenic Kras in vivo suggests that oncogenic Kras signaling-a hallmark and key driver of PDAC-is contingent on stromal inputs. Mechanistically, stroma-activated cancer cells show widespread increases in histone acetylation at transcriptionally enhanced genes, implicating the PDAC epigenome as a presumptive point of convergence between these pathways and a potential therapeutic target. Notably, inhibition of the bromodomain and extraterminal (BET) family of epigenetic readers, and of Bromodomain-containing protein 2 (BRD2) in particular, blocks stroma-inducible transcriptional regulation in vitro and tumor progression in vivo. Our work suggests the existence of a molecular "AND-gate" such that tumor activation is the consequence of mutant Kras and stromal cues, providing insight into the role of the tumor microenvironment in the origin and treatment of Ras-driven tumors.


Asunto(s)
Carcinoma Ductal Pancreático/fisiopatología , Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Código de Histonas , Metaboloma , Neoplasias Pancreáticas/fisiopatología , Células del Estroma/fisiología , Microambiente Tumoral/fisiología , Acetilación , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Citocinas/metabolismo , Metabolismo Energético , Elementos de Facilitación Genéticos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/fisiología , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Transcripción , Células Tumorales Cultivadas
7.
Mol Cell ; 39(6): 873-85, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20864035

RESUMEN

During an immune response, B cells undergo rapid proliferation and activation-induced cytidine deaminase (AID)-dependent remodeling of immunoglobulin (IG) genes within germinal centers (GCs) to generate memory B and plasma cells. Unfortunately, the genotoxic stress associated with the GC reaction also promotes most B cell malignancies. Here, we report that exogenous and intrinsic AID-induced DNA strand breaks activate ATM, which signals through an LKB1 intermediate to inactivate CRTC2, a transcriptional coactivator of CREB. Using genome-wide location analysis, we determined that CRTC2 inactivation unexpectedly represses a genetic program that controls GC B cell proliferation, self-renewal, and differentiation while opposing lymphomagenesis. Inhibition of this pathway results in increased GC B cell proliferation, reduced antibody secretion, and impaired terminal differentiation. Multiple distinct pathway disruptions were also identified in human GC B cell lymphoma patient samples. Combined, our data show that CRTC2 inactivation, via physiologic DNA damage response signaling, promotes B cell differentiation in response to genotoxic stress.


Asunto(s)
Linfocitos B/citología , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/inmunología , Citidina Desaminasa/genética , Daño del ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de la radiación , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/efectos de la radiación , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Expresión Génica/efectos de la radiación , Regulación de la Expresión Génica/inmunología , Centro Germinal/citología , Humanos , Cambio de Clase de Inmunoglobulina/fisiología , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Metformina/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
8.
Proc Natl Acad Sci U S A ; 112(51): 15713-8, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26644586

RESUMEN

Liver fibrosis is characterized by the persistent deposition of extracellular matrix components by hepatic stellate cell (HSC)-derived myofibroblasts. It is the histological manifestation of progressive, but reversible wound-healing processes. An unabated fibrotic response results in chronic liver disease and cirrhosis, a pathological precursor of hepatocellular carcinoma. We report here that JQ1, a small molecule inhibitor of bromodomain-containing protein 4 (BRD4), a member of bromodomain and extraterminal (BET) proteins, abrogate cytokine-induced activation of HSCs. Cistromic analyses reveal that BRD4 is highly enriched at enhancers associated with genes involved in multiple profibrotic pathways, where BRD4 is colocalized with profibrotic transcription factors. Furthermore, we show that JQ1 is not only protective, but can reverse the fibrotic response in carbon tetrachloride-induced fibrosis in mouse models. Our results implicate that BRD4 can act as a global genomic regulator to direct the fibrotic response through its coordinated regulation of myofibroblast transcription. This suggests BRD4 as a potential therapeutic target for patients with fibrotic complications.


Asunto(s)
Cirrosis Hepática Experimental/tratamiento farmacológico , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Azepinas/farmacología , Azepinas/uso terapéutico , Células Cultivadas , Células Estrelladas Hepáticas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Triazoles/uso terapéutico
10.
Nat Cancer ; 5(2): 283-298, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38195933

RESUMEN

Lipids and their modifying enzymes regulate diverse features of the tumor microenvironment and cancer progression. The secreted enzyme autotaxin (ATX) hydrolyzes extracellular lysophosphatidylcholine to generate the multifunctional lipid mediator lysophosphatidic acid (LPA) and supports the growth of several tumor types, including pancreatic ductal adenocarcinoma (PDAC). Here we show that ATX suppresses the accumulation of eosinophils in the PDAC microenvironment. Genetic or pharmacologic ATX inhibition increased the number of intratumor eosinophils, which promote tumor cell apoptosis locally and suppress tumor progression. Mechanistically, ATX suppresses eosinophil accumulation via an autocrine feedback loop, wherein ATX-LPA signaling negatively regulates the activity of the AP-1 transcription factor c-Jun, in turn suppressing the expression of the potent eosinophil chemoattractant CCL11 (eotaxin-1). Eosinophils were identified in human PDAC specimens, and rare individuals with high intratumor eosinophil abundance had the longest overall survival. Together with recent findings, this study reveals the context-dependent, immune-modulatory potential of ATX-LPA signaling in cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Eosinófilos/metabolismo , Quimiocina CCL11 , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Procesos Neoplásicos , Lisofosfatidilcolinas/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Microambiente Tumoral
11.
Annu Rev Pathol ; 18: 123-148, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36130070

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) features a prominent stromal microenvironment with remarkable cellular and spatial heterogeneity that meaningfully impacts disease biology and treatment resistance. Recent advances in tissue imaging capabilities, single-cell analytics, and disease modeling have shed light on organizing principles that shape the stromal complexity of PDAC tumors. These insights into the functional and spatial dependencies that coordinate cancer cell biology and the relationships that exist between cells and extracellular matrix components present in tumors are expected to unveil therapeutic vulnerabilities. We review recent advances in the field and discuss current understandings of mechanisms by which the tumor microenvironment shapes PDAC pathogenesis and therapy resistance.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Resistencia a Antineoplásicos , Microambiente Tumoral , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología
12.
Artículo en Inglés | MEDLINE | ID: mdl-37696660

RESUMEN

The altered metabolism of tumor cells is a well-known hallmark of cancer and is driven by multiple factors such as mutations in oncogenes and tumor suppressor genes, the origin of the tissue where the tumor arises, and the microenvironment of the tumor. These metabolic changes support the growth of cancer cells by providing energy and the necessary building blocks to sustain proliferation. Targeting these metabolic alterations therapeutically is a potential strategy to treat cancer, but it is challenging due to the metabolic plasticity of tumors. Cancer cells have developed ways to scavenge nutrients through autophagy and macropinocytosis and can also form metabolic networks with stromal cells in the tumor microenvironment. Understanding the role of the tumor microenvironment in tumor metabolism is crucial for effective therapeutic targeting. This review will discuss tumor metabolism and the contribution of the stroma in supporting tumor growth through metabolic interactions.

13.
Onco Targets Ther ; 16: 695-702, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37635751

RESUMEN

GOT2 is at the nexus of several critical metabolic pathways in homeostatic cellular and dysregulated cancer metabolism. Despite this, recent work has emphasized the remarkable plasticity of cancer cells to employ compensatory pathways when GOT2 is inhibited. Here, we review the metabolic roles of GOT2, highlighting findings in both normal and cancer cells. We emphasize how cancer cells repurpose cell intrinsic metabolism and their flexibility when GOT2 is inhibited. We close by using this framework to discuss key considerations for future investigations into cancer metabolism.

14.
bioRxiv ; 2023 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-36993424

RESUMEN

Accurately identifying phenotype-relevant cell subsets from heterogeneous cell populations is crucial for delineating the underlying mechanisms driving biological or clinical phenotypes. Here, by deploying a learning with rejection strategy, we developed a novel supervised learning framework called PENCIL to identify subpopulations associated with categorical or continuous phenotypes from single-cell data. By embedding a feature selection function into this flexible framework, for the first time, we were able to select informative features and identify cell subpopulations simultaneously, which enables the accurate identification of phenotypic subpopulations otherwise missed by methods incapable of concurrent gene selection. Furthermore, the regression mode of PENCIL presents a novel ability for supervised phenotypic trajectory learning of subpopulations from single-cell data. We conducted comprehensive simulations to evaluate PENCILs versatility in simultaneous gene selection, subpopulation identification and phenotypic trajectory prediction. PENCIL is fast and scalable to analyze 1 million cells within 1 hour. Using the classification mode, PENCIL detected T-cell subpopulations associated with melanoma immunotherapy outcomes. Moreover, when applied to scRNA-seq of a mantle cell lymphoma patient with drug treatment across multiple time points, the regression mode of PENCIL revealed a transcriptional treatment response trajectory. Collectively, our work introduces a scalable and flexible infrastructure to accurately identify phenotype-associated subpopulations from single-cell data.

15.
Nat Commun ; 14(1): 797, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36781852

RESUMEN

The tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC) is a complex ecosystem that drives tumor progression; however, in-depth single cell characterization of the PDAC TME and its role in response to therapy is lacking. Here, we perform single-cell RNA sequencing on freshly collected human PDAC samples either before or after chemotherapy. Overall, we find a heterogeneous mixture of basal and classical cancer cell subtypes, along with distinct cancer-associated fibroblast and macrophage subpopulations. Strikingly, classical and basal-like cancer cells exhibit similar transcriptional responses to chemotherapy and do not demonstrate a shift towards a basal-like transcriptional program among treated samples. We observe decreased ligand-receptor interactions in treated samples, particularly between TIGIT on CD8 + T cells and its receptor on cancer cells, and identify TIGIT as the major inhibitory checkpoint molecule of CD8 + T cells. Our results suggest that chemotherapy profoundly impacts the PDAC TME and may promote resistance to immunotherapy.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Microambiente Tumoral/genética , Ecosistema , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Análisis de Secuencia de ARN , Neoplasias Pancreáticas
16.
Proc Natl Acad Sci U S A ; 106(32): 13433-8, 2009 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-19666576

RESUMEN

Epigenetic alterations, including gain or loss of DNA methylation, are a hallmark of nearly every malignancy. Changes in DNA methylation can impact expression of cancer-related genes including apoptosis regulators and tumor suppressors. Because such epigenetic changes are reversible, they are being aggressively investigated as potential therapeutic targets. Here we use the Emu-TCL1 transgenic mouse model of chronic lymphocytic leukemia (CLL) to determine the timing and patterns of aberrant DNA methylation, and to investigate the mechanisms that lead to aberrant DNA methylation. We show that CLL cells from Emu-TCL1 mice at various stages recapitulate epigenetic alterations seen in human CLL. Aberrant methylation of promoter sequences is observed as early as 3 months of age in these animals, well before disease onset. Abnormally methylated promoter regions include binding sites for the transcription factor FOXD3. We show that loss of Foxd3 expression due to an NF-kappaB p50/p50:HDAC1 repressor complex occurs in TCL1-positive B cells before methylation. Therefore, specific transcriptional repression is an early event leading to epigenetic silencing of target genes in murine and human CLL. These results provide strong rationale for the development of strategies to target NF-kappaB components in CLL and potentially other B-cell malignancies.


Asunto(s)
Epigénesis Genética , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Animales , Metilación de ADN , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factores de Transcripción Forkhead/metabolismo , Regulación Leucémica de la Expresión Génica , Silenciador del Gen , Histona Desacetilasa 1 , Histona Desacetilasas/metabolismo , Humanos , Ratones , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo
17.
Cancer Discov ; 12(11): 2501-2503, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36321307

RESUMEN

Cancer-associated fibroblasts share defined features with fibroblasts in secondary lymphoid organs, including the ability to regulate immune cell localization. In this issue of Cancer Discovery, Grout and colleagues perform multimodal analysis of human lung cancer specimens and identify two distinct fibroblast populations associated with spatial exclusion of T cells from tumor cell nests. See related article by Grout et al., p. 2606 (3).


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Pulmonares , Humanos , Fibroblastos Asociados al Cáncer/patología , Linfocitos T/patología , Neoplasias Pulmonares/patología , Pulmón/patología , Fibroblastos/patología
18.
Cancer Discov ; 12(10): 2414-2433, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-35894778

RESUMEN

Despite significant recent advances in precision medicine, pancreatic ductal adenocarcinoma (PDAC) remains near uniformly lethal. Although immune-modulatory therapies hold promise to meaningfully improve outcomes for patients with PDAC, the development of such therapies requires an improved understanding of the immune evasion mechanisms that characterize the PDAC microenvironment. Here, we show that cancer cell-intrinsic glutamic-oxaloacetic transaminase 2 (GOT2) shapes the immune microenvironment to suppress antitumor immunity. Mechanistically, we find that GOT2 functions beyond its established role in the malate-aspartate shuttle and promotes the transcriptional activity of nuclear receptor peroxisome proliferator-activated receptor delta (PPARδ), facilitated by direct fatty acid binding. Although GOT2 is dispensable for cancer cell proliferation in vivo, the GOT2-PPARδ axis promotes spatial restriction of both CD4+ and CD8+ T cells from the tumor microenvironment. Our results demonstrate a noncanonical function for an established mitochondrial enzyme in transcriptional regulation of immune evasion, which may be exploitable to promote a productive antitumor immune response. SIGNIFICANCE: Prior studies demonstrate the important moonlighting functions of metabolic enzymes in cancer. We find that the mitochondrial transaminase GOT2 binds directly to fatty acid ligands that regulate the nuclear receptor PPARδ, and this functional interaction critically regulates the immune microenvironment of pancreatic cancer to promote tumor progression. See related commentary by Nwosu and di Magliano, p. 2237.. This article is highlighted in the In This Issue feature, p. 2221.


Asunto(s)
Carcinoma Ductal Pancreático , PPAR delta , Neoplasias Pancreáticas , Aspartato Aminotransferasas , Ácido Aspártico/metabolismo , Carcinoma Ductal Pancreático/patología , Ácidos Grasos , Humanos , Ligandos , Malatos/metabolismo , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Neoplasias Pancreáticas
19.
Cancer Discov ; 12(2): 484-501, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34548310

RESUMEN

Cancer-associated fibroblast (CAF) heterogeneity is increasingly appreciated, but the origins and functions of distinct CAF subtypes remain poorly understood. The abundant and transcriptionally diverse CAF population in pancreatic ductal adenocarcinoma (PDAC) is thought to arise from a common cell of origin, pancreatic stellate cells (PSC), with diversification resulting from cytokine and growth factor gradients within the tumor microenvironment. Here we analyzed the differentiation and function of PSCs during tumor progression in vivo. Contrary to expectations, we found that PSCs give rise to a numerically minor subset of PDAC CAFs. Targeted ablation of PSC-derived CAFs within their host tissue revealed nonredundant functions for this defined CAF population in shaping the PDAC microenvironment, including production of specific extracellular matrix components and tissue stiffness regulation. Together, these findings link stromal evolution from distinct cells of origin to transcriptional heterogeneity among PDAC CAFs and demonstrate unique functions for CAFs of a defined cellular origin. SIGNIFICANCE: By tracking and ablating a specific CAF population, we find that a numerically minor CAF subtype from a defined cell of origin plays unique roles in establishing the pancreatic tumor microenvironment. Together with prior studies, this work suggests that mesenchymal lineage heterogeneity and signaling gradients diversify PDAC CAFs.See related commentary by Cukierman, p. 296.This article is highlighted in the In This Issue feature, p. 275.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Neoplasias Pancreáticas/genética , Animales , Femenino , Humanos , Masculino , Ratones , Neoplasias Pancreáticas/patología
20.
Cancer Discov ; 11(2): 230-232, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531426

RESUMEN

Cancer-associated fibroblasts conduct an aberrant wound-healing response, including mechanisms that restrain and others that support tumor progression. In this issue of Cancer Discovery, Francescone and colleagues demonstrate expression of presynaptic protein NetG1 on fibroblasts in pancreatic ductal adenocarcinoma and characterize tumor-supportive functions of NetG1 in this context, including metabolic and immune-modulatory mechanisms.See related article by Francescone et al., p. 446.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Fibroblastos , Humanos , Páncreas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA