Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
RNA ; 29(10): 1575-1590, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37460153

RESUMEN

Current methods for detecting unlabeled antisense oligonucleotide (ASO) drugs rely on immunohistochemistry (IHC) and/or conjugated molecules, which lack sufficient sensitivity, specificity, and resolution to fully investigate their biodistribution. Our aim was to demonstrate the qualitative and quantitative distribution of unlabeled bepirovirsen, a clinical stage ASO, in livers and kidneys of dosed mice using novel staining and imaging technologies at subcellular resolution. ASOs were detected in formalin-fixed paraffin-embedded (FFPE) and frozen tissues using an automated chromogenic in situ hybridization (ISH) assay: miRNAscope. This was then combined with immunohistochemical detection of cell lineage markers. ASO distribution in hepatocytes versus nonparenchymal cell lineages was quantified using HALO AI image analysis. To complement this, hyperspectral coherent anti-Stokes Raman scattering (HS-CARS) imaging microscopy was used to specifically detect the unique cellular Raman spectral signatures following ASO treatment. Bepirovirsen was localized primarily in nonparenchymal liver cells and proximal renal tubules. Codetection of ASO with distinct cell lineage markers of liver and kidney populations aided target cell identity facilitating quantification. Positive liver signal was quantified using HALO AI, with 12.9% of the ASO localized to the hepatocytes and 87.1% in nonparenchymal cells. HS-CARS imaging specifically detected ASO fingerprints based on the unique vibrational signatures following unlabeled ASO treatment in a totally nonperturbative manner at subcellular resolution. Together, these novel detection and imaging modalities represent a significant increase in our ability to detect unlabeled ASOs in tissues, demonstrating improved levels of specificity and resolution. These methods help us understand their underlying mechanisms of action and ultimately improve the therapeutic potential of these important drugs for treating globally significant human diseases.


Asunto(s)
Hígado , Oligonucleótidos Antisentido , Ratones , Humanos , Animales , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Distribución Tisular , Hígado/diagnóstico por imagen , Hígado/metabolismo , Hibridación in Situ , Coloración y Etiquetado
2.
Anal Chem ; 95(29): 10957-10965, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37450658

RESUMEN

Understanding drug fingerprints in complex biological samples is essential for the development of a drug. Hyperspectral coherent anti-Stokes Raman scattering (HS-CARS) microscopy, a label-free nondestructive chemical imaging technique, can profile biological samples based on their endogenous vibrational contrast. Here, we propose a deep learning-assisted HS-CARS imaging approach for the investigation of drug fingerprints and their localization at single-cell resolution. To identify and localize drug fingerprints in complex biological systems, an attention-based deep neural network, hyperspectral attention net (HAN), was developed. By formulating the task to a multiple instance learning problem, HAN highlights informative regions through the attention mechanism when being trained on whole-image labels. Using the proposed technique, we investigated the drug fingerprints of a hepatitis B virus therapy in murine liver tissues. With the increase in drug dosage, higher classification accuracy was observed, with an average area under the curve (AUC) of 0.942 for the high-dose group. Besides, highly informative tissue structures predicted by HAN demonstrated a high degree of similarity with the drug localization shown by the in situ hybridization staining results. These results demonstrate the potential of the proposed deep learning-assisted optical imaging technique for the label-free profiling, identification, and localization of drug fingerprints in biological samples, which can be extended to nonperturbative investigations of complex biological systems under various biological conditions.


Asunto(s)
Microscopía , Espectrometría Raman , Animales , Ratones , Microscopía/métodos , Espectrometría Raman/métodos , Hígado , Redes Neurales de la Computación
3.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34236045

RESUMEN

The AP-1 transcription factor c-Jun is required for Ras-driven tumorigenesis in many tissues and is considered as a classical proto-oncogene. To determine the requirement for c-Jun in a mouse model of K-RasG12D-induced lung adenocarcinoma, we inducibly deleted c-Jun in the adult lung. Surprisingly, we found that inactivation of c-Jun, or mutation of its JNK phosphorylation sites, actually increased lung tumor burden. Mechanistically, we found that protein levels of the Jun family member JunD were increased in the absence of c-Jun. In c-Jun-deficient cells, JunD phosphorylation was increased, and expression of a dominant-active JNKK2-JNK1 transgene further increased lung tumor formation. Strikingly, deletion of JunD completely abolished Ras-driven lung tumorigenesis. This work identifies JunD, not c-Jun, as the crucial substrate of JNK signaling and oncogene required for Ras-induced lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinogénesis , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas ras/metabolismo , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Genes jun/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-jun/genética , Factor de Transcripción AP-1/metabolismo
4.
Cancer Cell ; 36(1): 68-83.e9, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31257073

RESUMEN

RAC1 P29 is the third most commonly mutated codon in human cutaneous melanoma, after BRAF V600 and NRAS Q61. Here, we study the role of RAC1P29S in melanoma development and reveal that RAC1P29S activates PAK, AKT, and a gene expression program initiated by the SRF/MRTF transcriptional pathway, which results in a melanocytic to mesenchymal phenotypic switch. Mice with ubiquitous expression of RAC1P29S from the endogenous locus develop lymphoma. When expressed only in melanocytes, RAC1P29S cooperates with oncogenic BRAF or with NF1-loss to promote tumorigenesis. RAC1P29S also drives resistance to BRAF inhibitors, which is reversed by SRF/MRTF inhibitors. These findings establish RAC1P29S as a promoter of melanoma initiation and mediator of therapy resistance, while identifying SRF/MRTF as a potential therapeutic target.


Asunto(s)
Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/genética , Melanoma/etiología , Melanoma/patología , Mutación , Proteína de Unión al GTP rac1/genética , Alelos , Sustitución de Aminoácidos , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Melanocitos/metabolismo , Melanoma/mortalidad , Melanoma/terapia , Ratones , Ratones Transgénicos , Modelos Biológicos , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Factor de Respuesta Sérica , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncogenesis ; 8(3): 13, 2019 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-30783098

RESUMEN

Colorectal cancer (CRC) patients develop recurrence after chemotherapy owing to the survival of stem cell-like cells referred to as cancer stem-like cells (CSCs). The origin of CSCs is linked to the epithelial-mesenchymal transition (EMT) process. Currently, it remains poorly understood how EMT programmes enable CSCs residing in the tumour microenvironment to escape the effects of chemotherapy. This study identifies a key molecular pathway that is responsible for the formation of drug-resistant CSC populations. Using a modified yeast-2-hybrid system and 2D gel-based proteomics methods, we show that the E3-ubiquitin ligase FBXW7 directly binds and degrades the EMT-inducing transcription factor ZEB2 in a phosphorylation-dependent manner. Loss of FBXW7 induces an EMT that can be effectively reversed by knockdown of ZEB2. The FBXW7-ZEB2 axis regulates such important cancer cell features, as stemness/dedifferentiation, chemoresistance and cell migration in vitro, ex vivo and in animal models of metastasis. High expression of ZEB2 in cancer tissues defines the reduced ZEB2 expression in the cancer-associated stroma in patients and in murine intestinal organoids, demonstrating a tumour-stromal crosstalk that modulates a niche and EMT activation. Our study thus uncovers a new molecular mechanism, by which the CRC cells display differences in resistance to chemotherapy and metastatic potential.

6.
Cell Rep ; 25(13): 3545-3553.e2, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30590030

RESUMEN

RAS family GTPases contribute directly to the regulation of type I phosphoinositide 3-kinases (PI3Ks) via RAS-binding domains in the PI3K catalytic p110 subunits. Disruption of this domain of p110α impairs RAS-mutant-oncogene-driven tumor formation and maintenance. Here, we test the effect of blocking the interaction of RAS with p110α on epidermal growth factor receptor (EGFR)-mutant-driven lung tumorigenesis. Disrupting the RAS-PI3K interaction inhibits activation of both AKT and RAC1 in EGFR-mutant lung cancer cells, leading to reduced growth and survival, and inhibits EGFR-mutant-induced tumor onset and promotes major regression of established tumors in an autochthonous mouse model of EGFR-mutant-induced lung adenocarcinoma. The RAS-PI3K interaction is thus an important signaling node and potential therapeutic target in EGFR-mutant lung cancer, even though RAS oncogenes are not themselves mutated in this setting, suggesting different strategies for tackling tyrosine kinase inhibitor resistance in lung cancer.


Asunto(s)
Receptores ErbB/genética , Neoplasias Pulmonares/genética , Mutación/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/química , Unión Proteica/efectos de los fármacos , Dominios Proteicos
7.
Elife ; 72018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30231971

RESUMEN

Squamous cell carcinoma (SCC) can progress to malignant metastatic cancer, including an aggressive subtype known as spindle cell carcinoma (spSCC). spSCC formation involves epithelial-to-mesenchymal transition (EMT), yet the molecular basis of this event remains unknown. The transcriptional co-activator YAP undergoes recurrent amplification in human SCC and overexpression of YAP drives SCC formation in mice. Here, we show that human spSCC tumours also feature strong nuclear localisation of YAP and overexpression of activated YAP (NLS-YAP-5SA) with Keratin-5 (K5-CreERt) is sufficient to induce rapid formation of both SCC and spSCC in mice. spSCC tumours arise at sites of epithelial scratch wounding, where tumour-initiating epithelial cells undergo EMT to generate spSCC. Expression of the EMT transcription factor ZEB1 arises upon wounding and is a defining characteristic of spSCC in mice and humans. Thus, the wound healing response synergises with YAP to drive metaplastic transformation of SCC to spSCC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Fosfoproteínas/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Animales , Núcleo Celular/metabolismo , Epidermis/patología , Transición Epitelial-Mesenquimal , Humanos , Ratones , Factores de Transcripción , Proteínas Señalizadoras YAP , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
8.
Mol Cancer Res ; 16(12): 1977-1990, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30097457

RESUMEN

Wnt/ß-catenin signaling plays a critical role during development of both normal and malignant colorectal cancer tissues. Phosphorylation of ß-catenin protein alters its trafficking and function. Such conventional allosteric regulation usually involves a highly specialized set of molecular interactions, which may specifically turn on a particular cell phenotype. This study identifies a novel transcription modulator with an FLYWCH/Zn-finger DNA-binding domain, called "FLYWCH1." Using a modified yeast-2-hybrid based Ras-Recruitment system, it is demonstrated that FLYWCH1 directly binds to unphosphorylated (nuclear) ß-catenin efficiently suppressing the transcriptional activity of Wnt/ß-catenin signaling that cannot be rescued by TCF4. FLYWCH1 rearranges the transcriptional activity of ß-catenin/TCF4 to selectively block the expression of specific downstream genes associated with colorectal cancer cell migration and morphology, including ZEB1, EPHA4, and E-cadherin. Accordingly, overexpression of FLYWCH1 reduces cell motility and increases cell attachment. The expression of FLYWCH1 negatively correlates with the expression level of ZEB1 and EPHA4 in normal versus primary and metastatic colorectal cancer tissues in patients. Thus, FLYWCH1 antagonizes ß-catenin/TCF4 signaling during cell polarity/migration in colorectal cancer. IMPLICATIONS: This study uncovers a new molecular mechanism by which FLYWCH1 with a possible tumor suppressive role represses ß-catenin-induced ZEB1 and increases cadherin-mediated cell attachment preventing colorectal cancer metastasis.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proteínas de Unión al ADN/química , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Análisis de Matrices Tisulares , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Dedos de Zinc
9.
J Clin Invest ; 128(4): 1326-1337, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29346117

RESUMEN

The tumor suppressor FBW7 targets oncoproteins such as c-MYC for ubiquitylation and is mutated in several human cancers. We noted that in a substantial percentage of colon cancers, FBW7 protein is undetectable despite the presence of FBW7 mRNA. To understand the molecular mechanism of FBW7 regulation in these cancers, we employed proteomics and identified the deubiquitinase (DUB) USP9X as an FBW7 interactor. USP9X antagonized FBW7 ubiquitylation, and Usp9x deletion caused Fbw7 destabilization. Mice lacking Usp9x in the gut showed reduced secretory cell differentiation and increased progenitor proliferation, phenocopying Fbw7 loss. In addition, Usp9x inactivation impaired intestinal regeneration and increased tumor burden in colitis-associated intestinal cancer. c-Myc heterozygosity abrogated increased progenitor proliferation and tumor burden in Usp9x-deficient mice, suggesting that Usp9x suppresses tumor formation by regulating Fbw7 protein stability and thereby reducing c-Myc. Thus, we identify a tumor suppressor mechanism in the mammalian intestine that arises from the posttranslational regulation of FBW7 by USP9X independent of somatic FBW7 mutations.


Asunto(s)
Neoplasias Colorrectales/enzimología , Endopeptidasas/biosíntesis , Proteína 7 que Contiene Repeticiones F-Box-WD/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas Supresoras de Tumor/biosíntesis , Ubiquitina Tiolesterasa/biosíntesis , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Endopeptidasas/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Células HCT116 , Humanos , Ratones , Ratones Mutantes , Muramidasa , Mutación , Fragmentos de Péptidos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética
10.
Immunity ; 47(6): 1083-1099.e6, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29246442

RESUMEN

The immunosuppressive protein PD-L1 is upregulated in many cancers and contributes to evasion of the host immune system. The relative importance of the tumor microenvironment and cancer cell-intrinsic signaling in the regulation of PD-L1 expression remains unclear. We report that oncogenic RAS signaling can upregulate tumor cell PD-L1 expression through a mechanism involving increases in PD-L1 mRNA stability via modulation of the AU-rich element-binding protein tristetraprolin (TTP). TTP negatively regulates PD-L1 expression through AU-rich elements in the 3' UTR of PD-L1 mRNA. MEK signaling downstream of RAS leads to phosphorylation and inhibition of TTP by the kinase MK2. In human lung and colorectal tumors, RAS pathway activation is associated with elevated PD-L1 expression. In vivo, restoration of TTP expression enhances anti-tumor immunity dependent on degradation of PD-L1 mRNA. We demonstrate that RAS can drive cell-intrinsic PD-L1 expression, thus presenting therapeutic opportunities to reverse the innately immunoresistant phenotype of RAS mutant cancers.


Asunto(s)
Antígeno B7-H1/inmunología , Neoplasias Colorrectales/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/inmunología , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Tristetraprolina/inmunología , Escape del Tumor , Animales , Antígeno B7-H1/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células Epiteliales/inmunología , Células Epiteliales/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas p21(ras)/genética , División del ARN , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/inmunología , Transducción de Señal , Tristetraprolina/genética
11.
Cell Rep ; 21(4): 966-978, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29069604

RESUMEN

The cell of origin of pancreatic ductal adenocarcinoma (PDAC) has been controversial. Here, we show that identical oncogenic drivers trigger PDAC originating from both ductal and acinar cells with similar histology but with distinct pathophysiology and marker expression dependent on cell of origin. Whereas acinar-derived tumors exhibited low AGR2 expression and were preceded by pancreatic intraepithelial neoplasias (PanINs), duct-derived tumors displayed high AGR2 and developed independently of a PanIN stage via non-mucinous lesions. Using orthotopic transplantation and chimera experiments, we demonstrate that PanIN-like lesions can be induced by PDAC as bystanders in adjacent healthy tissues, explaining the co-existence of mucinous and non-mucinous lesions and highlighting the need to distinguish between true precursor PanINs and PanIN-like bystander lesions. Our results suggest AGR2 as a tool to stratify PDAC according to cell of origin, highlight that not all PanIN-like lesions are precursors of PDAC, and add an alternative progression route to the current model of PDAC development.


Asunto(s)
Células Acinares/patología , Carcinogénesis , Carcinoma Ductal Pancreático/patología , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Células Acinares/metabolismo , Animales , Carcinoma Ductal Pancreático/metabolismo , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Mucoproteínas/genética , Mucoproteínas/metabolismo , Proteínas Oncogénicas , Conductos Pancreáticos/metabolismo , Neoplasias Pancreáticas/metabolismo
12.
Sci Rep ; 7(1): 518, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28364121

RESUMEN

The myocardin-related transcription factor/serum response factor (MRTF/SRF) pathway represents a promising therapeutic target to prevent fibrosis. We have tested the effects of new pharmacological inhibitors of MRTF/SRF signalling in a preclinical model of fibrosis. CCG-222740, a novel MRTF/SRF inhibitor, markedly decreased SRF reporter gene activity and showed a greater inhibitory effect on MRTF/SRF target genes than the previously described MRTF-A inhibitor CCG-203971. CCG-222740 was also five times more potent, with an IC50 of 5 µM, in a fibroblast-mediated collagen contraction assay, was less cytotoxic, and a more potent inhibitor of alpha-smooth muscle actin protein expression than CCG-203971. Local delivery of CCG-222740 and CCG-203971 in a validated and clinically relevant rabbit model of scar tissue formation after glaucoma filtration surgery increased the long-term success of the surgery by 67% (P < 0.0005) and 33% (P < 0.01), respectively, and significantly decreased fibrosis and scarring histologically. Unlike mitomycin-C, neither CCG-222740 nor CCG-203971 caused any detectable epithelial toxicity or systemic side effects with very low drug levels measured in the aqueous, vitreous, and serum. We conclude that inhibitors of MRTF/SRF-regulated gene transcription such as CCG-222740, potentially represent a new therapeutic strategy to prevent scar tissue formation in the eye and other tissues.


Asunto(s)
Cicatriz/metabolismo , Cicatriz/patología , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Animales , Células Cultivadas , Cicatriz/prevención & control , Colágeno/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Matriz Extracelular , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Humanos , Conejos , Transducción de Señal/efectos de los fármacos
13.
Cell ; 168(5): 843-855.e13, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28215706

RESUMEN

The transcription-related DNA damage response was analyzed on a genome-wide scale with great spatial and temporal resolution. Upon UV irradiation, a slowdown of transcript elongation and restriction of gene activity to the promoter-proximal ∼25 kb is observed. This is associated with a shift from expression of long mRNAs to shorter isoforms, incorporating alternative last exons (ALEs) that are more proximal to the transcription start site. Notably, this includes a shift from a protein-coding ASCC3 mRNA to a shorter ALE isoform of which the RNA, rather than an encoded protein, is critical for the eventual recovery of transcription. The non-coding ASCC3 isoform counteracts the function of the protein-coding isoform, indicating crosstalk between them. Thus, the ASCC3 gene expresses both coding and non-coding transcript isoforms with opposite effects on transcription recovery after UV-induced DNA damage.


Asunto(s)
Empalme Alternativo/efectos de la radiación , ADN Helicasas/genética , ARN no Traducido/genética , Transcripción Genética , Rayos Ultravioleta , Línea Celular , Exones , Humanos , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elongación de la Transcripción Genética/efectos de la radiación , Iniciación de la Transcripción Genética/efectos de la radiación
14.
Sci Rep ; 7: 41722, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28155887

RESUMEN

The influence of parasite genetic factors on immune responses and development of severe pathology of malaria is largely unknown. In this study, we performed genome-wide transcriptomic profiling of mouse whole blood during blood-stage infections of two strains of the rodent malaria parasite Plasmodium chabaudi that differ in virulence. We identified several transcriptomic signatures associated with the virulent infection, including signatures for platelet aggregation, stronger and prolonged anemia and lung inflammation. The first two signatures were detected prior to pathology. The anemia signature indicated deregulation of host erythropoiesis, and the lung inflammation signature was linked to increased neutrophil infiltration, more cell death and greater parasite sequestration in the lungs. This comparative whole-blood transcriptomics profiling of virulent and avirulent malaria shows the validity of this approach to inform severity of the infection and provide insight into pathogenic mechanisms.


Asunto(s)
Interacciones Huésped-Parásitos/genética , Malaria/genética , Malaria/parasitología , Plasmodium/fisiología , Transcriptoma , Animales , Análisis por Conglomerados , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Perfilación de la Expresión Génica , Estadios del Ciclo de Vida , Malaria/sangre , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Especificidad de Órganos/genética , Plasmodium chabaudi/fisiología , Agregación Plaquetaria , Virulencia
15.
Cancer Cell ; 31(1): 79-93, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28073006

RESUMEN

Chromosomal instability (CIN) contributes to cancer evolution, intratumor heterogeneity, and drug resistance. CIN is driven by chromosome segregation errors and a tolerance phenotype that permits the propagation of aneuploid genomes. Through genomic analysis of colorectal cancers and cell lines, we find frequent loss of heterozygosity and mutations in BCL9L in aneuploid tumors. BCL9L deficiency promoted tolerance of chromosome missegregation events, propagation of aneuploidy, and genetic heterogeneity in xenograft models likely through modulation of Wnt signaling. We find that BCL9L dysfunction contributes to aneuploidy tolerance in both TP53-WT and mutant cells by reducing basal caspase-2 levels and preventing cleavage of MDM2 and BID. Efforts to exploit aneuploidy tolerance mechanisms and the BCL9L/caspase-2/BID axis may limit cancer diversity and evolution.


Asunto(s)
Aneuploidia , Caspasa 2/fisiología , Neoplasias Colorrectales/genética , Cisteína Endopeptidasas/fisiología , Proteínas de Unión al ADN/fisiología , Factores de Transcripción/fisiología , Anciano , Anciano de 80 o más Años , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Caspasa 2/análisis , Segregación Cromosómica , Cisteína Endopeptidasas/análisis , Proteínas de Unión al ADN/genética , Células HCT116 , Humanos , Ratones , Persona de Mediana Edad , Mutación , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/fisiología
16.
J Neurosci ; 37(9): 2305-2316, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130357

RESUMEN

Midbrain dopaminergic neurons are highly heterogeneous. They differ in their connectivity and firing patterns and, therefore, in their functional properties. The molecular underpinnings of this heterogeneity are largely unknown, and there is a paucity of markers that distinguish these functional subsets. In this paper, we report the identification and characterization of a novel subset of midbrain dopaminergic neurons located in the ventral tegmental area that expresses the basic helix-loop-helix transcription factor, Neurogenic Differentiation Factor-6 (NEUROD6). Retrograde fluorogold tracing experiments demonstrate that Neurod6+ midbrain dopaminergic neurons neurons project to two distinct septal regions: the dorsal and intermediate region of the lateral septum. Loss-of-function studies in mice demonstrate that Neurod6 and the closely related family member Neurod1 are both specifically required for the survival of this lateral-septum projecting neuronal subset during development. Our findings underscore the complex organization of midbrain dopaminergic neurons and provide an entry point for future studies of the functions of the Neurod6+ subset of midbrain dopaminergic neurons.SIGNIFICANCE STATEMENT Midbrain dopaminergic neurons regulate diverse brain functions, including voluntary movement and cognitive and emotive behaviors. These neurons are heterogeneous, and distinct subsets are thought to regulate different behaviors. However, we currently lack the means to identify and modify gene function in specific subsets of midbrain dopaminergic neurons. In this study, we identify the transcription factor NEUROD6 as a specific marker for a novel subset of midbrain dopaminergic neurons in the ventral midbrain that project to the lateral septum, and we reveal essential roles for Neurod1 and Neurod6 in the survival of these neurons during development. Our findings highlight the molecular and anatomical heterogeneity of midbrain dopaminergic neurons and contribute to a better understanding of this functionally complex group of neurons.


Asunto(s)
Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neuronas Dopaminérgicas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Núcleos Septales/citología , Área Tegmental Ventral/citología , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biotina/análogos & derivados , Biotina/metabolismo , Calbindinas/metabolismo , Recuento de Células , Dextranos/metabolismo , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Vías Nerviosas/fisiología , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Retinal-Deshidrogenasa , Núcleos Septales/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/embriología , Área Tegmental Ventral/crecimiento & desarrollo
17.
Blood ; 129(4): 415-419, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27903529

RESUMEN

Microvascular endothelial cells (ECs) display a high degree of phenotypic and functional heterogeneity among different organs. Organ-specific ECs control their tissue microenvironment by angiocrine factors in health and disease. Liver sinusoidal endothelial cells (LSECs) are uniquely differentiated to fulfill important organ-specific functions in development, under homeostatic conditions, and in regeneration and liver pathology. Recently, Bmp2 has been identified by us as an organ-specific angiokine derived from LSECs. To study angiocrine Bmp2 signaling in the liver, we conditionally deleted Bmp2 in LSECs using EC subtype-specific Stab2-Cre mice. Genetic inactivation of hepatic angiocrine Bmp2 signaling in Stab2-Cre;Bmp2fl/fl (Bmp2LSECKO) mice caused massive iron overload in the liver and increased serum iron levels and iron deposition in several organs similar to classic hereditary hemochromatosis. Iron overload was mediated by decreased hepatic expression of hepcidin, a key regulator of iron homeostasis. Thus, angiocrine Bmp2 signaling within the hepatic vascular niche represents a constitutive pathway indispensable for iron homeostasis in vivo that is nonredundant with Bmp6. Notably, we demonstrate that organ-specific angiocrine signaling is essential not only for the homeostasis of the respective organ but also for the homeostasis of the whole organism.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Células Endoteliales/metabolismo , Hemocromatosis/genética , Hepcidinas/genética , Homeostasis/genética , Hierro/metabolismo , Hígado/metabolismo , Animales , Proteína Morfogenética Ósea 2/deficiencia , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Capilares/metabolismo , Capilares/patología , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Hemocromatosis/metabolismo , Hemocromatosis/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Hepcidinas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Hígado/irrigación sanguínea , Hígado/patología , Masculino , Ratones , Ratones Transgénicos , Comunicación Paracrina , Transducción de Señal , Transcripción Genética
18.
Nat Cell Biol ; 18(12): 1346-1356, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27798604

RESUMEN

The mammary gland is composed of a complex cellular hierarchy with unusual postnatal plasticity. The identities of stem/progenitor cell populations, as well as tumour-initiating cells that give rise to breast cancer, are incompletely understood. Here we show that Lgr6 marks rare populations of cells in both basal and luminal mammary gland compartments in mice. Lineage tracing analysis showed that Lgr6+ cells are unipotent progenitors, which expand clonally during puberty but diminish in adulthood. In pregnancy or following stimulation with ovarian hormones, adult Lgr6+ cells regained proliferative potency and their progeny formed alveoli over repeated pregnancies. Oncogenic mutations in Lgr6+ cells resulted in expansion of luminal cells, culminating in mammary gland tumours. Conversely, depletion of Lgr6+ cells in the MMTV-PyMT model of mammary tumorigenesis significantly impaired tumour growth. Thus, Lgr6 marks mammary gland progenitor cells that can initiate tumours, and cells of luminal breast tumours required for efficient tumour maintenance.


Asunto(s)
Neoplasias de la Mama/patología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/patología , Alelos , Animales , Animales Recién Nacidos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Carcinogénesis/patología , Linaje de la Célula , Proliferación Celular , Células Clonales , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Homeostasis , Hormonas/farmacología , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/genética , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Embarazo , Células Madre/metabolismo , Regulación hacia Arriba
19.
Science ; 353(6307)2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27708074

RESUMEN

Tumors comprise functionally diverse subpopulations of cells with distinct proliferative potential. Here, we show that dynamic epigenetic states defined by the linker histone H1.0 determine which cells within a tumor can sustain the long-term cancer growth. Numerous cancer types exhibit high inter- and intratumor heterogeneity of H1.0, with H1.0 levels correlating with tumor differentiation status, patient survival, and, at the single-cell level, cancer stem cell markers. Silencing of H1.0 promotes maintenance of self-renewing cells by inducing derepression of megabase-sized gene domains harboring downstream effectors of oncogenic pathways. Self-renewing epigenetic states are not stable, and reexpression of H1.0 in subsets of tumor cells establishes transcriptional programs that restrict cancer cells' long-term proliferative potential and drive their differentiation. Our results uncover epigenetic determinants of tumor-maintaining cells.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Histonas/genética , Neoplasias/genética , Neoplasias/patología , Adenina/química , Línea Celular Tumoral , ADN/química , Metilación de ADN , Elementos de Facilitación Genéticos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias/mortalidad , Nucleosomas/metabolismo , ARN Interferente Pequeño/genética , Timina/química
20.
Cell ; 167(1): 203-218.e17, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27641500

RESUMEN

Many body surfaces harbor organ-specific γδ T cell compartments that contribute to tissue integrity. Thus, murine dendritic epidermal T cells (DETCs) uniquely expressing T cell receptor (TCR)-Vγ5 chains protect from cutaneous carcinogens. The DETC repertoire is shaped by Skint1, a butyrophilin-like (Btnl) gene expressed specifically by thymic epithelial cells and suprabasal keratinocytes. However, the generality of this mechanism has remained opaque, since neither Skint1 nor DETCs are evolutionarily conserved. Here, Btnl1 expressed by murine enterocytes is shown to shape the local TCR-Vγ7(+) γδ compartment. Uninfluenced by microbial or food antigens, this activity evokes the developmental selection of TCRαß(+) repertoires. Indeed, Btnl1 and Btnl6 jointly induce TCR-dependent responses specifically in intestinal Vγ7(+) cells. Likewise, human gut epithelial cells express BTNL3 and BTNL8 that jointly induce selective TCR-dependent responses of human colonic Vγ4(+) cells. Hence, a conserved mechanism emerges whereby epithelia use organ-specific BTNL/Btnl genes to shape local T cell compartments.


Asunto(s)
Butirofilinas/inmunología , Mucosa Intestinal/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Butirofilinas/genética , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Timo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA