Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31.620
Filtrar
Más filtros

Publication year range
1.
Cell ; 186(10): 2111-2126.e20, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37172564

RESUMEN

Microglia are specialized brain-resident macrophages that play crucial roles in brain development, homeostasis, and disease. However, until now, the ability to model interactions between the human brain environment and microglia has been severely limited. To overcome these limitations, we developed an in vivo xenotransplantation approach that allows us to study functionally mature human microglia (hMGs) that operate within a physiologically relevant, vascularized immunocompetent human brain organoid (iHBO) model. Our data show that organoid-resident hMGs gain human-specific transcriptomic signatures that closely resemble their in vivo counterparts. In vivo two-photon imaging reveals that hMGs actively engage in surveilling the human brain environment, react to local injuries, and respond to systemic inflammatory cues. Finally, we demonstrate that the transplanted iHBOs developed here offer the unprecedented opportunity to study functional human microglia phenotypes in health and disease and provide experimental evidence for a brain-environment-induced immune response in a patient-specific model of autism with macrocephaly.


Asunto(s)
Microglía , Organoides , Humanos , Encéfalo , Macrófagos , Fenotipo
2.
Cell ; 186(12): 2593-2609.e18, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37209683

RESUMEN

Here, we describe an approach to correct the genetic defect in fragile X syndrome (FXS) via recruitment of endogenous repair mechanisms. A leading cause of autism spectrum disorders, FXS results from epigenetic silencing of FMR1 due to a congenital trinucleotide (CGG) repeat expansion. By investigating conditions favorable to FMR1 reactivation, we find MEK and BRAF inhibitors that induce a strong repeat contraction and full FMR1 reactivation in cellular models. We trace the mechanism to DNA demethylation and site-specific R-loops, which are necessary and sufficient for repeat contraction. A positive feedback cycle comprising demethylation, de novo FMR1 transcription, and R-loop formation results in the recruitment of endogenous DNA repair mechanisms that then drive excision of the long CGG repeat. Repeat contraction is specific to FMR1 and restores the production of FMRP protein. Our study therefore identifies a potential method of treating FXS in the future.


Asunto(s)
Síndrome del Cromosoma X Frágil , Expansión de Repetición de Trinucleótido , Humanos , Estructuras R-Loop , Metilación de ADN , Síndrome del Cromosoma X Frágil/genética , Epigénesis Genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo
3.
Cell ; 185(16): 2961-2974.e19, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35839760

RESUMEN

Wheat crops are frequently devastated by pandemic stripe rust caused by Puccinia striiformis f. sp. tritici (Pst). Here, we identify and characterize a wheat receptor-like cytoplasmic kinase gene, TaPsIPK1, that confers susceptibility to this pathogen. PsSpg1, a secreted fungal effector vital for Pst virulence, can bind TaPsIPK1, enhance its kinase activity, and promote its nuclear localization, where it phosphorylates the transcription factor TaCBF1d for gene regulation. The phosphorylation of TaCBF1d switches its transcriptional activity on the downstream genes. CRISPR-Cas9 inactivation of TaPsIPK1 in wheat confers broad-spectrum resistance against Pst without impacting important agronomic traits in two years of field tests. The disruption of TaPsIPK1 leads to immune priming without constitutive activation of defense responses. Taken together, TaPsIPK1 is a susceptibility gene known to be targeted by rust effectors, and it has great potential for developing durable resistance against rust by genetic modifications.


Asunto(s)
Basidiomycota , Triticum , Basidiomycota/genética , Basidiomycota/metabolismo , Enfermedades de las Plantas , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Triticum/genética , Triticum/metabolismo , Triticum/microbiología , Virulencia/genética
4.
Cell ; 185(23): 4409-4427.e18, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36368308

RESUMEN

Fully understanding autism spectrum disorder (ASD) genetics requires whole-genome sequencing (WGS). We present the latest release of the Autism Speaks MSSNG resource, which includes WGS data from 5,100 individuals with ASD and 6,212 non-ASD parents and siblings (total n = 11,312). Examining a wide variety of genetic variants in MSSNG and the Simons Simplex Collection (SSC; n = 9,205), we identified ASD-associated rare variants in 718/5,100 individuals with ASD from MSSNG (14.1%) and 350/2,419 from SSC (14.5%). Considering genomic architecture, 52% were nuclear sequence-level variants, 46% were nuclear structural variants (including copy-number variants, inversions, large insertions, uniparental isodisomies, and tandem repeat expansions), and 2% were mitochondrial variants. Our study provides a guidebook for exploring genotype-phenotype correlations in families who carry ASD-associated rare variants and serves as an entry point to the expanded studies required to dissect the etiology in the ∼85% of the ASD population that remain idiopathic.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Humanos , Trastorno del Espectro Autista/genética , Predisposición Genética a la Enfermedad , Variaciones en el Número de Copia de ADN/genética , Genómica
5.
Cell ; 185(20): 3705-3719.e14, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179667

RESUMEN

The intestinal microbiota is an important modulator of graft-versus-host disease (GVHD), which often complicates allogeneic hematopoietic stem cell transplantation (allo-HSCT). Broad-spectrum antibiotics such as carbapenems increase the risk for intestinal GVHD, but mechanisms are not well understood. In this study, we found that treatment with meropenem, a commonly used carbapenem, aggravates colonic GVHD in mice via the expansion of Bacteroides thetaiotaomicron (BT). BT has a broad ability to degrade dietary polysaccharides and host mucin glycans. BT in meropenem-treated allogeneic mice demonstrated upregulated expression of enzymes involved in the degradation of mucin glycans. These mice also had thinning of the colonic mucus layer and decreased levels of xylose in colonic luminal contents. Interestingly, oral xylose supplementation significantly prevented thinning of the colonic mucus layer in meropenem-treated mice. Specific nutritional supplementation strategies, including xylose supplementation, may combat antibiotic-mediated microbiome injury to reduce the risk for intestinal GVHD in allo-HSCT patients.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bacteroides , Carbapenémicos/farmacología , Carbapenémicos/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Meropenem , Ratones , Mucinas/metabolismo , Moco/metabolismo , Polisacáridos/metabolismo , Xilosa
6.
Cell ; 184(24): 5916-5931.e17, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34767757

RESUMEN

There is increasing interest in the potential contribution of the gut microbiome to autism spectrum disorder (ASD). However, previous studies have been underpowered and have not been designed to address potential confounding factors in a comprehensive way. We performed a large autism stool metagenomics study (n = 247) based on participants from the Australian Autism Biobank and the Queensland Twin Adolescent Brain project. We found negligible direct associations between ASD diagnosis and the gut microbiome. Instead, our data support a model whereby ASD-related restricted interests are associated with less-diverse diet, and in turn reduced microbial taxonomic diversity and looser stool consistency. In contrast to ASD diagnosis, our dataset was well powered to detect microbiome associations with traits such as age, dietary intake, and stool consistency. Overall, microbiome differences in ASD may reflect dietary preferences that relate to diagnostic features, and we caution against claims that the microbiome has a driving role in ASD.


Asunto(s)
Trastorno Autístico/microbiología , Conducta Alimentaria , Microbioma Gastrointestinal , Adolescente , Factores de Edad , Trastorno Autístico/diagnóstico , Conducta , Niño , Preescolar , Heces/microbiología , Femenino , Humanos , Masculino , Fenotipo , Filogenia , Especificidad de la Especie
7.
Cell ; 184(25): 6037-6051.e14, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34852237

RESUMEN

RNA viruses generate defective viral genomes (DVGs) that can interfere with replication of the parental wild-type virus. To examine their therapeutic potential, we created a DVG by deleting the capsid-coding region of poliovirus. Strikingly, intraperitoneal or intranasal administration of this genome, which we termed eTIP1, elicits an antiviral response, inhibits replication, and protects mice from several RNA viruses, including enteroviruses, influenza, and SARS-CoV-2. While eTIP1 replication following intranasal administration is limited to the nasal cavity, its antiviral action extends non-cell-autonomously to the lungs. eTIP1 broad-spectrum antiviral effects are mediated by both local and distal type I interferon responses. Importantly, while a single eTIP1 dose protects animals from SARS-CoV-2 infection, it also stimulates production of SARS-CoV-2 neutralizing antibodies that afford long-lasting protection from SARS-CoV-2 reinfection. Thus, eTIP1 is a safe and effective broad-spectrum antiviral generating short- and long-term protection against SARS-CoV-2 and other respiratory infections in animal models.


Asunto(s)
Proteínas de la Cápside/genética , Virus Interferentes Defectuosos/metabolismo , Replicación Viral/efectos de los fármacos , Administración Intranasal , Animales , Antivirales/farmacología , Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos ampliamente neutralizantes/farmacología , COVID-19 , Proteínas de la Cápside/metabolismo , Línea Celular , Virus Interferentes Defectuosos/patogenicidad , Modelos Animales de Enfermedad , Genoma Viral/genética , Humanos , Gripe Humana , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Poliovirus/genética , Poliovirus/metabolismo , Infecciones del Sistema Respiratorio/virología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad
8.
Cell ; 184(18): 4772-4783.e15, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34388390

RESUMEN

Throughout development and aging, human cells accumulate mutations resulting in genomic mosaicism and genetic diversity at the cellular level. Mosaic mutations present in the gonads can affect both the individual and the offspring and subsequent generations. Here, we explore patterns and temporal stability of clonal mosaic mutations in male gonads by sequencing ejaculated sperm. Through 300× whole-genome sequencing of blood and sperm from healthy men, we find each ejaculate carries on average 33.3 ± 12.1 (mean ± SD) clonal mosaic variants, nearly all of which are detected in serial sampling, with the majority absent from sampled somal tissues. Their temporal stability and mutational signature suggest origins during embryonic development from a largely immutable stem cell niche. Clonal mosaicism likely contributes a transmissible, predicted pathogenic exonic variant for 1 in 15 men, representing a life-long threat of transmission for these individuals and a significant burden on human population health.


Asunto(s)
Crecimiento y Desarrollo , Mosaicismo , Espermatozoides/metabolismo , Adolescente , Envejecimiento/sangre , Alelos , Células Clonales , Estudios de Cohortes , Humanos , Masculino , Modelos Biológicos , Mutación/genética , Factores de Riesgo , Factores de Tiempo , Adulto Joven
9.
Cell ; 184(19): 5053-5069.e23, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34390642

RESUMEN

Genetic perturbations of cortical development can lead to neurodevelopmental disease, including autism spectrum disorder (ASD). To identify genomic regions crucial to corticogenesis, we mapped the activity of gene-regulatory elements generating a single-cell atlas of gene expression and chromatin accessibility both independently and jointly. This revealed waves of gene regulation by key transcription factors (TFs) across a nearly continuous differentiation trajectory, distinguished the expression programs of glial lineages, and identified lineage-determining TFs that exhibited strong correlation between linked gene-regulatory elements and expression levels. These highly connected genes adopted an active chromatin state in early differentiating cells, consistent with lineage commitment. Base-pair-resolution neural network models identified strong cell-type-specific enrichment of noncoding mutations predicted to be disruptive in a cohort of ASD individuals and identified frequently disrupted TF binding sites. This approach illustrates how cell-type-specific mapping can provide insights into the programs governing human development and disease.


Asunto(s)
Corteza Cerebral/embriología , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Análisis de la Célula Individual , Astrocitos/citología , Diferenciación Celular , Linaje de la Célula/genética , Análisis por Conglomerados , Aprendizaje Profundo , Epigénesis Genética , Lógica Difusa , Glutamatos/metabolismo , Humanos , Mutación/genética , Neuronas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética
10.
Cell ; 180(3): 568-584.e23, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31981491

RESUMEN

We present the largest exome sequencing study of autism spectrum disorder (ASD) to date (n = 35,584 total samples, 11,986 with ASD). Using an enhanced analytical framework to integrate de novo and case-control rare variation, we identify 102 risk genes at a false discovery rate of 0.1 or less. Of these genes, 49 show higher frequencies of disruptive de novo variants in individuals ascertained to have severe neurodevelopmental delay, whereas 53 show higher frequencies in individuals ascertained to have ASD; comparing ASD cases with mutations in these groups reveals phenotypic differences. Expressed early in brain development, most risk genes have roles in regulation of gene expression or neuronal communication (i.e., mutations effect neurodevelopmental and neurophysiological changes), and 13 fall within loci recurrently hit by copy number variants. In cells from the human cortex, expression of risk genes is enriched in excitatory and inhibitory neuronal lineages, consistent with multiple paths to an excitatory-inhibitory imbalance underlying ASD.


Asunto(s)
Trastorno Autístico/genética , Corteza Cerebral/crecimiento & desarrollo , Secuenciación del Exoma/métodos , Regulación del Desarrollo de la Expresión Génica , Neurobiología/métodos , Estudios de Casos y Controles , Linaje de la Célula , Estudios de Cohortes , Exoma , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Masculino , Mutación Missense , Neuronas/metabolismo , Fenotipo , Factores Sexuales , Análisis de la Célula Individual/métodos
11.
Cell ; 183(7): 1742-1756, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33357399

RESUMEN

It is unclear how disease mutations impact intrinsically disordered protein regions (IDRs), which lack a stable folded structure. These mutations, while prevalent in disease, are frequently neglected or annotated as variants of unknown significance. Biomolecular phase separation, a physical process often mediated by IDRs, has increasingly appreciated roles in cellular organization and regulation. We find that autism spectrum disorder (ASD)- and cancer-associated proteins are enriched for predicted phase separation propensities, suggesting that IDR mutations disrupt phase separation in key cellular processes. More generally, we hypothesize that combinations of small-effect IDR mutations perturb phase separation, potentially contributing to "missing heritability" in complex disease susceptibility.


Asunto(s)
Enfermedad/genética , Mutación/genética , Cromatina/metabolismo , Humanos , Proteínas Intrínsecamente Desordenadas/genética , Modelos Biológicos , Proteoma/metabolismo
12.
Cell ; 181(7): 1518-1532.e14, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32497502

RESUMEN

The rise of antibiotic resistance and declining discovery of new antibiotics has created a global health crisis. Of particular concern, no new antibiotic classes have been approved for treating Gram-negative pathogens in decades. Here, we characterize a compound, SCH-79797, that kills both Gram-negative and Gram-positive bacteria through a unique dual-targeting mechanism of action (MoA) with undetectably low resistance frequencies. To characterize its MoA, we combined quantitative imaging, proteomic, genetic, metabolomic, and cell-based assays. This pipeline demonstrates that SCH-79797 has two independent cellular targets, folate metabolism and bacterial membrane integrity, and outperforms combination treatments in killing methicillin-resistant Staphylococcus aureus (MRSA) persisters. Building on the molecular core of SCH-79797, we developed a derivative, Irresistin-16, with increased potency and showed its efficacy against Neisseria gonorrhoeae in a mouse vaginal infection model. This promising antibiotic lead suggests that combining multiple MoAs onto a single chemical scaffold may be an underappreciated approach to targeting challenging bacterial pathogens.


Asunto(s)
Bacterias Gramnegativas/efectos de los fármacos , Pirroles/metabolismo , Pirroles/farmacología , Quinazolinas/metabolismo , Quinazolinas/farmacología , Animales , Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Femenino , Ácido Fólico/metabolismo , Bacterias Grampositivas/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Ovariectomía , Proteómica , Pseudomonas aeruginosa/efectos de los fármacos
13.
Cell ; 178(4): 867-886.e24, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398341

RESUMEN

Somatosensory over-reactivity is common among patients with autism spectrum disorders (ASDs) and is hypothesized to contribute to core ASD behaviors. However, effective treatments for sensory over-reactivity and ASDs are lacking. We found distinct somatosensory neuron pathophysiological mechanisms underlie tactile abnormalities in different ASD mouse models and contribute to some ASD-related behaviors. Developmental loss of ASD-associated genes Shank3 or Mecp2 in peripheral mechanosensory neurons leads to region-specific brain abnormalities, revealing links between developmental somatosensory over-reactivity and the genesis of aberrant behaviors. Moreover, acute treatment with a peripherally restricted GABAA receptor agonist that acts directly on mechanosensory neurons reduced tactile over-reactivity in six distinct ASD models. Chronic treatment of Mecp2 and Shank3 mutant mice improved body condition, some brain abnormalities, anxiety-like behaviors, and some social impairments but not memory impairments, motor deficits, or overgrooming. Our findings reveal a potential therapeutic strategy targeting peripheral mechanosensory neurons to treat tactile over-reactivity and select ASD-related behaviors.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Agonistas del GABA/farmacología , Ácidos Isonicotínicos/farmacología , Fenotipo , Células Receptoras Sensoriales/efectos de los fármacos , Tacto/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Ansiedad/tratamiento farmacológico , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/genética , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Agonistas del GABA/uso terapéutico , Ácidos Isonicotínicos/uso terapéutico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos , Proteínas del Tejido Nervioso/genética , Inhibición Prepulso/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo
14.
Cell ; 177(6): 1600-1618.e17, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31150625

RESUMEN

Autism spectrum disorder (ASD) manifests as alterations in complex human behaviors including social communication and stereotypies. In addition to genetic risks, the gut microbiome differs between typically developing (TD) and ASD individuals, though it remains unclear whether the microbiome contributes to symptoms. We transplanted gut microbiota from human donors with ASD or TD controls into germ-free mice and reveal that colonization with ASD microbiota is sufficient to induce hallmark autistic behaviors. The brains of mice colonized with ASD microbiota display alternative splicing of ASD-relevant genes. Microbiome and metabolome profiles of mice harboring human microbiota predict that specific bacterial taxa and their metabolites modulate ASD behaviors. Indeed, treatment of an ASD mouse model with candidate microbial metabolites improves behavioral abnormalities and modulates neuronal excitability in the brain. We propose that the gut microbiota regulates behaviors in mice via production of neuroactive metabolites, suggesting that gut-brain connections contribute to the pathophysiology of ASD.


Asunto(s)
Trastorno del Espectro Autista/microbiología , Síntomas Conductuales/microbiología , Microbioma Gastrointestinal/fisiología , Animales , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/fisiopatología , Bacterias , Conducta Animal/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Microbiota , Factores de Riesgo
15.
Cell ; 177(2): 243-255.e15, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30827682

RESUMEN

Mammals cannot see light over 700 nm in wavelength. This limitation is due to the physical thermodynamic properties of the photon-detecting opsins. However, the detection of naturally invisible near-infrared (NIR) light is a desirable ability. To break this limitation, we developed ocular injectable photoreceptor-binding upconversion nanoparticles (pbUCNPs). These nanoparticles anchored on retinal photoreceptors as miniature NIR light transducers to create NIR light image vision with negligible side effects. Based on single-photoreceptor recordings, electroretinograms, cortical recordings, and visual behavioral tests, we demonstrated that mice with these nanoantennae could not only perceive NIR light, but also see NIR light patterns. Excitingly, the injected mice were also able to differentiate sophisticated NIR shape patterns. Moreover, the NIR light pattern vision was ambient-daylight compatible and existed in parallel with native daylight vision. This new method will provide unmatched opportunities for a wide variety of emerging bio-integrated nanodevice designs and applications. VIDEO ABSTRACT.


Asunto(s)
Nanopartículas/uso terapéutico , Células Fotorreceptoras de Vertebrados/fisiología , Visión Ocular/fisiología , Animales , Femenino , Rayos Infrarrojos , Inyecciones/métodos , Luz , Masculino , Mamíferos/fisiología , Ratones , Ratones Endogámicos C57BL , Opsinas/metabolismo , Retina/metabolismo , Retina/fisiología , Células Fotorreceptoras Retinianas Conos/fisiología , Visión Ocular/genética
16.
Annu Rev Neurosci ; 47(1): 41-61, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38382543

RESUMEN

To perform computations with the efficiency necessary for animal survival, neocortical microcircuits must be capable of reconfiguring in response to experience, while carefully regulating excitatory and inhibitory connectivity to maintain stable function. This dynamic fine-tuning is accomplished through a rich array of cellular homeostatic plasticity mechanisms that stabilize important cellular and network features such as firing rates, information flow, and sensory tuning properties. Further, these functional network properties can be stabilized by different forms of homeostatic plasticity, including mechanisms that target excitatory or inhibitory synapses, or that regulate intrinsic neuronal excitability. Here we discuss which aspects of neocortical circuit function are under homeostatic control, how this homeostasis is realized on the cellular and molecular levels, and the pathological consequences when circuit homeostasis is impaired. A remaining challenge is to elucidate how these diverse homeostatic mechanisms cooperate within complex circuits to enable them to be both flexible and stable.


Asunto(s)
Encéfalo , Homeostasis , Red Nerviosa , Plasticidad Neuronal , Homeostasis/fisiología , Animales , Humanos , Plasticidad Neuronal/fisiología , Red Nerviosa/fisiología , Encéfalo/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Neocórtex/fisiología
17.
Cell ; 170(1): 114-126.e15, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28666113

RESUMEN

Rice feeds half the world's population, and rice blast is often a destructive disease that results in significant crop loss. Non-race-specific resistance has been more effective in controlling crop diseases than race-specific resistance because of its broad spectrum and durability. Through a genome-wide association study, we report the identification of a natural allele of a C2H2-type transcription factor in rice that confers non-race-specific resistance to blast. A survey of 3,000 sequenced rice genomes reveals that this allele exists in 10% of rice, suggesting that this favorable trait has been selected through breeding. This allele causes a single nucleotide change in the promoter of the bsr-d1 gene, which results in reduced expression of the gene through the binding of the repressive MYB transcription factor and, consequently, an inhibition of H2O2 degradation and enhanced disease resistance. Our discovery highlights this novel allele as a strategy for breeding durable resistance in rice.


Asunto(s)
Oryza/genética , Proteínas de Plantas/genética , Factores de Transcripción/genética , Secuencia de Bases , Cruzamiento , Resistencia a la Enfermedad , Técnicas de Inactivación de Genes , Genoma de Planta , Estudio de Asociación del Genoma Completo , Enfermedades de las Plantas , Regiones Promotoras Genéticas
18.
Immunity ; 55(1): 145-158.e7, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34879222

RESUMEN

Children with autism spectrum disorders often display dysregulated immune responses and related gastrointestinal symptoms. However, the underlying mechanisms leading to the development of both phenotypes have not been elucidated. Here, we show that mouse offspring exhibiting autism-like phenotypes due to prenatal exposure to maternal inflammation were more susceptible to developing intestinal inflammation following challenges later in life. In contrast to its prenatal role in neurodevelopmental phenotypes, interleukin-17A (IL-17A) generated immune-primed phenotypes in offspring through changes in the maternal gut microbiota that led to postnatal alterations in the chromatin landscape of naive CD4+ T cells. The transfer of stool samples from pregnant mice with enhanced IL-17A responses into germ-free dams produced immune-primed phenotypes in offspring. Our study provides mechanistic insights into why children exposed to heightened inflammation in the womb might have an increased risk of developing inflammatory diseases in addition to neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/inmunología , Linfocitos T CD4-Positivos/inmunología , Cromatina/metabolismo , Microbioma Gastrointestinal/inmunología , Inflamación/inmunología , Interleucina-17/metabolismo , Intestinos/inmunología , Trastornos del Neurodesarrollo/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Trastorno del Espectro Autista/microbiología , Niño , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Femenino , Humanos , Inmunización , Inflamación/microbiología , Ratones , Trastornos del Neurodesarrollo/microbiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/microbiología
19.
Cell ; 167(5): 1385-1397.e11, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863250

RESUMEN

The association of histone modification changes with autism spectrum disorder (ASD) has not been systematically examined. We conducted a histone acetylome-wide association study (HAWAS) by performing H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) on 257 postmortem samples from ASD and matched control brains. Despite etiological heterogeneity, ≥68% of syndromic and idiopathic ASD cases shared a common acetylome signature at >5,000 cis-regulatory elements in prefrontal and temporal cortex. Similarly, multiple genes associated with rare genetic mutations in ASD showed common "epimutations." Acetylome aberrations in ASD were not attributable to genetic differentiation at cis-SNPs and highlighted genes involved in synaptic transmission, ion transport, epilepsy, behavioral abnormality, chemokinesis, histone deacetylation, and immunity. By correlating histone acetylation with genotype, we discovered >2,000 histone acetylation quantitative trait loci (haQTLs) in human brain regions, including four candidate causal variants for psychiatric diseases. Due to the relative stability of histone modifications postmortem, we anticipate that the HAWAS approach will be applicable to multiple diseases.


Asunto(s)
Trastorno del Espectro Autista/genética , Cerebelo/metabolismo , Código de Histonas , Corteza Prefrontal/metabolismo , Sitios de Carácter Cuantitativo , Lóbulo Temporal/metabolismo , Acetilación , Trastorno del Espectro Autista/metabolismo , Autopsia , Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
20.
Annu Rev Genet ; 56: 391-422, 2022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36055969

RESUMEN

Recent advances in genomics have revealed a wide spectrum of genetic variants associated with neurodevelopmental disorders at an unprecedented scale. An increasing number of studies have consistently identified mutations-both inherited and de novo-impacting the function of specific brain circuits. This suggests that, during brain development, alterations in distinct neural circuits, cell types, or broad regulatory pathways ultimately shaping synapses might be a dysfunctional process underlying these disorders. Here, we review findings from human studies and animal model research to provide a comprehensive description of synaptic and circuit mechanisms implicated in neurodevelopmental disorders. We discuss how specific synaptic connections might be commonly disrupted in different disorders and the alterations in cognition and behaviors emerging from imbalances in neuronal circuits. Moreover, we review new approaches that have been shown to restore or mitigate dysfunctional processes during specific critical windows of brain development. Considering the heterogeneity of neurodevelopmental disorders, we also highlight the recent progress in developing improved clinical biomarkers and strategies that will help to identify novel therapeutic compounds and opportunities for early intervention.


Asunto(s)
Trastornos del Neurodesarrollo , Animales , Humanos , Trastornos del Neurodesarrollo/genética , Modelos Animales de Enfermedad , Genómica , Mutación , Sinapsis/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda