Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 52(2): 357-373.e9, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32049051

RESUMEN

Clearance of apoptotic cells by macrophages prevents excessive inflammation and supports immune tolerance. Here, we examined the effect of blocking apoptotic cell clearance on anti-tumor immune response. We generated an antibody that selectively inhibited efferocytosis by phagocytic receptor MerTK. Blockade of MerTK resulted in accumulation of apoptotic cells within tumors and triggered a type I interferon response. Treatment of tumor-bearing mice with anti-MerTK antibody stimulated T cell activation and synergized with anti-PD-1 or anti-PD-L1 therapy. The anti-tumor effect induced by anti-MerTK treatment was lost in Stinggt/gt mice, but not in Cgas-/- mice. Abolishing cGAMP production in Cgas-/- tumor cells, depletion of extracellular ATP, or inactivation of the ATP-gated P2X7R channel also compromised the effects of MerTK blockade. Mechanistically, extracellular ATP acted via P2X7R to enhance the transport of extracellular cGAMP into macrophages and subsequent STING activation. Thus, MerTK blockade increases tumor immunogenicity and potentiates anti-tumor immunity, which has implications for cancer immunotherapy.


Asunto(s)
Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Neoplasias/inmunología , Nucleótidos Cíclicos/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Tirosina Quinasa c-Mer/inmunología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Antígeno B7-H1/inmunología , Células Cultivadas , Femenino , Inmunidad Innata , Inmunoterapia , Interferón Tipo I/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Nucleotidiltransferasas/deficiencia , Nucleotidiltransferasas/metabolismo , Fagocitosis , Receptor de Muerte Celular Programada 1/inmunología , Receptores Purinérgicos P2X7/deficiencia , Transducción de Señal/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa c-Mer/genética
2.
Nature ; 594(7863): 418-423, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33953400

RESUMEN

Although RAF monomer inhibitors (type I.5, BRAF(V600)) are clinically approved for the treatment of BRAFV600-mutant melanoma, they are ineffective in non-BRAFV600 mutant cells1-3. Belvarafenib is a potent and selective RAF dimer (type II) inhibitor that exhibits clinical activity in patients with BRAFV600E- and NRAS-mutant melanomas. Here we report the first-in-human phase I study investigating the maximum tolerated dose, and assessing the safety and preliminary efficacy of belvarafenib in BRAFV600E- and RAS-mutated advanced solid tumours (NCT02405065, NCT03118817). By generating belvarafenib-resistant NRAS-mutant melanoma cells and analysing circulating tumour DNA from patients treated with belvarafenib, we identified new recurrent mutations in ARAF within the kinase domain. ARAF mutants conferred resistance to belvarafenib in both a dimer- and a kinase activity-dependent manner. Belvarafenib induced ARAF mutant dimers, and dimers containing mutant ARAF were active in the presence of inhibitor. ARAF mutations may serve as a general resistance mechanism for RAF dimer inhibitors as the mutants exhibit reduced sensitivity to a panel of type II RAF inhibitors. The combination of RAF plus MEK inhibition may be used to delay ARAF-driven resistance and suggests a rational combination for clinical use. Together, our findings reveal specific and compensatory functions for the ARAF isoform and implicate ARAF mutations as a driver of resistance to RAF dimer inhibitors.


Asunto(s)
Resistencia a Antineoplásicos/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Mutación , Proteínas Proto-Oncogénicas A-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas A-raf/genética , Quinasas raf/antagonistas & inhibidores , Animales , Línea Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Melanoma/patología , Ratones , Multimerización de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas A-raf/química , Quinasas raf/química
3.
EMBO Rep ; 24(3): e55532, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36621885

RESUMEN

Preclinical and clinical studies demonstrate that T cell-dependent bispecific antibodies (TDBs) induce systemic changes in addition to tumor killing, leading to adverse events. Here, we report an in-depth characterization of acute responses to TDBs in tumor-bearing mice. Contrary to modest changes in tumors, rapid and substantial lymphocyte accumulation and endothelial cell (EC) activation occur around large blood vessels in normal organs including the liver. We hypothesize that organ-specific ECs may account for the differential responses in normal tissues and tumors, and we identify a list of genes selectively upregulated by TDB in large liver vessels. Using one of the genes as an example, we demonstrate that CD9 facilitates ICAM-1 to support T cell-EC interaction in response to soluble factors released from a TDB-mediated cytotoxic reaction. Our results suggest that multiple factors may cooperatively promote T cell infiltration into normal organs as a secondary response to TDB-mediated tumor killing. These data shed light on how different vascular beds respond to cancer immunotherapy and may help improve their safety and efficacy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Ratones , Animales , Linfocitos T , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Neoplasias/tratamiento farmacológico , Comunicación Celular , Células Endoteliales
4.
Proc Natl Acad Sci U S A ; 119(46): e2207327119, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36343233

RESUMEN

Developing peptide-based tools to fine-tune growth signaling pathways, in particular molecules with exquisite selectivity and high affinities, opens up opportunities for cellular reprogramming in tissue regeneration. Here, we present a library based on cystine-knot peptides (CKPs) that incorporate multiple loops for randomization and selection via directed evolution. Resulting binders could be assembled into multimeric structures to fine-tune cellular signaling. An example is presented for the Wnt pathway, which plays a key role in the homeostasis and regeneration of tissues such as lung, skin, and intestine. We discovered picomolar affinity CKP agonists of the human LPR6 receptor by exploring the limits of the topological manipulation of LRP6 dimerization. Structural analyses revealed that the agonists bind at the first ß-propeller domain of LRP6, mimicking the natural Wnt inhibitors DKK1 and SOST. However, the CKP agonists exhibit a different mode of action as they amplify the signaling of natural Wnt ligands but do not activate the pathway by themselves. In an alveolosphere organoid model, the CKP agonists induced alveolar stem cell activity. They also stimulated growth in primary human intestinal organoids. The approach described here advances the important frontier of next-generation agonist design and could be applied to other signaling pathways to discover tunable agonist ligands.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , Humanos , beta Catenina/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas Wnt/metabolismo , Cistina , Ligandos , Péptidos
5.
Nature ; 562(7727): 429-433, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30297801

RESUMEN

Despite the efficacy of Hedgehog pathway inhibitors in the treatment of basal cell carcinoma (BCC)1, residual disease persists in some patients and may contribute to relapse when treatment is discontinued2. Here, to study the effect of the Smoothened inhibitor vismodegib on tumour clearance, we have used a Ptch1-Trp53 mouse model of BCC3 and found that mice treated with vismodegib harbour quiescent residual tumours that regrow upon cessation of treatment. Profiling experiments revealed that residual BCCs initiate a transcriptional program that closely resembles that of stem cells of the interfollicular epidermis and isthmus, whereas untreated BCCs are more similar to the hair follicle bulge. This cell identity switch was enabled by a mostly permissive chromatin state accompanied by rapid Wnt pathway activation and reprogramming of super enhancers to drive activation of key transcription factors involved in cellular identity. Accordingly, treatment of BCC with both vismodegib and a Wnt pathway inhibitor reduced the residual tumour burden and enhanced differentiation. Our study identifies a resistance mechanism in which tumour cells evade treatment by adopting an alternative identity that does not rely on the original oncogenic driver for survival.


Asunto(s)
Anilidas/farmacología , Carcinoma Basocelular/patología , Diferenciación Celular/efectos de los fármacos , Proteínas Hedgehog/antagonistas & inhibidores , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/patología , Anilidas/administración & dosificación , Anilidas/uso terapéutico , Animales , Carcinoma Basocelular/tratamiento farmacológico , Carcinoma Basocelular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Epidérmicas/efectos de los fármacos , Células Epidérmicas/metabolismo , Células Epidérmicas/patología , Folículo Piloso/efectos de los fármacos , Folículo Piloso/metabolismo , Folículo Piloso/patología , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Piridinas/administración & dosificación , Piridinas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Receptor Smoothened/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/patología , Vía de Señalización Wnt/efectos de los fármacos
6.
Nature ; 543(7647): 676-680, 2017 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-28358093

RESUMEN

Cancer stem cells (CSCs) have been hypothesized to represent the driving force behind tumour progression and metastasis, making them attractive cancer targets. However, conclusive experimental evidence for their functional relevance is still lacking for most malignancies. Here we show that the leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5) identifies intestinal CSCs in mouse tumours engineered to recapitulate the clinical progression of human colorectal cancer. We demonstrate that selective Lgr5+ cell ablation restricts primary tumour growth, but does not result in tumour regression. Instead, tumours are maintained by proliferative Lgr5- cells that continuously attempt to replenish the Lgr5+ CSC pool, leading to rapid re-initiation of tumour growth upon treatment cessation. Notably, CSCs are critical for the formation and maintenance of liver metastasis derived from colorectal cancers. Together, our data highlight distinct CSC dependencies for primary versus metastasic tumour growth, and suggest that targeting CSCs may represent a therapeutic opportunity for managing metastatic disease.


Asunto(s)
Neoplasias Colorrectales/patología , Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Separación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Inyecciones Subcutáneas , Intestinos/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Ratones , Metástasis de la Neoplasia/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Organoides/patología , Organoides/trasplante , Receptores Acoplados a Proteínas G/análisis
7.
Nature ; 550(7675): 249-254, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-29022589

RESUMEN

Adenosine-to-inosine (A-to-I) RNA editing is a conserved post-transcriptional mechanism mediated by ADAR enzymes that diversifies the transcriptome by altering selected nucleotides in RNA molecules. Although many editing sites have recently been discovered, the extent to which most sites are edited and how the editing is regulated in different biological contexts are not fully understood. Here we report dynamic spatiotemporal patterns and new regulators of RNA editing, discovered through an extensive profiling of A-to-I RNA editing in 8,551 human samples (representing 53 body sites from 552 individuals) from the Genotype-Tissue Expression (GTEx) project and in hundreds of other primate and mouse samples. We show that editing levels in non-repetitive coding regions vary more between tissues than editing levels in repetitive regions. Globally, ADAR1 is the primary editor of repetitive sites and ADAR2 is the primary editor of non-repetitive coding sites, whereas the catalytically inactive ADAR3 predominantly acts as an inhibitor of editing. Cross-species analysis of RNA editing in several tissues revealed that species, rather than tissue type, is the primary determinant of editing levels, suggesting stronger cis-directed regulation of RNA editing for most sites, although the small set of conserved coding sites is under stronger trans-regulation. In addition, we curated an extensive set of ADAR1 and ADAR2 targets and showed that many editing sites display distinct tissue-specific regulation by the ADAR enzymes in vivo. Further analysis of the GTEx data revealed several potential regulators of editing, such as AIMP2, which reduces editing in muscles by enhancing the degradation of the ADAR proteins. Collectively, our work provides insights into the complex cis- and trans-regulation of A-to-I editing.


Asunto(s)
Adenosina Desaminasa , Primates/genética , Edición de ARN/genética , Proteínas de Unión al ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Femenino , Genotipo , Células HEK293 , Humanos , Masculino , Ratones , Músculos/metabolismo , Proteínas Nucleares/metabolismo , Especificidad de Órganos/genética , Proteolisis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis Espacio-Temporal , Especificidad de la Especie , Transcriptoma/genética
8.
Nat Chem Biol ; 14(6): 582-590, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29632413

RESUMEN

Regeneration of the adult intestinal epithelium is mediated by a pool of cycling stem cells, which are located at the base of the crypt, that express leucine-rich-repeat-containing G-protein-coupled receptor 5 (LGR5). The Frizzled (FZD) 7 receptor (FZD7) is enriched in LGR5+ intestinal stem cells and plays a critical role in their self-renewal. Yet, drug discovery approaches and structural bases for targeting specific FZD isoforms remain poorly defined. FZD proteins interact with Wnt signaling proteins via, in part, a lipid-binding groove on the extracellular cysteine-rich domain (CRD) of the FZD receptor. Here we report the identification of a potent peptide that selectively binds to the FZD7 CRD at a previously uncharacterized site and alters the conformation of the CRD and the architecture of its lipid-binding groove. Treatment with the FZD7-binding peptide impaired Wnt signaling in cultured cells and stem cell function in intestinal organoids. Together, our data illustrate that targeting the lipid-binding groove holds promise as an approach for achieving isoform-selective FZD receptor inhibition.


Asunto(s)
Receptores Frizzled/antagonistas & inhibidores , Receptores Frizzled/metabolismo , Intestinos/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Sitios de Unión , Células CHO , Membrana Celular/metabolismo , Cricetulus , Cristalografía por Rayos X , Descubrimiento de Drogas , Femenino , Citometría de Flujo , Células HEK293 , Humanos , Intestinos/citología , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Péptidos/química , Unión Proteica , Multimerización de Proteína , Regeneración , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Células Madre/patología , Resonancia por Plasmón de Superficie , Vía de Señalización Wnt
9.
Nat Chem Biol ; 14(9): 902, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29728602

RESUMEN

The version of this article originally published contained older versions of the Life Sciences Reporting Summary and the Supplementary Text and Figures. The error has been corrected in the HTML and PDF versions of the article.

10.
Dev Biol ; 426(2): 155-164, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27157655

RESUMEN

The Xenopus community has embraced recent advances in sequencing technology, resulting in the accumulation of numerous RNA-Seq and ChIP-Seq datasets. However, easily accessing and comparing datasets generated by multiple laboratories is challenging. Thus, we have created a central space to view, search and analyze data, providing essential information on gene expression changes and regulatory elements present in the genome. XenMine (www.xenmine.org) is a user-friendly website containing published genomic datasets from both Xenopus tropicalis and Xenopus laevis. We have established an analysis pipeline where all published datasets are uniformly processed with the latest genome releases. Information from these datasets can be extracted and compared using an array of pre-built or custom templates. With these search tools, users can easily extract sequences for all putative regulatory domains surrounding a gene of interest, identify the expression values of a gene of interest over developmental time, and analyze lists of genes for gene ontology terms and publications. Additionally, XenMine hosts an in-house genome browser that allows users to visualize all available ChIP-Seq data, extract specifically marked sequences, and aid in identifying important regulatory elements within the genome. Altogether, XenMine is an excellent tool for visualizing, accessing and querying analyzed datasets rapidly and efficiently.


Asunto(s)
Minería de Datos , Bases de Datos Genéticas , Genoma , Genómica/métodos , Xenopus/genética , Animales , Secuencia de Bases , Conjuntos de Datos como Asunto , Expresión Génica , Ontología de Genes , Internet , ARN/biosíntesis , ARN/genética , Secuencias Reguladoras de Ácidos Nucleicos , Programas Informáticos
11.
PLoS Genet ; 10(5): e1004304, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24786518

RESUMEN

Personal exome and genome sequencing provides access to loss-of-function and rare deleterious alleles whose interpretation is expected to provide insight into individual disease burden. However, for each allele, accurate interpretation of its effect will depend on both its penetrance and the trait's expressivity. In this regard, an important factor that can modify the effect of a pathogenic coding allele is its level of expression; a factor which itself characteristically changes across tissues. To better inform the degree to which pathogenic alleles can be modified by expression level across multiple tissues, we have conducted exome, RNA and deep, targeted allele-specific expression (ASE) sequencing in ten tissues obtained from a single individual. By combining such data, we report the impact of rare and common loss-of-function variants on allelic expression exposing stronger allelic bias for rare stop-gain variants and informing the extent to which rare deleterious coding alleles are consistently expressed across tissues. This study demonstrates the potential importance of transcriptome data to the interpretation of pathogenic protein-coding variants.


Asunto(s)
Alelos , Proteínas/genética , Exoma , Humanos , Reacción en Cadena de la Polimerasa
12.
Am J Hum Genet ; 93(4): 641-51, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24075185

RESUMEN

Identifying genomic variation is a crucial step for unraveling the relationship between genotype and phenotype and can yield important insights into human diseases. Prevailing methods rely on cost-intensive whole-genome sequencing (WGS) or whole-exome sequencing (WES) approaches while the identification of genomic variants from often existing RNA sequencing (RNA-seq) data remains a challenge because of the intrinsic complexity in the transcriptome. Here, we present a highly accurate approach termed SNPiR to identify SNPs in RNA-seq data. We applied SNPiR to RNA-seq data of samples for which WGS and WES data are also available and achieved high specificity and sensitivity. Of the SNPs called from the RNA-seq data, >98% were also identified by WGS or WES. Over 70% of all expressed coding variants were identified from RNA-seq, and comparable numbers of exonic variants were identified in RNA-seq and WES. Despite our method's limitation in detecting variants in expressed regions only, our results demonstrate that SNPiR outperforms current state-of-the-art approaches for variant detection from RNA-seq data and offers a cost-effective and reliable alternative for SNP discovery.


Asunto(s)
Exoma , Genoma Humano , Genómica/métodos , Polimorfismo de Nucleótido Simple , ARN/genética , Análisis de Secuencia de ARN/métodos , Exones , Humanos , Sistemas de Lectura Abierta , Sensibilidad y Especificidad
13.
Nat Methods ; 10(2): 128-32, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23291724

RESUMEN

We show that RNA editing sites can be called with high confidence using RNA sequencing data from multiple samples across either individuals or species, without the need for matched genomic DNA sequence. We identified many previously unidentified editing sites in both humans and Drosophila; our results nearly double the known number of human protein recoding events. We also found that human genes harboring conserved editing sites within Alu repeats are enriched for neuronal functions.


Asunto(s)
Edición de ARN , Análisis de Secuencia de ARN/métodos , Adenosina/genética , Elementos Alu , Animales , Biología Computacional/métodos , Drosophila melanogaster/genética , Humanos , Inosina/genética
14.
J Theor Biol ; 388: 96-107, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26506471

RESUMEN

Consider two sites under compensatory fitness interaction, such as a Watson-Crick base pair in an RNA helix or two interacting residues in a protein. A mutation at any one of these two sites may reduce the fitness of an individual. However, fitness may be restored by the occurrence of a second mutation at the other site. Kimura modeled this process using a two-locus haploid fitness scheme with two alleles at each locus. He predicted that compensatory evolution following this model is very rare unless selection against the deleterious single mutations is weak and linkage between the interacting sites is tight. Here we investigate the question whether the rate of compensatory evolution increases if we take the context of the two directly interacting sites into account. By "context", we mean the effect of neighboring sites in an RNA helix. Interaction between the focal pair of sites under consideration and the context may lead to so-called indirect compensation. Thus, extending Kimura's classical model of compensatory evolution, we study the effects of both direct and indirect compensation on the rate of compensatory evolution. It is shown that the effects of indirect compensation are very strong. We find that recombination does not slow down the rate of compensatory evolution as predicted by the classical model. Instead, compensatory substitutions may be relatively frequent, even if linkage between the focal interacting sites is loose, selection against deleterious mutations is strong, and mutation rate is low. We compare our theoretical results with data on RNA secondary structures from vertebrate introns.


Asunto(s)
Algoritmos , Evolución Molecular , Modelos Genéticos , ARN/genética , Alelos , Animales , Emparejamiento Base/genética , Aptitud Genética/genética , Haplotipos , Humanos , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , ARN/química , Selección Genética
15.
Nat Methods ; 9(6): 579-81, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22484847

RESUMEN

We developed a computational framework to robustly identify RNA editing sites using transcriptome and genome deep-sequencing data from the same individual. As compared with previous methods, our approach identified a large number of Alu and non-Alu RNA editing sites with high specificity. We also found that editing of non-Alu sites appears to be dependent on nearby edited Alu sites, possibly through the locally formed double-stranded RNA structure.


Asunto(s)
Elementos Alu , Edición de ARN , Análisis de Secuencia de ARN/métodos , Desaminasas APOBEC-1 , Línea Celular , Citidina Desaminasa/metabolismo , Humanos , Mapeo Nucleótido/métodos , ARN Bicatenario
16.
Cell Stem Cell ; 30(9): 1166-1178.e8, 2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37597516

RESUMEN

The intestinal epithelium has high intrinsic turnover rate, and the precise renewal of the epithelium is dependent on the microenvironment. The intestine is innervated by a dense network of peripheral nerves that controls various aspects of intestinal physiology. However, the role of neurons in regulating epithelial cell regeneration remains largely unknown. Here, we investigated the effects of gut-innervating adrenergic nerves on epithelial cell repair following irradiation (IR)-induced injury. We observed that adrenergic nerve density in the small intestine increased post IR, while chemical adrenergic denervation impaired epithelial regeneration. Single-cell RNA sequencing experiments revealed a decrease in IL-22 signaling post IR in denervated animals. Combining pharmacologic and genetic tools, we demonstrate that ß-adrenergic receptor signaling drives IL-22 production from type 3 innate lymphoid cells (ILC3s) post IR, which in turn promotes epithelial regeneration. These results define an adrenergic-ILC3 axis important for intestinal regeneration.


Asunto(s)
Neuronas Adrenérgicas , Inmunidad Innata , Mucosa Intestinal , Linfocitos , Regeneración , Animales , Transducción de Señal , Neuronas Adrenérgicas/fisiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/inervación , Mucosa Intestinal/fisiología , Ratones , Interleucina-22
17.
Nat Metab ; 5(12): 2094-2110, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38123718

RESUMEN

The thyroid functions as an apex endocrine organ that controls growth, differentiation and metabolism1, and thyroid diseases comprise the most common endocrine disorders2. Nevertheless, high-resolution views of the cellular composition and signals that govern the thyroid have been lacking3,4. Here, we show that Notch signalling controls homeostasis and thermoregulation in adult mammals through a mitochondria-based mechanism in a subset of thyrocytes. We discover two thyrocyte subtypes in mouse and human thyroids, identified in single-cell analyses by different levels of metabolic activity and Notch signalling. Therapeutic antibody blockade of Notch in adult mice inhibits a thyrocyte-specific transcriptional program and induces thyrocyte defects due to decreased mitochondrial activity and ROS production. Thus, disrupting Notch signalling in adult mice causes hypothyroidism, characterized by reduced levels of circulating thyroid hormone and dysregulation of whole-body thermoregulation. Inducible genetic deletion of Notch1 and 2 in thyrocytes phenocopies this antibody-induced hypothyroidism, establishing a direct role for Notch in adult murine thyrocytes. We confirm that hypothyroidism is enriched in children with Alagille syndrome, a genetic disorder marked by Notch mutations, suggesting that these findings translate to humans.


Asunto(s)
Hipotiroidismo , Células Epiteliales Tiroideas , Adulto , Niño , Humanos , Ratones , Animales , Mamíferos , Homeostasis
18.
Blood Adv ; 7(4): 491-507, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35914228

RESUMEN

Self-renewal and differentiation of stem and progenitor cells are tightly regulated to ensure tissue homeostasis. This regulation is enabled both remotely by systemic circulating cues, such as cytokines and hormones, and locally by various niche-confined factors. R-spondin 3 (RSPO3) is one of the most potent enhancers of Wnt signaling, and its expression is usually restricted to the stem cell niche where it provides localized enhancement of Wnt signaling to regulate stem cell expansion and differentiation. Disruption of this niche-confined expression can disturb proper tissue organization and lead to cancers. Here, we investigate the consequences of disrupting the niche-restricted expression of RSPO3 in various tissues, including the hematopoietic system. We show that normal Rspo3 expression is confined to the perivascular niche in the bone marrow. Induction of increased systemic levels of circulating RSPO3 outside of the niche results in prominent loss of early B-cell progenitors and anemia but surprisingly has no effect on hematopoietic stem cells. Using molecular, pharmacologic, and genetic approaches, we show that these RSPO3-induced hematopoietic phenotypes are Wnt and RSPO3 dependent and mediated through noncanonical Wnt signaling. Our study highlights a distinct role for a Wnt/RSPO3 signaling axis in the regulation of hematopoiesis, as well as possible challenges related to therapeutic use of RSPOs for regenerative medicine.


Asunto(s)
Hematopoyesis , Nicho de Células Madre , Hematopoyesis/genética , Células Madre Hematopoyéticas , Diferenciación Celular/genética , Vía de Señalización Wnt/fisiología
19.
Mol Biol Evol ; 28(4): 1519-29, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21172832

RESUMEN

Small noncoding RNAs as well as folded RNA structures in genic regions are crucial for many cellular processes. They are involved in posttranscriptional gene regulation (microRNAs), RNA modification (small nucleolar RNAs), regulation of splicing, correct localization of proteins, and many other processes. In most cases, a distinct secondary structure of the molecule is necessary for its correct function. Hence, selection should act to retain the structure of the molecule, although the underlying sequence is allowed to vary. Here, we present the first genome-wide estimates of selective constraints in folded RNA molecules in the nuclear genomes of drosophilids and hominids. In comparison to putatively neutrally evolving sites, we observe substantially reduced rates of substitutions at paired and unpaired sites of folded molecules. We estimated evolutionary constraints to be in the ranges of (0.974,0.991) and (0.895,1.000) for paired nucleotides in drosophilids and hominids, respectively. These values are significantly higher than for constraints at nonsynonymous sites of protein-coding genes in both genera. Nonetheless, valleys of only moderately reduced fitness (s ≈ 10(-4)) are sufficient to generate the observed fraction of nucleotide changes that are removed by purifying selection. In addition, a comparison of selective coefficients between drosophilids and hominids revealed significantly higher constraints in drosophilids, which can be attributed to the difference in long-term effective population size between these two groups of species. This difference is particularly apparent at the independently evolving (unpaired) sites.


Asunto(s)
Evolución Biológica , Secuencia Conservada/genética , Drosophila/genética , Hominidae/genética , Conformación de Ácido Nucleico , ARN/química , ARN/genética , Animales , Composición de Base , Humanos , Selección Genética
20.
Nat Biotechnol ; 40(1): 86-93, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34531539

RESUMEN

Genetic and non-genetic heterogeneity within cancer cell populations represent major challenges to anticancer therapies. We currently lack robust methods to determine how preexisting and adaptive features affect cellular responses to therapies. Here, by conducting clonal fitness mapping and transcriptional characterization using expressed barcodes and single-cell RNA sequencing (scRNA-seq), we have developed tracking differential clonal response by scRNA-seq (TraCe-seq). TraCe-seq is a method that captures at clonal resolution the origin, fate and differential early adaptive transcriptional programs of cells in a complex population in response to distinct treatments. We used TraCe-seq to benchmark how next-generation dual epidermal growth factor receptor (EGFR) inhibitor-degraders compare to standard EGFR kinase inhibitors in EGFR-mutant lung cancer cells. We identified a loss of antigrowth activity associated with targeted degradation of EGFR protein and an essential role of the endoplasmic reticulum (ER) protein processing pathway in anti-EGFR therapeutic efficacy. Our results suggest that targeted degradation is not always superior to enzymatic inhibition and establish TraCe-seq as an approach to study how preexisting transcriptional programs affect treatment responses.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de la Célula Individual/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA