Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 13(2)2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38247798

RESUMEN

Driver mutations are considered the cornerstone of cancer initiation. They are defined as mutations that convey a competitive fitness advantage, and hence, their mutation frequency in premalignant tissue is expected to exceed the basal mutation rate. In old terms, that translates to "the survival of the fittest" and implies that a selective process underlies the frequency of cancer driver mutations. In that sense, each tissue is its own niche that creates a molecular selective pressure that may favor the propagation of a mutation or not. At the heart of this stands one of the biggest riddles in cancer biology: the tissue-predisposition to cancer driver mutations. The frequency of cancer driver mutations among tissues is non-uniform: for instance, mutations in APC are particularly frequent in colorectal cancer, and 99% of chronic myeloid leukemia patients harbor the driver BCR-ABL1 fusion mutation, which is rarely found in solid tumors. Here, we provide a mechanistic framework that aims to explain how tissue-specific features, ranging from epigenetic underpinnings to the expression of viral transposable elements, establish a molecular basis for selecting cancer driver mutations in a tissue-specific manner.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Lesiones Precancerosas , Humanos , Susceptibilidad a Enfermedades , Mutación/genética , Tasa de Mutación
2.
Blood Adv ; 8(3): 766-779, 2024 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-38147624

RESUMEN

ABSTRACT: It is still not fully understood how genetic haploinsufficiency in del(5q) myelodysplastic syndrome (MDS) contributes to malignant transformation of hematopoietic stem cells. We asked how compound haploinsufficiency for Csnk1a1 and Egr1 in the common deleted region on chromosome 5 affects hematopoietic stem cells. Additionally, Trp53 was disrupted as the most frequently comutated gene in del(5q) MDS using CRISPR/Cas9 editing in hematopoietic progenitors of wild-type (WT), Csnk1a1-/+, Egr1-/+, Csnk1a1/Egr1-/+ mice. A transplantable acute leukemia only developed in the Csnk1a1-/+Trp53-edited recipient. Isolated blasts were indefinitely cultured ex vivo and gave rise to leukemia after transplantation, providing a tool to study disease mechanisms or perform drug screenings. In a small-scale drug screening, the collaborative effect of Csnk1a1 haploinsufficiency and Trp53 sensitized blasts to the CSNK1 inhibitor A51 relative to WT or Csnk1a1 haploinsufficient cells. In vivo, A51 treatment significantly reduced blast counts in Csnk1a1 haploinsufficient/Trp53 acute leukemias and restored hematopoiesis in the bone marrow. Transcriptomics on blasts and their normal counterparts showed that the derived leukemia was driven by MAPK and Myc upregulation downstream of Csnk1a1 haploinsufficiency cooperating with a downregulated p53 axis. A collaborative effect of Csnk1a1 haploinsufficiency and p53 loss on MAPK and Myc upregulation was confirmed on the protein level. Downregulation of Myc protein expression correlated with efficient elimination of blasts in A51 treatment. The "Myc signature" closely resembled the transcriptional profile of patients with del(5q) MDS with TP53 mutation.


Asunto(s)
Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Animales , Humanos , Ratones , Médula Ósea/metabolismo , Deleción Cromosómica , Haploinsuficiencia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Eur J Immunol ; 52(1): 34-43, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34648664

RESUMEN

Acute myeloid leukemia (AML) is a highly aggressive disease with high relapse and mortality rates. Recent years have shown a surge in novel therapeutic development for AML, both in clinical and preclinical stages. These developments include targeted therapies based on AML-specific molecular signatures as well as more general immune modulation and vaccination studies. In this review, we will explore the evolving arena of AML therapy and suggest some intriguing connections between immune system modulation and targeted therapy. Improved understanding of the immune system involvement in various stages of the disease and the crosstalk between immune effectors, targeted therapy, and AML cells can provide a better framework for designing the next generation of AML therapies.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Terapia Molecular Dirigida , Humanos
4.
Trends Cell Biol ; 32(1): 8-17, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34373150

RESUMEN

Somatic mutations have traditionally been associated with cancer, yet more recently, it was realized that they also appear in nontransformed cells beginning in early life. Remarkably, some of these mutations, commonly viewed as cancer driver mutations, are widely spread among cells of noncancerous tissues, sometimes affecting the majority of the tissue cells. This spreading process intensifies upon aging or exposure to extrinsic insults, such as UV irradiation, inhaling smoke, and inflammatory cues. Whereas classic driver mutations in normal cells are mostly viewed as a first step in the carcinogenesis process, here, we speculate that in certain states, they can play beneficial homeostatic roles while confronting stress and aging tissue repair.


Asunto(s)
Neoplasias , Envejecimiento/genética , Carcinogénesis/genética , Hematopoyesis , Humanos , Mutación/genética , Neoplasias/genética
5.
Nature ; 586(7827): 133-138, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32728212

RESUMEN

Somatic mutations in p53, which inactivate the tumour-suppressor function of p53 and often confer oncogenic gain-of-function properties, are very common in cancer1,2. Here we studied the effects of hotspot gain-of-function mutations in Trp53 (the gene that encodes p53 in mice) in mouse models of WNT-driven intestinal cancer caused by Csnk1a1 deletion3,4 or ApcMin mutation5. Cancer in these models is known to be facilitated by loss of p533,6. We found that mutant versions of p53 had contrasting effects in different segments of the gut: in the distal gut, mutant p53 had the expected oncogenic effect; however, in the proximal gut and in tumour organoids it had a pronounced tumour-suppressive effect. In the tumour-suppressive mode, mutant p53 eliminated dysplasia and tumorigenesis in Csnk1a1-deficient and ApcMin/+ mice, and promoted normal growth and differentiation of tumour organoids derived from these mice. In these settings, mutant p53 was more effective than wild-type p53 at inhibiting tumour formation. Mechanistically, the tumour-suppressive effects of mutant p53 were driven by disruption of the WNT pathway, through preventing the binding of TCF4 to chromatin. Notably, this tumour-suppressive effect was completely abolished by the gut microbiome. Moreover, a single metabolite derived from the gut microbiota-gallic acid-could reproduce the entire effect of the microbiome. Supplementing gut-sterilized p53-mutant mice and p53-mutant organoids with gallic acid reinstated the TCF4-chromatin interaction and the hyperactivation of WNT, thus conferring a malignant phenotype to the organoids and throughout the gut. Our study demonstrates the substantial plasticity of a cancer mutation and highlights the role of the microenvironment in determining its functional outcome.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Microbioma Gastrointestinal/genética , Genes Supresores de Tumor , Mutación , Oncogenes/genética , Proteína p53 Supresora de Tumor/genética , Animales , Antibacterianos/farmacología , Carcinogénesis/efectos de los fármacos , Femenino , Ácido Gálico/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Masculino , Ratones , Organoides/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
6.
Prostate ; 79(4): 403-413, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30488478

RESUMEN

The substantial availability of hypoxia-inducible factor 1 (HIF-1) for pathophysiological states, such as malignancies and ischemia, is primarily regulated post-translationally through the ubiquitin proteolytic system. The balance between degradation and stabilization of HIF-1α protein is determined by specific E3 ligases. In our search for new E3 ligases that might affect HIF-1α protein expression, we studied the effects of beta-transducin repeat-containing protein (ß-TrCP) on the hypoxic pathway in cancer cells. ß-TrCP is overexpressed in many tumors and regulates various cellular processes through mediating the degradation of important targets. Unexpectedly, we found that ß-TrCP overexpression increases HIF-1α protein expression level as well as HIF-1 transcriptional activity by stabilizing HIF-1α protein and preventing its ubiquitination and proteasomal degradation in prostate cancer cells. By using a proteomic approach, we succeeded in demonstrating that ß-TrCP interferes with the association between HIF-1α and HSP70/CHIP, a HIF-1α established E3 ligase complex. Whereas the E3 ligase activity of ß-TrCP is well known, antagonizing another E3 ligase is a new mechanism of action of this important E3. We suggest that destroying or suppressing ß-TrCP and thereby interrupting the HIF-1 pathway, could be valuable antitumor therapy.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias de la Próstata/metabolismo , Regulación hacia Arriba/fisiología , Proteínas con Repetición de beta-Transducina/fisiología , Línea Celular Tumoral , Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Fosfotreonina/metabolismo , Proteómica , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/farmacología
7.
Cell ; 175(1): 171-185.e25, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30146162

RESUMEN

CKIα ablation induces p53 activation, and CKIα degradation underlies the therapeutic effect of lenalidomide in a pre-leukemia syndrome. Here we describe the development of CKIα inhibitors, which co-target the transcriptional kinases CDK7 and CDK9, thereby augmenting CKIα-induced p53 activation and its anti-leukemic activity. Oncogene-driving super-enhancers (SEs) are highly sensitive to CDK7/9 inhibition. We identified multiple newly gained SEs in primary mouse acute myeloid leukemia (AML) cells and demonstrate that the inhibitors abolish many SEs and preferentially suppress the transcription elongation of SE-driven oncogenes. We show that blocking CKIα together with CDK7 and/or CDK9 synergistically stabilize p53, deprive leukemia cells of survival and proliferation-maintaining SE-driven oncogenes, and induce apoptosis. Leukemia progenitors are selectively eliminated by the inhibitors, explaining their therapeutic efficacy with preserved hematopoiesis and leukemia cure potential; they eradicate leukemia in MLL-AF9 and Tet2-/-;Flt3ITD AML mouse models and in several patient-derived AML xenograft models, supporting their potential efficacy in curing human leukemia.


Asunto(s)
Caseína Quinasa Ialfa/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Caseína Quinasa Ialfa/fisiología , Proliferación Celular/efectos de los fármacos , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/fisiología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/fisiología , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos/genética , Hematopoyesis , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
EMBO J ; 36(20): 3046-3061, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28963394

RESUMEN

The intestinal epithelium holds an immense regenerative capacity mobilized by intestinal stem cells (ISCs), much of it supported by Wnt pathway activation. Several unique regulatory mechanisms ensuring optimal levels of Wnt signaling have been recognized in ISCs. Here, we identify another Wnt signaling amplifier, CKIε, which is specifically upregulated in ISCs and is essential for ISC maintenance, especially in the absence of its close isoform CKIδ. Co-ablation of CKIδ/ε in the mouse gut epithelium results in rapid ISC elimination, with subsequent growth arrest, crypt-villous shrinking, and rapid mouse death. Unexpectedly, Wnt activation is preserved in all CKIδ/ε-deficient enterocyte populations, with the exception of Lgr5+ ISCs, which exhibit Dvl2-dependent Wnt signaling attenuation. CKIδ/ε-depleted gut organoids cease proliferating and die rapidly, yet survive and resume self-renewal upon reconstitution of Dvl2 expression. Our study underscores a unique regulation mode of the Wnt pathway in ISCs, possibly providing new means of stem cell enrichment for regenerative medicine.


Asunto(s)
Caseína Cinasa 1 épsilon/metabolismo , Quinasa Idelta de la Caseína/metabolismo , Mucosa Intestinal/fisiología , Células Madre/fisiología , Vía de Señalización Wnt , Animales , Proliferación Celular , Células Epiteliales/fisiología , Ratones
9.
Proc Natl Acad Sci U S A ; 114(38): E8035-E8044, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28878021

RESUMEN

Casein kinase 1α (CK1α), a component of the ß-catenin destruction complex, is a critical regulator of Wnt signaling; its ablation induces both Wnt and p53 activation. To characterize the role of CK1α (encoded by Csnk1a1) in skin physiology, we crossed mice harboring floxed Csnk1a1 with mice expressing K14-Cre-ERT2 to generate mice in which tamoxifen induces the deletion of Csnk1a1 exclusively in keratinocytes [single-knockout (SKO) mice]. As expected, CK1α loss was accompanied by ß-catenin and p53 stabilization, with the preferential induction of p53 target genes, but phenotypically most striking was hyperpigmentation of the skin, importantly without tumorigenesis, for at least 9 mo after Csnk1a1 ablation. The number of epidermal melanocytes and eumelanin levels were dramatically increased in SKO mice. To clarify the putative role of p53 in epidermal hyperpigmentation, we established K14-Cre-ERT2 CK1α/p53 double-knockout (DKO) mice and found that coablation failed to induce epidermal hyperpigmentation, demonstrating that it was p53-dependent. Transcriptome analysis of the epidermis revealed p53-dependent up-regulation of Kit ligand (KitL). SKO mice treated with ACK2 (a Kit-neutralizing antibody) or imatinib (a Kit inhibitor) abrogated the CK1α ablation-induced hyperpigmentation, demonstrating that it requires the KitL/Kit pathway. Pro-opiomelanocortin (POMC), a precursor of α-melanocyte-stimulating hormone (α-MSH), was not activated in the CK1α ablation-induced hyperpigmentation, which is in contrast to the mechanism of p53-dependent UV tanning. Nevertheless, acute sunburn effects were successfully prevented in the hyperpigmented skin of SKO mice. CK1α inhibition induces skin-protective eumelanin but no carcinogenic pheomelanin and may therefore constitute an effective strategy for safely increasing eumelanin via UV-independent pathways, protecting against acute sunburn.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Queratinocitos/metabolismo , Pigmentación de la Piel , Quemadura Solar/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Quinasa de la Caseína I/antagonistas & inhibidores , Quinasa de la Caseína I/genética , Epidermis/metabolismo , Epidermis/patología , Queratinocitos/patología , Melaninas/biosíntesis , Melaninas/genética , Melanocitos/metabolismo , Melanocitos/patología , Ratones , Ratones Noqueados , Quemadura Solar/genética , Quemadura Solar/patología , Proteína p53 Supresora de Tumor/genética , beta Catenina/genética , beta Catenina/metabolismo
10.
Cancer Res ; 77(14): 3740-3744, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28667073

RESUMEN

Parainflammation is a unique variant of inflammation, characterized by epithelial-autonomous activation of inflammatory response. Parainflammation has been shown to strongly promote mouse gut tumorigenesis upon p53 loss. In a recent study, we explored the prevalence of parainflammation in human cancer and determined its relationship to certain molecular and clinical parameters affecting treatment and prognosis. Parainflammation can be identified from a 40-gene signature and is found in both carcinoma cell lines and a variety of primary tumors, independently of tumor microenvironment. Here, we discuss the implications of our findings in analyses of tumor microenvironment, suggesting that as tumor cell gene expression may often mimic immune and inflammatory infiltration, caution should be applied when interpreting tumor expression data. We also address the connection between parainflammation and prevalence of p53 mutations in specific types of tumors, and cancer prevention by regular usage of NSAIDs. We suggest that parainflammation may serve as a novel biomarker for screening patients who may particularly benefit from NSAID treatment. Cancer Res; 77(14); 3740-4. ©2017 AACR.


Asunto(s)
Carcinoma/inmunología , Carcinoma/patología , Inflamación/inmunología , Inflamación/patología , Animales , Carcinogénesis/inmunología , Carcinogénesis/patología , Humanos , Ratones , Microambiente Tumoral/inmunología
11.
Genome Biol ; 17(1): 145, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27386949

RESUMEN

BACKGROUND: Chronic inflammation has been recognized as one of the hallmarks of cancer. We recently showed that parainflammation, a unique variant of inflammation between homeostasis and chronic inflammation, strongly promotes mouse gut tumorigenesis upon p53 loss. Here we explore the prevalence of parainflammation in human cancer and determine its relationship to certain molecular and clinical parameters affecting treatment and prognosis. RESULTS: We generated a transcriptome signature to identify parainflammation in many primary human tumors and carcinoma cell lines as distinct from their normal tissue counterparts and the tumor microenvironment and show that parainflammation-positive tumors are enriched for p53 mutations and associated with poor prognosis. Non-steroidal anti-inflammatory drug (NSAID) treatment suppresses parainflammation in both murine and human cancers, possibly explaining a protective effect of NSAIDs against cancer. CONCLUSIONS: We conclude that parainflammation, a low-grade form of inflammation, is widely prevalent in human cancer, particularly in cancer types commonly harboring p53 mutations. Our data suggest that parainflammation may be a driver for p53 mutagenesis and a guide for cancer prevention by NSAID treatment.


Asunto(s)
Carcinogénesis/genética , Inflamación/genética , Neoplasias/genética , Transcriptoma/genética , Proteína p53 Supresora de Tumor/genética , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Genoma Humano , Humanos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Ratones , Mutación , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico
12.
Oncotarget ; 7(33): 52643-52660, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27462916

RESUMEN

SV40 large T-antigen (T-ag) has been known for decades to inactivate the tumor suppressor p53 by sequestration and additional mechanisms. Our present study revealed that the struggle between p53 and T-ag begins very early in the infection cycle. We found that p53 is activated early after SV40 infection and defends the host against the infection. Using live cell imaging and single cell analyses we found that p53 dynamics are variable among individual cells, with only a subset of cells activating p53 immediately after SV40 infection. This cell-to-cell variabilty had clear consequences on the outcome of the infection. None of the cells with elevated p53 at the beginning of the infection proceeded to express T-ag, suggesting a p53-dependent decision between abortive and productive infection. In addition, we show that artificial elevation of p53 levels prior to the infection reduces infection efficiency, supporting a role for p53 in defending against SV40. We further found that the p53-mediated host defense mechanism against SV40 is not facilitated by apoptosis nor via interferon-stimulated genes. Instead p53 binds to the viral DNA at the T-ag promoter region, prevents its transcriptional activation by Sp1, and halts the progress of the infection. These findings shed new light on the long studied struggle between SV40 T-ag and p53, as developed during virus-host coevolution. Our studies indicate that the fate of SV40 infection is determined as soon as the viral DNA enters the nucleus, before the onset of viral gene expression.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Regulación Viral de la Expresión Génica , Virus 40 de los Simios/genética , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/virología , Línea Celular , Regulación Neoplásica de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Células MCF-7 , Microscopía Confocal , Regiones Promotoras Genéticas/genética , Unión Proteica , Virus 40 de los Simios/fisiología , Factor de Transcripción Sp1/metabolismo , Imagen de Lapso de Tiempo/métodos , Proteína p53 Supresora de Tumor/metabolismo
13.
Nat Immunol ; 17(3): 230-40, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26882261

RESUMEN

Inflammation is emerging as one of the hallmarks of cancer, yet its role in most tumors remains unclear. Whereas a minority of solid tumors are associated with overt inflammation, long-term treatment with non-steroidal anti-inflammatory drugs is remarkably effective in reducing cancer rate and death. This indicates that inflammation might have many as-yet-unrecognized facets, among which an indolent course might be far more prevalent than previously appreciated. In this Review, we explore the various inflammatory processes underlying the development and progression of colorectal cancer and discuss anti-inflammatory means for its prevention and treatment.


Asunto(s)
Adenocarcinoma/inmunología , Adenoma/inmunología , Carcinogénesis/inmunología , Neoplasias Colorrectales/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inflamación , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/prevención & control , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/prevención & control , Daño del ADN/inmunología , Progresión de la Enfermedad , Humanos , Inmunidad Innata/inmunología , Inmunidad Mucosa/inmunología , Interleucina-1beta/antagonistas & inhibidores , Quinasas Janus/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores de Interleucina-6/antagonistas & inhibidores , Factores de Transcripción STAT/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
14.
Mol Cell Biol ; 36(7): 1124-35, 2016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26811327

RESUMEN

Wnt pathway-driven proliferation and renewal of the intestinal epithelium must be tightly controlled to prevent development of cancer and barrier dysfunction. Although type I interferons (IFN) produced in the gut under the influence of microbiota are known for their antiproliferative effects, the role of these cytokines in regulating intestinal epithelial cell renewal is largely unknown. Here we report a novel role for IFN in the context of intestinal knockout of casein kinase 1α (CK1α), which controls the ubiquitination and degradation of both ß-catenin and the IFNAR1 chain of the IFN receptor. Ablation of CK1α leads to the activation of both ß-catenin and IFN pathways and prevents the unlimited proliferation of intestinal epithelial cells despite constitutive ß-catenin activity. IFN signaling contributes to the activation of the p53 pathway and the appearance of apoptotic and senescence markers in the CK1α-deficient gut. Concurrent genetic ablation of CK1α and IFNAR1 leads to intestinal hyperplasia, robust attenuation of apoptosis, and rapid and lethal loss of barrier function. These data indicate that IFN play an important role in controlling the proliferation and function of the intestinal epithelium in the context of ß-catenin activation.


Asunto(s)
Interferón Tipo I/fisiología , Mucosa Intestinal/citología , Receptor de Interferón alfa y beta/metabolismo , Animales , Apoptosis , Caseína Quinasa Ialfa/genética , Caseína Quinasa Ialfa/metabolismo , Proliferación Celular , Daño del ADN , Mucosa Intestinal/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Ubiquitinación
15.
Nat Immunol ; 16(12): 1235-44, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26502405

RESUMEN

Ectopic lymphoid-like structures (ELSs) are often observed in cancer, yet their function is obscure. Although ELSs signify good prognosis in certain malignancies, we found that hepatic ELSs indicated poor prognosis for hepatocellular carcinoma (HCC). We studied an HCC mouse model that displayed abundant ELSs and found that they constituted immunopathological microniches wherein malignant hepatocyte progenitor cells appeared and thrived in a complex cellular and cytokine milieu until gaining self-sufficiency. The egress of progenitor cells and tumor formation were associated with the autocrine production of cytokines previously provided by the niche. ELSs developed via cooperation between the innate immune system and adaptive immune system, an event facilitated by activation of the transcription factor NF-κB and abolished by depletion of T cells. Such aberrant immunological foci might represent new targets for cancer therapy.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Tejido Linfoide/inmunología , Células Madre Neoplásicas/inmunología , Nicho de Células Madre/inmunología , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Hibridación Genómica Comparativa , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Quinasa I-kappa B/metabolismo , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Immunoblotting , Hibridación in Situ , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Tejido Linfoide/metabolismo , Tejido Linfoide/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Células Madre Neoplásicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nicho de Células Madre/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transcriptoma/genética , Transcriptoma/inmunología
16.
Cancer Res ; 75(15): 3010-9, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26069251

RESUMEN

The microRNA (miRNA) landscape changes during the progression of cancer. We defined a metastasis-associated miRNA landscape using a systematic approach. We profiled and validated miRNA and mRNA expression in a unique series of human colorectal metastasis tissues together with their matched primary tumors and corresponding normal tissues. We identified an exclusive miRNA signature that is differentially expressed in metastases. Three of these miRNAs were identified as key drivers of an EMT-regulating network acting though a number of novel targets. These targets include SIAH1, SETD2, ZEB2, and especially FOXN3, which we demonstrated for the first time as a direct transcriptional suppressor of N-cadherin. The modulation of N-cadherin expression had significant impact on migration, invasion, and metastasis in two different in vivo models. The significant deregulation of the miRNAs defining the network was confirmed in an independent patient set as well as in a database of diverse malignancies derived from more than 6,000 patients. Our data define a novel metastasis-orchestrating network based on systematic hypothesis generation from metastasis tissues.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/secundario , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Animales , Antígenos CD/genética , Cadherinas/genética , Proteínas de Ciclo Celular/genética , Bases de Datos Factuales , Transición Epitelial-Mesenquimal/genética , Factores de Transcripción Forkhead , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Homeodominio/genética , Humanos , Ratones Desnudos , Metástasis de la Neoplasia/genética , Proteínas Nucleares/genética , Valores de Referencia , Proteínas Represoras/genética , Reproducibilidad de los Resultados , Ubiquitina-Proteína Ligasas/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
17.
Trends Immunol ; 36(4): 217-28, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25801910

RESUMEN

Senescent cells, albeit not proliferating, are metabolically and transcriptionally active, thereby capable of affecting their microenvironment, notably via the production of inflammatory mediators. These mediators maintain and propagate the senescence process to neighboring cells, and then recruit immune cells for clearing senescent cells. Among the inflammatory cues are molecules with pronounced tumor-controlling properties, both growth and invasion factors and inhibitory factors, working directly or via recruited immune cells. These senescence-inflammatory effects also prevail within tumors, mediated by the senescent tumor cells and the senescent tumor stroma. Here, we review the course and impact of senescence-associated inflammatory responses in aging and cancer. We propose that controlling senescence-associated inflammation by targeting specific inflammatory mediators may have a beneficial therapeutic effect in treatment of cancer and aging-related diseases.


Asunto(s)
Envejecimiento/inmunología , Neoplasias/inmunología , Humanos , Inflamación/inmunología , Neoplasias/patología
18.
Mol Cell Oncol ; 2(1): e968028, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27308384

RESUMEN

In contrast to common genomic amplifications that support cancer cell growth by rewiring intracellular signaling, VEGFA amplification drives tumor cell proliferation via the tumor microenvironment. VEGFA amplification is present in a subset of mouse and human hepatocellular carcinomas (HCCs) that appear to be particularly sensitive to sorafenib treatment, indicating its potential value as a biomarker for HCC treatment.

19.
Proc Natl Acad Sci U S A ; 111(49): 17582-7, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422452

RESUMEN

The inflamed tumor microenvironment plays a critical role in tumorigenesis. However, the mechanisms through which immune cells, particularly macrophages, promote tumorigenesis have only been partially elucidated, and the full scope of signaling pathways supplying macrophages with protumorigenic phenotypes still remain largely unknown. Here we report that germ-line absence of c-Jun N-terminal phosphorylation at serines 63 and 73 impedes inflammation-associated hepatocarcinogenesis, yet deleting c-Jun only in hepatocytes does not inhibit hepatocellular carcinoma (HCC) formation. Moreover, in human HCC-bearing livers, c-Jun phosphorylation is found in inflammatory cells, whereas it is mostly absent from malignant hepatocytes. Interestingly, macrophages in livers of mice with chronic hepatitis gradually switch their phenotype along the course of disease. Macrophage phenotype and density are dictated by c-Jun phosphorylation, in vitro and in vivo. Transition of macrophage phenotype, from antitumorigenic to protumorigenic, occurs before tumorigenesis, resulting in the production of various chemokines, including chemokine (C-C motif) ligand 17 (CCL17) and CCL22. Such signals, emanating from the liver microenvironment, direct the recruitment of regulatory T cells, which are known to facilitate HCC growth. Our findings identify c-Jun phosphorylation as a key mediator of macrophage education and point to the recruitment of immunosuppressive regulatory T cells as a possible protumorigenic mechanism.


Asunto(s)
Macrófagos/citología , Macrófagos/inmunología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Apoptosis , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Quimiocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Hepatitis/metabolismo , Hepatocitos/citología , Humanos , Inmunidad Innata , Inflamación , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Fosforilación , Pronóstico , Estructura Terciaria de Proteína , Microambiente Tumoral
20.
Cell Rep ; 8(6): 1943-1956, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25242330

RESUMEN

Colorectal cancer (CRC) initiation and growth is often attributed to stem cells, yet little is known about the regulation of these cells. We show here that a subpopulation of Prox1-transcription-factor-expressing cells have stem cell activity in intestinal adenomas, but not in the normal intestine. Using in vivo models and 3D ex vivo organoid cultures of mouse adenomas and human CRC, we found that Prox1 deletion reduced the number of stem cells and cell proliferation and decreased intestinal tumor growth via induction of annexin A1 and reduction of the actin-binding protein filamin A, which has been implicated as a prognostic marker in CRC. Loss of Prox1 also decreased autophagy and the survival of hypoxic tumor cells in tumor transplants. Thus, Prox1 is essential for the expansion of the stem cell pool in intestinal adenomas and CRC without being critical for the normal functions of the gut.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Anexina A1/antagonistas & inhibidores , Anexina A1/genética , Anexina A1/metabolismo , Autofagia , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Filaminas/antagonistas & inhibidores , Filaminas/genética , Filaminas/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Isquemia/patología , Isquemia/prevención & control , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Trasplante Heterólogo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...