Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26.929
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 180(3): 454-470.e18, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32004459

RESUMO

Metagenomic inferences of bacterial strain diversity and infectious disease transmission studies largely assume a dominant, within-individual haplotype. We hypothesize that within-individual bacterial population diversity is critical for homeostasis of a healthy microbiome and infection risk. We characterized the evolutionary trajectory and functional distribution of Staphylococcus epidermidis-a keystone skin microbe and opportunistic pathogen. Analyzing 1,482 S. epidermidis genomes from 5 healthy individuals, we found that skin S. epidermidis isolates coalesce into multiple founder lineages rather than a single colonizer. Transmission events, natural selection, and pervasive horizontal gene transfer result in population admixture within skin sites and dissemination of antibiotic resistance genes within-individual. We provide experimental evidence for how admixture can modulate virulence and metabolism. Leveraging data on the contextual microbiome, we assess how interspecies interactions can shape genetic diversity and mobile gene elements. Our study provides insights into how within-individual evolution of human skin microbes shapes their functional diversification.


Assuntos
Evolução Molecular , Transferência Genética Horizontal , Interações entre Hospedeiro e Microrganismos/genética , Microbiota/genética , Polimorfismo de Nucleotídeo Único , Pele/microbiologia , Staphylococcus epidermidis/genética , Adulto , DNA Bacteriano/genética , Farmacorresistência Bacteriana/genética , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Filogenia , Staphylococcus epidermidis/isolamento & purificação , Staphylococcus epidermidis/patogenicidade , Virulência/genética , Adulto Jovem
2.
Cell ; 181(7): 1661-1679.e22, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32526207

RESUMO

The human gut microbiome harbors hundreds of bacterial species with diverse biochemical capabilities. Dozens of drugs have been shown to be metabolized by single isolates from the gut microbiome, but the extent of this phenomenon is rarely explored in the context of microbial communities. Here, we develop a quantitative experimental framework for mapping the ability of the human gut microbiome to metabolize small molecule drugs: Microbiome-Derived Metabolism (MDM)-Screen. Included are a batch culturing system for sustained growth of subject-specific gut microbial communities, an ex vivo drug metabolism screen, and targeted and untargeted functional metagenomic screens to identify microbiome-encoded genes responsible for specific metabolic events. Our framework identifies novel drug-microbiome interactions that vary between individuals and demonstrates how the gut microbiome might be used in drug development and personalized medicine.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Microbioma Gastrointestinal/fisiologia , Microbiota/efeitos dos fármacos , Adulto , Animais , Bactérias/classificação , Biomarcadores Farmacológicos/metabolismo , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/genética , Voluntários Saudáveis , Humanos , Masculino , Metagenoma/genética , Metagenômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/genética , Preparações Farmacêuticas/metabolismo , Medicina de Precisão/métodos , RNA Ribossômico 16S/genética
3.
Cell ; 179(2): 432-447.e21, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31585082

RESUMO

Cell-cell communication involves a large number of molecular signals that function as words of a complex language whose grammar remains mostly unknown. Here, we describe an integrative approach involving (1) protein-level measurement of multiple communication signals coupled to output responses in receiving cells and (2) mathematical modeling to uncover input-output relationships and interactions between signals. Using human dendritic cell (DC)-T helper (Th) cell communication as a model, we measured 36 DC-derived signals and 17 Th cytokines broadly covering Th diversity in 428 observations. We developed a data-driven, computationally validated model capturing 56 already described and 290 potentially novel mechanisms of Th cell specification. By predicting context-dependent behaviors, we demonstrate a new function for IL-12p70 as an inducer of Th17 in an IL-1 signaling context. This work provides a unique resource to decipher the complex combinatorial rules governing DC-Th cell communication and guide their manipulation for vaccine design and immunotherapies.


Assuntos
Comunicação Celular/imunologia , Células Dendríticas/imunologia , Interleucina-12/fisiologia , Células Th17/imunologia , Adolescente , Adulto , Idoso , Células Cultivadas , Técnicas de Cocultura , Voluntários Saudáveis , Humanos , Interleucina-1/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , Adulto Jovem
4.
Cell ; 178(1): 216-228.e21, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31204103

RESUMO

The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) is the leading target for next-generation vaccines against the disease-causing blood-stage of malaria. However, little is known about how human antibodies confer functional immunity against this antigen. We isolated a panel of human monoclonal antibodies (mAbs) against PfRH5 from peripheral blood B cells from vaccinees in the first clinical trial of a PfRH5-based vaccine. We identified a subset of mAbs with neutralizing activity that bind to three distinct sites and another subset of mAbs that are non-functional, or even antagonistic to neutralizing antibodies. We also identify the epitope of a novel group of non-neutralizing antibodies that significantly reduce the speed of red blood cell invasion by the merozoite, thereby potentiating the effect of all neutralizing PfRH5 antibodies as well as synergizing with antibodies targeting other malaria invasion proteins. Our results provide a roadmap for structure-guided vaccine development to maximize antibody efficacy against blood-stage malaria.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/imunologia , Eritrócitos/parasitologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Adolescente , Adulto , Animais , Sítios de Ligação , Proteínas de Transporte/imunologia , Reações Cruzadas/imunologia , Epitopos/imunologia , Feminino , Células HEK293 , Voluntários Saudáveis , Humanos , Malária Falciparum/parasitologia , Masculino , Merozoítos/fisiologia , Pessoa de Meia-Idade , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/imunologia , Coelhos , Ratos , Ratos Sprague-Dawley , Adulto Jovem
5.
Cell ; 177(2): 446-462.e16, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30951671

RESUMO

Poor reproducibility within and across studies arising from lack of knowledge regarding the performance of extracellular RNA (exRNA) isolation methods has hindered progress in the exRNA field. A systematic comparison of 10 exRNA isolation methods across 5 biofluids revealed marked differences in the complexity and reproducibility of the resulting small RNA-seq profiles. The relative efficiency with which each method accessed different exRNA carrier subclasses was determined by estimating the proportions of extracellular vesicle (EV)-, ribonucleoprotein (RNP)-, and high-density lipoprotein (HDL)-specific miRNA signatures in each profile. An interactive web-based application (miRDaR) was developed to help investigators select the optimal exRNA isolation method for their studies. miRDar provides comparative statistics for all expressed miRNAs or a selected subset of miRNAs in the desired biofluid for each exRNA isolation method and returns a ranked list of exRNA isolation methods prioritized by complexity, expression level, and reproducibility. These results will improve reproducibility and stimulate further progress in exRNA biomarker development.


Assuntos
Ácidos Nucleicos Livres/isolamento & purificação , MicroRNA Circulante/isolamento & purificação , RNA/isolamento & purificação , Adulto , Líquidos Corporais/química , Linhagem Celular , Vesículas Extracelulares/metabolismo , Feminino , Voluntários Saudáveis , Humanos , Masculino , MicroRNAs/isolamento & purificação , MicroRNAs/metabolismo , RNA/metabolismo , Reprodutibilidade dos Testes , Análise de Sequência de RNA/métodos
6.
Cell ; 178(6): 1313-1328.e13, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31491384

RESUMO

Emerging evidence indicates a central role for the microbiome in immunity. However, causal evidence in humans is sparse. Here, we administered broad-spectrum antibiotics to healthy adults prior and subsequent to seasonal influenza vaccination. Despite a 10,000-fold reduction in gut bacterial load and long-lasting diminution in bacterial diversity, antibody responses were not significantly affected. However, in a second trial of subjects with low pre-existing antibody titers, there was significant impairment in H1N1-specific neutralization and binding IgG1 and IgA responses. In addition, in both studies antibiotics treatment resulted in (1) enhanced inflammatory signatures (including AP-1/NR4A expression), observed previously in the elderly, and increased dendritic cell activation; (2) divergent metabolic trajectories, with a 1,000-fold reduction in serum secondary bile acids, which was highly correlated with AP-1/NR4A signaling and inflammasome activation. Multi-omics integration revealed significant associations between bacterial species and metabolic phenotypes, highlighting a key role for the microbiome in modulating human immunity.


Assuntos
Antibacterianos/farmacologia , Anticorpos Antivirais/imunologia , Microbioma Gastrointestinal/fisiologia , Imunidade/efeitos dos fármacos , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Adolescente , Adulto , Formação de Anticorpos , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Imunogenicidade da Vacina/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Masculino , Adulto Jovem
7.
Nat Immunol ; 22(5): 654-665, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33888898

RESUMO

Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.


Assuntos
Imunidade Adaptativa/imunologia , Suscetibilidade a Doenças/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Imunidade Adaptativa/genética , Adolescente , Adulto , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , População Negra/genética , Células Dendríticas/imunologia , Suscetibilidade a Doenças/sangue , Suscetibilidade a Doenças/parasitologia , Feminino , Voluntários Saudáveis , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Interferon gama/metabolismo , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Masculino , RNA-Seq , Análise de Sistemas , Linfócitos T/imunologia , Linfócitos T/metabolismo , População Branca/genética , Adulto Jovem
8.
Nat Immunol ; 20(2): 141-151, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30643265

RESUMO

Rheumatoid arthritis is characterized by progressive joint inflammation and affects ~1% of the human population. We noted single-nucleotide polymorphisms (SNPs) in the apoptotic cell-engulfment genes ELMO1, DOCK2, and RAC1 linked to rheumatoid arthritis. As ELMO1 promotes cytoskeletal reorganization during engulfment, we hypothesized that ELMO1 loss would worsen inflammatory arthritis. Surprisingly, Elmo1-deficient mice showed reduced joint inflammation in acute and chronic arthritis models. Genetic and cell-biology studies revealed that ELMO1 associates with receptors linked to neutrophil function in arthritis and regulates activation and early neutrophil recruitment to the joints, without general inhibition of inflammatory responses. Further, neutrophils from the peripheral blood of human donors that carry the SNP in ELMO1 associated with arthritis display increased migratory capacity, whereas ELMO1 knockdown reduces human neutrophil migration to chemokines linked to arthritis. These data identify 'noncanonical' roles for ELMO1 as an important cytoplasmic regulator of specific neutrophil receptors and promoter of arthritis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Neutrófilos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose/imunologia , Artrite Experimental/diagnóstico , Artrite Experimental/genética , Artrite Experimental/patologia , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Quimiotaxia/genética , Quimiotaxia/imunologia , Colágeno/imunologia , Complemento C5a/imunologia , Complemento C5a/metabolismo , Citoplasma/imunologia , Citoplasma/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Microscopia Intravital , Articulações/citologia , Articulações/imunologia , Leucotrieno B4/imunologia , Leucotrieno B4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Polimorfismo de Nucleotídeo Único , Proteômica , Índice de Gravidade de Doença , Transdução de Sinais/imunologia , Imagem com Lapso de Tempo
9.
Nature ; 633(8030): 654-661, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39261724

RESUMO

Heart failure is a leading cause of morbidity and mortality1,2. Elevated intracardiac pressures and myocyte stretch in heart failure trigger the release of counter-regulatory natriuretic peptides, which act through their receptor (NPR1) to affect vasodilation, diuresis and natriuresis, lowering venous pressures and relieving venous congestion3-8. Recombinant natriuretic peptide infusions were developed to treat heart failure but have been limited by a short duration of effect9,10. Here we report that in a human genetic analysis of over 700,000 individuals, lifelong exposure to coding variants of the NPR1 gene is associated with changes in blood pressure and risk of heart failure. We describe the development of REGN5381, an investigational monoclonal agonist antibody that targets the membrane-bound guanylate cyclase receptor NPR1. REGN5381, an allosteric agonist of NPR1, induces an active-like receptor conformation that results in haemodynamic effects preferentially on venous vasculature, including reductions in systolic blood pressure and venous pressure in animal models. In healthy human volunteers, REGN5381 produced the expected haemodynamic effects, reflecting reductions in venous pressures, without obvious changes in diuresis and natriuresis. These data support the development of REGN5381 for long-lasting and selective lowering of venous pressures that drive symptomatology in patients with heart failure.


Assuntos
Anticorpos Monoclonais , Pressão Sanguínea , Receptores do Fator Natriurético Atrial , Vasoconstrição , Veias , Adulto , Animais , Cães , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Adulto Jovem , Regulação Alostérica/efeitos dos fármacos , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Diurese/efeitos dos fármacos , Voluntários Saudáveis , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Macaca fascicularis , Músculo Liso Vascular/efeitos dos fármacos , Natriurese/efeitos dos fármacos , Receptores do Fator Natriurético Atrial/metabolismo , Receptores do Fator Natriurético Atrial/agonistas , Receptores do Fator Natriurético Atrial/genética , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Veias/efeitos dos fármacos , Veias/fisiologia
10.
Nature ; 632(8023): 131-138, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39020167

RESUMO

A single dose of psilocybin, a psychedelic that acutely causes distortions of space-time perception and ego dissolution, produces rapid and persistent therapeutic effects in human clinical trials1-4. In animal models, psilocybin induces neuroplasticity in cortex and hippocampus5-8. It remains unclear how human brain network changes relate to subjective and lasting effects of psychedelics. Here we tracked individual-specific brain changes with longitudinal precision functional mapping (roughly 18 magnetic resonance imaging visits per participant). Healthy adults were tracked before, during and for 3 weeks after high-dose psilocybin (25 mg) and methylphenidate (40 mg), and brought back for an additional psilocybin dose 6-12 months later. Psilocybin massively disrupted functional connectivity (FC) in cortex and subcortex, acutely causing more than threefold greater change than methylphenidate. These FC changes were driven by brain desynchronization across spatial scales (areal, global), which dissolved network distinctions by reducing correlations within and anticorrelations between networks. Psilocybin-driven FC changes were strongest in the default mode network, which is connected to the anterior hippocampus and is thought to create our sense of space, time and self. Individual differences in FC changes were strongly linked to the subjective psychedelic experience. Performing a perceptual task reduced psilocybin-driven FC changes. Psilocybin caused persistent decrease in FC between the anterior hippocampus and default mode network, lasting for weeks. Persistent reduction of hippocampal-default mode network connectivity may represent a neuroanatomical and mechanistic correlate of the proplasticity and therapeutic effects of psychedelics.


Assuntos
Encéfalo , Alucinógenos , Rede Nervosa , Psilocibina , Adolescente , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Encéfalo/citologia , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Mapeamento Encefálico , Rede de Modo Padrão/citologia , Rede de Modo Padrão/diagnóstico por imagem , Rede de Modo Padrão/efeitos dos fármacos , Rede de Modo Padrão/fisiologia , Alucinógenos/farmacologia , Alucinógenos/administração & dosagem , Voluntários Saudáveis , Hipocampo/citologia , Hipocampo/diagnóstico por imagem , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Imageamento por Ressonância Magnética , Metilfenidato/farmacologia , Metilfenidato/administração & dosagem , Rede Nervosa/citologia , Rede Nervosa/diagnóstico por imagem , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Psilocibina/farmacologia , Psilocibina/administração & dosagem , Percepção Espacial/efeitos dos fármacos , Percepção do Tempo/efeitos dos fármacos , Ego
11.
Nature ; 623(7985): 106-114, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37880365

RESUMO

Maturation of the human fetal brain should follow precisely scheduled structural growth and folding of the cerebral cortex for optimal postnatal function1. We present a normative digital atlas of fetal brain maturation based on a prospective international cohort of healthy pregnant women2, selected using World Health Organization recommendations for growth standards3. Their fetuses were accurately dated in the first trimester, with satisfactory growth and neurodevelopment from early pregnancy to 2 years of age4,5. The atlas was produced using 1,059 optimal quality, three-dimensional ultrasound brain volumes from 899 of the fetuses and an automated analysis pipeline6-8. The atlas corresponds structurally to published magnetic resonance images9, but with finer anatomical details in deep grey matter. The between-study site variability represented less than 8.0% of the total variance of all brain measures, supporting pooling data from the eight study sites to produce patterns of normative maturation. We have thereby generated an average representation of each cerebral hemisphere between 14 and 31 weeks' gestation with quantification of intracranial volume variability and growth patterns. Emergent asymmetries were detectable from as early as 14 weeks, with peak asymmetries in regions associated with language development and functional lateralization between 20 and 26 weeks' gestation. These patterns were validated in 1,487 three-dimensional brain volumes from 1,295 different fetuses in the same cohort. We provide a unique spatiotemporal benchmark of fetal brain maturation from a large cohort with normative postnatal growth and neurodevelopment.


Assuntos
Encéfalo , Desenvolvimento Fetal , Feto , Pré-Escolar , Feminino , Humanos , Gravidez , Encéfalo/anatomia & histologia , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Feto/embriologia , Idade Gestacional , Substância Cinzenta/anatomia & histologia , Substância Cinzenta/embriologia , Substância Cinzenta/crescimento & desenvolvimento , Voluntários Saudáveis , Internacionalidade , Imageamento por Ressonância Magnética , Tamanho do Órgão , Estudos Prospectivos , Organização Mundial da Saúde , Imageamento Tridimensional , Ultrassonografia
12.
Nature ; 614(7949): 752-761, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36599369

RESUMO

Acute viral infections can have durable functional impacts on the immune system long after recovery, but how they affect homeostatic immune states and responses to future perturbations remain poorly understood1-4. Here we use systems immunology approaches, including longitudinal multimodal single-cell analysis (surface proteins, transcriptome and V(D)J sequences) to comparatively assess baseline immune statuses and responses to influenza vaccination in 33 healthy individuals after recovery from mild, non-hospitalized COVID-19 (mean, 151 days after diagnosis) and 40 age- and sex-matched control individuals who had never had COVID-19. At the baseline and independent of time after COVID-19, recoverees had elevated T cell activation signatures and lower expression of innate immune genes including Toll-like receptors in monocytes. Male individuals who had recovered from COVID-19 had coordinately higher innate, influenza-specific plasmablast, and antibody responses after vaccination compared with healthy male individuals and female individuals who had recovered from COVID-19, in part because male recoverees had monocytes with higher IL-15 responses early after vaccination coupled with elevated prevaccination frequencies of 'virtual memory'-like CD8+ T cells poised to produce more IFNγ after IL-15 stimulation. Moreover, the expression of the repressed innate immune genes in monocytes increased by day 1 to day 28 after vaccination in recoverees, therefore moving towards the prevaccination baseline of the healthy control individuals. By contrast, these genes decreased on day 1 and returned to the baseline by day 28 in the control individuals. Our study reveals sex-dimorphic effects of previous mild COVID-19 and suggests that viral infections in humans can establish new immunological set-points that affect future immune responses in an antigen-agnostic manner.


Assuntos
COVID-19 , Imunidade Inata , Memória Imunológica , Vacinas contra Influenza , Caracteres Sexuais , Linfócitos T , Vacinação , Feminino , Humanos , Masculino , Linfócitos T CD8-Positivos/imunologia , COVID-19/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Interleucina-15/imunologia , Receptores Toll-Like/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Monócitos , Imunidade Inata/genética , Imunidade Inata/imunologia , Análise de Célula Única , Voluntários Saudáveis
13.
Nature ; 606(7912): 172-179, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35545680

RESUMO

Missense driver mutations in cancer are concentrated in a few hotspots1. Various mechanisms have been proposed to explain this skew, including biased mutational processes2, phenotypic differences3-6 and immunoediting of neoantigens7,8; however, to our knowledge, no existing model weighs the relative contribution of these features to tumour evolution. We propose a unified theoretical 'free fitness' framework that parsimoniously integrates multimodal genomic, epigenetic, transcriptomic and proteomic data into a biophysical model of the rate-limiting processes underlying the fitness advantage conferred on cancer cells by driver gene mutations. Focusing on TP53, the most mutated gene in cancer1, we present an inference of mutant p53 concentration and demonstrate that TP53 hotspot mutations optimally solve an evolutionary trade-off between oncogenic potential and neoantigen immunogenicity. Our model anticipates patient survival in The Cancer Genome Atlas and patients with lung cancer treated with immunotherapy as well as the age of tumour onset in germline carriers of TP53 variants. The predicted differential immunogenicity between hotspot mutations was validated experimentally in patients with cancer and in a unique large dataset of healthy individuals. Our data indicate that immune selective pressure on TP53 mutations has a smaller role in non-cancerous lesions than in tumours, suggesting that targeted immunotherapy may offer an early prophylactic opportunity for the former. Determining the relative contribution of immunogenicity and oncogenic function to the selective advantage of hotspot mutations thus has important implications for both precision immunotherapies and our understanding of tumour evolution.


Assuntos
Carcinogênese , Evolução Molecular , Neoplasias Pulmonares , Mutação , Carcinogênese/genética , Carcinogênese/imunologia , Conjuntos de Dados como Assunto , Genes p53 , Aptidão Genética , Genômica , Voluntários Saudáveis , Humanos , Imunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Mutação/genética , Mutação de Sentido Incorreto , Reprodutibilidade dos Testes
14.
PLoS Biol ; 22(9): e3002772, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39316644

RESUMO

Dopamine-based reward and learning mechanisms have been suggested to contribute to placebo effects. However, the exact role of dopaminergic neurotransmission in their generation and maintenance is still unclear. This study aimed to shed light on the causal role of dopamine in establishing positive treatment expectations, as well as on the magnitude and duration of their effect on pain. To this end, we used an established placebo analgesia paradigm in combination with 2 opposing pharmacological modulations of dopaminergic tone, i.e., the dopamine antagonist sulpiride and the dopamine precursor L-dopa which were both applied in an experimental, double-blind, randomized, placebo-controlled trial with a between-subject design in N = 168 healthy volunteers. The study medication successfully altered dopaminergic tone during the conditioning procedure. Contrary to our hypotheses, the medication did not modulate the formation of positive treatment expectation and placebo analgesia tested 1 day later. Placebo analgesia was no longer detectable on day 8 after conditioning. Using a combined frequentist and Bayesian approach, our data provide strong evidence against a direct dopaminergic influence on the generation and maintenance of placebo effects. Further exploration of the neurochemical mechanisms underlying placebo analgesia remains paramount in the quest to exploit these effects for optimal treatment outcomes. Trial registration: ClinicalTrials.gov German Clinical Trials Register, ID: DRKS00029366, https://drks.de/search/en/trial/DRKS00029366.


Assuntos
Analgesia , Dopamina , Levodopa , Efeito Placebo , Humanos , Dopamina/metabolismo , Masculino , Adulto , Feminino , Analgesia/métodos , Método Duplo-Cego , Levodopa/farmacologia , Levodopa/uso terapêutico , Adulto Jovem , Sulpirida/farmacologia , Antagonistas de Dopamina/farmacologia , Dor/tratamento farmacológico , Voluntários Saudáveis , Teorema de Bayes
15.
Nature ; 593(7859): 405-410, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33911282

RESUMO

Somatic mutations drive the development of cancer and may contribute to ageing and other diseases1,2. Despite their importance, the difficulty of detecting mutations that are only present in single cells or small clones has limited our knowledge of somatic mutagenesis to a minority of tissues. Here, to overcome these limitations, we developed nanorate sequencing (NanoSeq), a duplex sequencing protocol with error rates of less than five errors per billion base pairs in single DNA molecules from cell populations. This rate is two orders of magnitude lower than typical somatic mutation loads, enabling the study of somatic mutations in any tissue independently of clonality. We used this single-molecule sensitivity to study somatic mutations in non-dividing cells across several tissues, comparing stem cells to differentiated cells and studying mutagenesis in the absence of cell division. Differentiated cells in blood and colon displayed remarkably similar mutation loads and signatures to their corresponding stem cells, despite mature blood cells having undergone considerably more divisions. We then characterized the mutational landscape of post-mitotic neurons and polyclonal smooth muscle, confirming that neurons accumulate somatic mutations at a constant rate throughout life without cell division, with similar rates to mitotically active tissues. Together, our results suggest that mutational processes that are independent of cell division are important contributors to somatic mutagenesis. We anticipate that the ability to reliably detect mutations in single DNA molecules could transform our understanding of somatic mutagenesis and enable non-invasive studies on large-scale cohorts.


Assuntos
Células Sanguíneas/metabolismo , Diferenciação Celular/genética , Análise Mutacional de DNA/métodos , Músculo Liso/metabolismo , Mutação , Neurônios/metabolismo , Imagem Individual de Molécula/métodos , Células-Tronco/metabolismo , Doença de Alzheimer/genética , Células Sanguíneas/citologia , Divisão Celular , Estudos de Coortes , Colo/citologia , Epitélio/metabolismo , Granulócitos/citologia , Granulócitos/metabolismo , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso/citologia , Mutagênese , Taxa de Mutação , Neurônios/citologia , Células-Tronco/citologia
16.
Nature ; 596(7870): 109-113, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34182569

RESUMO

SARS-CoV-2 mRNA-based vaccines are about 95% effective in preventing COVID-191-5. The dynamics of antibody-secreting plasmablasts and germinal centre B cells induced by these vaccines in humans remain unclear. Here we examined antigen-specific B cell responses in peripheral blood (n = 41) and draining lymph nodes in 14 individuals who had received 2 doses of BNT162b2, an mRNA-based vaccine that encodes the full-length SARS-CoV-2 spike (S) gene1. Circulating IgG- and IgA-secreting plasmablasts that target the S protein peaked one week after the second immunization and then declined, becoming undetectable three weeks later. These plasmablast responses preceded maximal levels of serum anti-S binding and neutralizing antibodies to an early circulating SARS-CoV-2 strain as well as emerging variants, especially in individuals who had previously been infected with SARS-CoV-2 (who produced the most robust serological responses). By examining fine needle aspirates of draining axillary lymph nodes, we identified germinal centre B cells that bound S protein in all participants who were sampled after primary immunization. High frequencies of S-binding germinal centre B cells and plasmablasts were sustained in these draining lymph nodes for at least 12 weeks after the booster immunization. S-binding monoclonal antibodies derived from germinal centre B cells predominantly targeted the receptor-binding domain of the S protein, and fewer clones bound to the N-terminal domain or to epitopes shared with the S proteins of the human betacoronaviruses OC43 and HKU1. These latter cross-reactive B cell clones had higher levels of somatic hypermutation as compared to those that recognized only the SARS-CoV-2 S protein, which suggests a memory B cell origin. Our studies demonstrate that SARS-CoV-2 mRNA-based vaccination of humans induces a persistent germinal centre B cell response, which enables the generation of robust humoral immunity.


Assuntos
Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Centro Germinativo/imunologia , Plasmócitos/imunologia , Vacinas Sintéticas/imunologia , Adulto , Idoso , Animais , Anticorpos Antivirais/imunologia , Vacina BNT162 , COVID-19/prevenção & controle , Chlorocebus aethiops , Células Clonais/citologia , Células Clonais/imunologia , Centro Germinativo/citologia , Voluntários Saudáveis , Humanos , Pessoa de Meia-Idade , Plasmócitos/citologia , SARS-CoV-2/imunologia , Fatores de Tempo , Células Vero , Vacinas de mRNA
17.
Nature ; 588(7836): 135-140, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33177712

RESUMO

The serum metabolome contains a plethora of biomarkers and causative agents of various diseases, some of which are endogenously produced and some that have been taken up from the environment1. The origins of specific compounds are known, including metabolites that are highly heritable2,3, or those that are influenced by the gut microbiome4, by lifestyle choices such as smoking5, or by diet6. However, the key determinants of most metabolites are still poorly understood. Here we measured the levels of 1,251 metabolites in serum samples from a unique and deeply phenotyped healthy human cohort of 491 individuals. We applied machine-learning algorithms to predict metabolite levels in held-out individuals on the basis of host genetics, gut microbiome, clinical parameters, diet, lifestyle and anthropometric measurements, and obtained statistically significant predictions for more than 76% of the profiled metabolites. Diet and microbiome had the strongest predictive power, and each explained hundreds of metabolites-in some cases, explaining more than 50% of the observed variance. We further validated microbiome-related predictions by showing a high replication rate in two geographically independent cohorts7,8 that were not available to us when we trained the algorithms. We used feature attribution analysis9 to reveal specific dietary and bacterial interactions. We further demonstrate that some of these interactions might be causal, as some metabolites that we predicted to be positively associated with bread were found to increase after a randomized clinical trial of bread intervention. Overall, our results reveal potential determinants of more than 800 metabolites, paving the way towards a mechanistic understanding of alterations in metabolites under different conditions and to designing interventions for manipulating the levels of circulating metabolites.


Assuntos
Dieta , Microbioma Gastrointestinal/fisiologia , Metaboloma/genética , Soro/metabolismo , Adulto , Pão , Estudos de Coortes , Feminino , Voluntários Saudáveis , Humanos , Estilo de Vida , Aprendizado de Máquina , Masculino , Metabolômica , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/genética , Oxigenases/genética , Padrões de Referência , Reprodutibilidade dos Testes , Estações do Ano
18.
Nature ; 587(7833): 270-274, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32726801

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the rapidly unfolding coronavirus disease 2019 (COVID-19) pandemic1,2. Clinical manifestations of COVID-19 vary, ranging from asymptomatic infection to respiratory failure. The mechanisms that determine such variable outcomes remain unresolved. Here we investigated CD4+ T cells that are reactive against the spike glycoprotein of SARS-CoV-2 in the peripheral blood of patients with COVID-19 and SARS-CoV-2-unexposed healthy donors. We detected spike-reactive CD4+ T cells not only in 83% of patients with COVID-19 but also in 35% of healthy donors. Spike-reactive CD4+ T cells in healthy donors were primarily active against C-terminal epitopes in the spike protein, which show a higher homology to spike glycoproteins of human endemic coronaviruses, compared with N-terminal epitopes. Spike-protein-reactive T cell lines generated from SARS-CoV-2-naive healthy donors responded similarly to the C-terminal region of the spike proteins of the human endemic coronaviruses 229E and OC43, as well as that of SARS-CoV-2. This results indicate that spike-protein cross-reactive T cells are present, which were probably generated during previous encounters with endemic coronaviruses. The effect of pre-existing SARS-CoV-2 cross-reactive T cells on clinical outcomes remains to be determined in larger cohorts. However, the presence of spike-protein cross-reactive T cells in a considerable fraction of the general population may affect the dynamics of the current pandemic, and has important implications for the design and analysis of upcoming trials investigating COVID-19 vaccines.


Assuntos
Betacoronavirus/imunologia , Linfócitos T CD4-Positivos/imunologia , Infecções por Coronavirus/imunologia , Pneumonia Viral/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , COVID-19 , Linhagem Celular , Coronavirus Humano 229E/imunologia , Coronavirus Humano NL63/imunologia , Coronavirus Humano OC43/imunologia , Reações Cruzadas , Epitopos de Linfócito T/imunologia , Feminino , Voluntários Saudáveis , Humanos , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Pandemias , SARS-CoV-2
19.
Arterioscler Thromb Vasc Biol ; 44(9): 2136-2141, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39114916

RESUMO

BACKGROUND: Although artificial and non-nutritive sweeteners are widely used and generally recognized as safe by the US and European Union regulatory agencies, there have been no clinical trials to assess either long-term cardiovascular disease risks or short-term cardiovascular disease-relevant phenotypes. Recent studies report that fasting plasma levels of erythritol, a commonly used sweetener, are clinically associated with heightened incident cardiovascular disease risks and enhance thrombosis potential in vitro and in animal models. Effects of dietary erythritol on thrombosis phenotypes in humans have not been examined. METHODS: Using a prospective interventional study design, we tested the impact of erythritol or glucose consumption on multiple indices of stimulus-dependent platelet responsiveness in healthy volunteers (n=10 per group). Erythritol plasma levels were quantified with liquid chromatography tandem mass spectrometry. Platelet function at baseline and following erythritol or glucose ingestion was assessed via both aggregometry and analysis of granule markers released. RESULTS: Dietary erythritol (30 g), but not glucose (30 g), lead to a >1000-fold increase in erythritol plasma concentration (6480 [5930-7300] versus 3.75 [3.35-3.87] µmol/L; P<0.0001) and exhibited acute enhancement of stimulus-dependent aggregation responses in all subjects, agonists, and doses examined. Erythritol ingestion also enhanced stimulus-dependent release of the platelet dense granule marker serotonin (P<0.0001 for TRAP6 [thrombin activator peptide 6] and P=0.004 for ADP) and the platelet α-granule marker CXCL4 (C-X-C motif ligand-4; P<0.0001 for TRAP6 and P=0.06 for ADP). In contrast, glucose ingestion triggered no significant increases in stimulus-dependent release of either serotonin or CXCL4. CONCLUSIONS: Ingestion of a typical quantity of the non-nutritive sweetener erythritol, but not glucose, enhances platelet reactivity in healthy volunteers, raising concerns that erythritol consumption may enhance thrombosis potential. Combined with recent large-scale clinical observational studies and mechanistic cell-based and animal model studies, the present findings suggest that discussion of whether erythritol should be reevaluated as a food additive with the Generally Recognized as Safe designation is warranted. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT04731363.


Assuntos
Plaquetas , Eritritol , Glucose , Voluntários Saudáveis , Agregação Plaquetária , Trombose , Humanos , Eritritol/sangue , Eritritol/administração & dosagem , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Masculino , Trombose/sangue , Trombose/induzido quimicamente , Trombose/prevenção & controle , Estudos Prospectivos , Agregação Plaquetária/efeitos dos fármacos , Feminino , Adulto , Adoçantes não Calóricos/administração & dosagem , Adoçantes não Calóricos/efeitos adversos , Adulto Jovem , Fator Plaquetário 4/sangue , Espectrometria de Massas em Tandem , Pessoa de Meia-Idade , Serotonina/sangue , Edulcorantes/administração & dosagem , Testes de Função Plaquetária
20.
J Immunol ; 210(6): 753-763, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36734626

RESUMO

OSE-127 is a humanized mAb targeting the IL-7Rα-chain (CD127), under development for inflammatory and autoimmune disease treatment. It is a strict antagonist of the IL-7R pathway, is not internalized by target cells, and is noncytotoxic. In this work, a first-in-human, phase I, randomized, double-blind, placebo-controlled, single-center study was carried out to determine the safety, pharmacokinetics, pharmacodynamics, and immunogenicity of OSE-127 administration. Sixty-three healthy subjects were randomly assigned to nine groups: six single ascending dose groups with i.v. administration (0.002-10 mg/kg), a single s.c. treatment group (1 mg/kg), and two double i.v. injection groups (6 or 10 mg/kg). Subjects were followed during <146 d. OSE-127's pharmacokinetic half-life after a single dose increased from 4.6 (1 mg/kg) to 11.7 d (10 mg/kg) and, after a second dose, from 12.5 (6 mg/kg) to 16.25 d (10 mg/kg). Receptor occupancy was ≥95% at doses ≥0.02 mg/kg, and this saturation level was maintained >100 d after two i.v. infusions at 10 mg/kg. IL-7 consumption was inhibited by OSE-127 administration, as demonstrated by a decreased IL-7 pathway gene signature in peripheral blood cells and by ex vivo T lymphocyte restimulation experiments. OSE-127 was well tolerated, with no evidence of cytokine-release syndrome and no significant alteration of blood lymphocyte counts or subset populations. Altogether, the observed lack of significant lymphopenia or serious adverse events, concomitant with the dose-dependent inhibition of IL-7 consumption by target cells, highlights that OSE-127 may show clinical activity in IL-7R pathway-involved diseases.


Assuntos
Anticorpos Monoclonais , Interleucina-7 , Humanos , Anticorpos Monoclonais/uso terapêutico , Voluntários Saudáveis , Anticorpos Monoclonais Humanizados/farmacologia , Método Duplo-Cego , Relação Dose-Resposta a Droga
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA