Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117.069
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 187(6): 1402-1421.e21, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38428422

RESUMO

Neonates are highly susceptible to inflammation and infection. Here, we investigate how late fetal liver (FL) mouse hematopoietic stem and progenitor cells (HSPCs) respond to inflammation, testing the hypothesis that deficits in the engagement of emergency myelopoiesis (EM) pathways limit neutrophil output and contribute to perinatal neutropenia. We show that fetal HSPCs have limited production of myeloid cells at steady state and fail to activate a classical adult-like EM transcriptional program. Moreover, we find that fetal HSPCs can respond to EM-inducing inflammatory stimuli in vitro but are restricted by maternal anti-inflammatory factors, primarily interleukin-10 (IL-10), from activating EM pathways in utero. Accordingly, we demonstrate that the loss of maternal IL-10 restores EM activation in fetal HSPCs but at the cost of fetal demise. These results reveal the evolutionary trade-off inherent in maternal anti-inflammatory responses that maintain pregnancy but render the fetus unresponsive to EM activation signals and susceptible to infection.


Assuntos
Inflamação , Interleucina-10 , Mielopoese , Animais , Camundongos , Gravidez/imunologia , Feto , Hematopoese , Células-Tronco Hematopoéticas/citologia , Inflamação/imunologia , Interleucina-10/imunologia , Animais Recém-Nascidos , Feminino
2.
Cell ; 185(2): 311-327.e24, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35063073

RESUMO

The role of postnatal experience in sculpting cortical circuitry, while long appreciated, is poorly understood at the level of cell types. We explore this in the mouse primary visual cortex (V1) using single-nucleus RNA sequencing, visual deprivation, genetics, and functional imaging. We find that vision selectively drives the specification of glutamatergic cell types in upper layers (L) (L2/3/4), while deeper-layer glutamatergic, GABAergic, and non-neuronal cell types are established prior to eye opening. L2/3 cell types form an experience-dependent spatial continuum defined by the graded expression of ∼200 genes, including regulators of cell adhesion and synapse formation. One of these genes, Igsf9b, a vision-dependent gene encoding an inhibitory synaptic cell adhesion molecule, is required for the normal development of binocular responses in L2/3. In summary, vision preferentially regulates the development of upper-layer glutamatergic cell types through the regulation of cell-type-specific gene expression programs.


Assuntos
Visão Ocular , Córtex Visual/citologia , Córtex Visual/embriologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Ácido Glutâmico/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , RNA-Seq , Transcriptoma/genética , Visão Binocular/genética , Ácido gama-Aminobutírico/metabolismo
3.
Cell ; 184(15): 4048-4063.e32, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34233165

RESUMO

Microglia, the resident immune cells of the brain, have emerged as crucial regulators of synaptic refinement and brain wiring. However, whether the remodeling of distinct synapse types during development is mediated by specialized microglia is unknown. Here, we show that GABA-receptive microglia selectively interact with inhibitory cortical synapses during a critical window of mouse postnatal development. GABA initiates a transcriptional synapse remodeling program within these specialized microglia, which in turn sculpt inhibitory connectivity without impacting excitatory synapses. Ablation of GABAB receptors within microglia impairs this process and leads to behavioral abnormalities. These findings demonstrate that brain wiring relies on the selective communication between matched neuronal and glial cell types.


Assuntos
Microglia/metabolismo , Inibição Neural/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Parvalbuminas/metabolismo , Fenótipo , Receptores de GABA-B/metabolismo , Sinapses/fisiologia , Transcrição Gênica
4.
Cell ; 184(22): 5608-5621.e18, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34637701

RESUMO

Mammals use glabrous (hairless) skin of their hands and feet to navigate and manipulate their environment. Cortical maps of the body surface across species contain disproportionately large numbers of neurons dedicated to glabrous skin sensation, in part reflecting a higher density of mechanoreceptors that innervate these skin regions. Here, we find that disproportionate representation of glabrous skin emerges over postnatal development at the first synapse between peripheral mechanoreceptors and their central targets in the brainstem. Mechanoreceptor synapses undergo developmental refinement that depends on proximity of their terminals to glabrous skin, such that those innervating glabrous skin make synaptic connections that expand their central representation. In mice incapable of sensing gentle touch, mechanoreceptors innervating glabrous skin still make more powerful synapses in the brainstem. We propose that the skin region a mechanoreceptor innervates controls the developmental refinement of its central synapses to shape the representation of touch in the brain.


Assuntos
Tronco Encefálico/metabolismo , Mecanorreceptores/metabolismo , Sinapses/metabolismo , Percepção do Tato/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Canais Iônicos/metabolismo , Camundongos Knockout , Neurônios/metabolismo , Imagem Óptica , Optogenética , Pele/inervação
5.
Cell ; 184(3): 741-758.e17, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33484631

RESUMO

Both transcription and three-dimensional (3D) architecture of the mammalian genome play critical roles in neurodevelopment and its disorders. However, 3D genome structures of single brain cells have not been solved; little is known about the dynamics of single-cell transcriptome and 3D genome after birth. Here, we generated a transcriptome (3,517 cells) and 3D genome (3,646 cells) atlas of the developing mouse cortex and hippocampus by using our high-resolution multiple annealing and looping-based amplification cycles for digital transcriptomics (MALBAC-DT) and diploid chromatin conformation capture (Dip-C) methods and developing multi-omic analysis pipelines. In adults, 3D genome "structure types" delineate all major cell types, with high correlation between chromatin A/B compartments and gene expression. During development, both transcriptome and 3D genome are extensively transformed in the first post-natal month. In neurons, 3D genome is rewired across scales, correlated with gene expression modules, and independent of sensory experience. Finally, we examine allele-specific structure of imprinted genes, revealing local and chromosome (chr)-wide differences. These findings uncover an unknown dimension of neurodevelopment.


Assuntos
Encéfalo/crescimento & desenvolvimento , Genoma , Sensação/genética , Transcrição Gênica , Alelos , Animais , Animais Recém-Nascidos , Linhagem da Célula/genética , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Redes Reguladoras de Genes , Loci Gênicos , Impressão Genômica , Camundongos , Família Multigênica , Neuroglia/metabolismo , Neurônios/metabolismo , Transcriptoma/genética , Córtex Visual/metabolismo
6.
Cell ; 176(5): 947-949, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30794778

RESUMO

The adult mammalian heart is minimally regenerative after injury, whereas neonatal hearts fully recover even after major damage. New work from the Red-Horse and Woo labs (Das et al., 2019) shows that collateral artery formation is a key mechanism contributing to successful regeneration in newborn mice and provides insights into how collateral arteries form.


Assuntos
Miócitos Cardíacos , Regeneração , Animais , Camundongos , Animais Recém-Nascidos , Artérias , Coração , Cavalos
7.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30686582

RESUMO

Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.


Assuntos
Circulação Colateral/fisiologia , Coração/crescimento & desenvolvimento , Regeneração/fisiologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Quimiocina CXCL12/metabolismo , Vasos Coronários/crescimento & desenvolvimento , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/fisiologia , Receptores CXCR4/metabolismo , Transdução de Sinais
8.
Cell ; 179(1): 132-146.e14, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31522887

RESUMO

Oligodendrocytes extend elaborate microtubule arbors that contact up to 50 axon segments per cell, then spiral around myelin sheaths, penetrating from outer to inner layers. However, how they establish this complex cytoarchitecture is unclear. Here, we show that oligodendrocytes contain Golgi outposts, an organelle that can function as an acentrosomal microtubule-organizing center (MTOC). We identify a specific marker for Golgi outposts-TPPP (tubulin polymerization promoting protein)-that we use to purify this organelle and characterize its proteome. In in vitro cell-free assays, recombinant TPPP nucleates microtubules. Primary oligodendrocytes from Tppp knockout (KO) mice have aberrant microtubule branching, mixed microtubule polarity, and shorter myelin sheaths when cultured on 3-dimensional (3D) microfibers. Tppp KO mice exhibit hypomyelination with shorter, thinner myelin sheaths and motor coordination deficits. Together, our data demonstrate that microtubule nucleation outside the cell body at Golgi outposts by TPPP is critical for elongation of the myelin sheath.


Assuntos
Proteínas de Transporte/metabolismo , Complexo de Golgi/metabolismo , Microtúbulos/metabolismo , Bainha de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Proteínas de Transporte/genética , Sistema Livre de Células/metabolismo , Células Cultivadas , Escherichia coli/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Centro Organizador dos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Células Precursoras de Oligodendrócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Tubulina (Proteína)/metabolismo
9.
Cell ; 178(1): 44-59.e7, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31104844

RESUMO

Hypothalamic Agrp neurons regulate food ingestion in adult mice. Whether these neurons are functional before animals start to ingest food is unknown. Here, we studied the functional ontogeny of Agrp neurons during breastfeeding using postnatal day 10 mice. In contrast to adult mice, we show that isolation from the nursing nest, not milk deprivation or ingestion, activated Agrp neurons. Non-nutritive suckling and warm temperatures blunted this effect. Using in vivo fiber photometry, neonatal Agrp neurons showed a rapid increase in activity upon isolation from the nest, an effect rapidly diminished following reunion with littermates. Neonates unable to release GABA from Agrp neurons expressed blunted emission of isolation-induced ultrasonic vocalizations. Chemogenetic overactivation of these neurons further increased emission of these ultrasonic vocalizations, but not milk ingestion. We uncovered important functional properties of hypothalamic Agrp neurons during mouse development, suggesting these neurons facilitate offspring-to-caregiver bonding.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Comportamento Alimentar/fisiologia , Hipotálamo/citologia , Neurônios/metabolismo , Proteína Relacionada com Agouti/genética , Animais , Animais Recém-Nascidos , Ingestão de Alimentos/fisiologia , Comportamento Materno/fisiologia , Camundongos , Camundongos Knockout , Leite , Proteínas Proto-Oncogênicas c-fos/metabolismo , Isolamento Social , Comportamento de Sucção/fisiologia , Temperatura , Vocalização Animal/fisiologia , Ácido gama-Aminobutírico/metabolismo
10.
Cell ; 175(7): 1827-1841.e17, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30550786

RESUMO

Newborn mice emit signals that promote parenting from mothers and fathers but trigger aggressive responses from virgin males. Although pup-directed attacks by males require vomeronasal function, the specific infant cues that elicit this behavior are unknown. We developed a behavioral paradigm based on reconstituted pup cues and showed that discrete infant morphological features combined with salivary chemosignals elicit robust male aggression. Seven vomeronasal receptors were identified based on infant-mediated activity, and the involvement of two receptors, Vmn2r65 and Vmn2r88, in infant-directed aggression was demonstrated by genetic deletion. Using the activation of these receptors as readouts for biochemical fractionation, we isolated two pheromonal compounds, the submandibular gland protein C and hemoglobins. Unexpectedly, none of the identified vomeronasal receptors and associated cues were specific to pups. Thus, infant-mediated aggression by virgin males relies on the recognition of pup's physical traits in addition to parental and infant chemical cues.


Assuntos
Agressão , Órgão Vomeronasal/metabolismo , Animais , Animais Recém-Nascidos , Deleção de Genes , Masculino , Camundongos , Camundongos Mutantes
11.
Cell ; 173(5): 1111-1122.e10, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29606355

RESUMO

The development of interventions to prevent congenital Zika syndrome (CZS) has been limited by the lack of an established nonhuman primate model. Here we show that infection of female rhesus monkeys early in pregnancy with Zika virus (ZIKV) recapitulates many features of CZS in humans. We infected 9 pregnant monkeys with ZIKV, 6 early in pregnancy (weeks 6-7 of gestation) and 3 later in pregnancy (weeks 12-14 of gestation), and compared findings with uninfected controls. 100% (6 of 6) of monkeys infected early in pregnancy exhibited prolonged maternal viremia and fetal neuropathology, including fetal loss, smaller brain size, and histopathologic brain lesions, including microcalcifications, hemorrhage, necrosis, vasculitis, gliosis, and apoptosis of neuroprogenitor cells. High-resolution MRI demonstrated concordant lesions indicative of deep gray matter injury. We also observed spinal, ocular, and neuromuscular pathology. Our data show that vascular compromise and neuroprogenitor cell dysfunction are hallmarks of CZS pathogenesis, suggesting novel strategies to prevent and to treat this disease.


Assuntos
Feto/virologia , Neurônios/patologia , Infecção por Zika virus/patologia , Zika virus/patogenicidade , Animais , Animais Recém-Nascidos , Apoptose , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Calcinose/patologia , Calcinose/veterinária , Feminino , Idade Gestacional , Macaca mulatta , Imageamento por Ressonância Magnética , Necrose , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/virologia , Neurônios/virologia , Gravidez , Índice de Gravidade de Doença , Vasculite/patologia , Vasculite/veterinária , Infecção por Zika virus/veterinária , Infecção por Zika virus/virologia
12.
Cell ; 165(4): 827-41, 2016 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-27153495

RESUMO

To maintain a symbiotic relationship between the host and its resident intestinal microbiota, appropriate mucosal T cell responses to commensal antigens must be established. Mice acquire both IgG and IgA maternally; the former has primarily been implicated in passive immunity to pathogens while the latter mediates host-commensal mutualism. Here, we report the surprising observation that mice generate T cell-independent and largely Toll-like receptor (TLR)-dependent IgG2b and IgG3 antibody responses against their gut microbiota. We demonstrate that maternal acquisition of these antibodies dampens mucosal T follicular helper responses and subsequent germinal center B cell responses following birth. This work reveals a feedback loop whereby T cell-independent, TLR-dependent antibodies limit mucosal adaptive immune responses to newly acquired commensal antigens and uncovers a broader function for maternal IgG.


Assuntos
Animais Recém-Nascidos/imunologia , Microbioma Gastrointestinal , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Leite Humano/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Animais Recém-Nascidos/microbiologia , Linfócitos B/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Receptores Toll-Like/imunologia
13.
Cell ; 166(4): 841-854, 2016 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-27453471

RESUMO

For placental mammals, the transition from the in utero maternal environment to postnatal life requires the activation of thermogenesis to maintain their core temperature. This is primarily accomplished by induction of uncoupling protein 1 (UCP1) in brown and beige adipocytes, the principal sites for uncoupled respiration. Despite its importance, how placental mammals license their thermogenic adipocytes to participate in postnatal uncoupled respiration is not known. Here, we provide evidence that the "alarmin" IL-33, a nuclear cytokine that activates type 2 immune responses, licenses brown and beige adipocytes for uncoupled respiration. We find that, in absence of IL-33 or ST2, beige and brown adipocytes develop normally but fail to express an appropriately spliced form of Ucp1 mRNA, resulting in absence of UCP1 protein and impairment in uncoupled respiration and thermoregulation. Together, these data suggest that IL-33 and ST2 function as a developmental switch to license thermogenesis during the perinatal period. PAPERCLIP.


Assuntos
Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Parto , Termogênese , Adipócitos/metabolismo , Animais , Animais Recém-Nascidos , Respiração Celular , Temperatura Baixa , Feminino , Interleucina-33/genética , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38355799

RESUMO

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Assuntos
Animais Recém-Nascidos , Embrião de Mamíferos , Desenvolvimento Embrionário , Gástrula , Análise de Célula Única , Imagem com Lapso de Tempo , Animais , Feminino , Camundongos , Gravidez , Animais Recém-Nascidos/embriologia , Animais Recém-Nascidos/genética , Diferenciação Celular/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário/genética , Gástrula/citologia , Gástrula/embriologia , Gastrulação/genética , Rim/citologia , Rim/embriologia , Mesoderma/citologia , Mesoderma/enzimologia , Neurônios/citologia , Neurônios/metabolismo , Retina/citologia , Retina/embriologia , Somitos/citologia , Somitos/embriologia , Fatores de Tempo , Fatores de Transcrição/genética , Transcrição Gênica , Especificidade de Órgãos/genética
15.
Nat Immunol ; 18(1): 64-73, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27869817

RESUMO

Atopic dermatitis is increasing worldwide in correlation with air pollution. Various organic components of pollutants activate the transcription factor AhR (aryl hydrocarbon receptor). Through the use of AhR-CA mice, whose keratinocytes express constitutively active AhR and that develop atopic-dermatitis-like phenotypes, we identified Artn as a keratinocyte-specific AhR target gene whose product (the neurotrophic factor artemin) was responsible for epidermal hyper-innervation that led to hypersensitivity to pruritus. The activation of AhR via air pollutants induced expression of artemin, alloknesis, epidermal hyper-innervation and inflammation. AhR activation and ARTN expression were positively correlated in the epidermis of patients with atopic dermatitis. Thus, AhR in keratinocytes senses environmental stimuli and elicits an atopic-dermatitis pathology. We propose a mechanism of air-pollution-induced atopic dermatitis via activation of AhR.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Dermatite Atópica/imunologia , Epiderme/inervação , Queratina-15/metabolismo , Queratinócitos/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Prurido/imunologia , Receptores de Hidrocarboneto Arílico/metabolismo , Poluentes Atmosféricos/efeitos adversos , Animais , Animais Recém-Nascidos , Orientação de Axônios/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Epiderme/patologia , Regulação da Expressão Gênica , Humanos , Queratina-15/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptor EphB2/genética , Receptor EphB2/metabolismo , Receptores de Hidrocarboneto Arílico/genética
16.
Nat Immunol ; 18(6): 622-632, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28459433

RESUMO

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Assuntos
Calgranulina A/imunologia , Calgranulina B/imunologia , Imunidade Inata/imunologia , Monócitos/imunologia , Sepse Neonatal/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Animais Recém-Nascidos , Calgranulina A/efeitos dos fármacos , Calgranulina B/efeitos dos fármacos , Epigênese Genética , Sangue Fetal , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imunidade Inata/efeitos dos fármacos , Immunoblotting , Recém-Nascido , Inflamação , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Monócitos/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Sepse Neonatal/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptor 4 Toll-Like/imunologia
17.
Immunity ; 53(1): 172-186.e6, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32610078

RESUMO

B-1 B cells derive from a developmental program distinct from that of conventional B cells, through B cell receptor (BCR)-dependent positive selection of fetally derived precursors. Here, we used direct labeling of B cells reactive with the N-acetyl-D-glucosamine (GlcNAc)-containing Lancefield group A carbohydrate of Streptococcus pyogenes to study the effects of bacterial antigens on the emergent B-1 B cell clonal repertoire. The number, phenotype, and BCR clonotypes of GlcNAc-reactive B-1 B cells were modulated by neonatal exposure to heat-killed S. pyogenes bacteria. GlcNAc-reactive B-1 clonotypes and serum antibodies were reduced in germ-free mice compared with conventionally raised mice. Colonization of germ-free mice with a conventional microbiota promoted GlcNAc-reactive B-1 B cell development and concomitantly elicited clonally related IgA+ plasma cells in the small intestine. Thus, exposure to microbial antigens in early life determines the clonality of the mature B-1 B cell repertoire and ensuing antibody responses, with implications for vaccination approaches and schedules.


Assuntos
Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Subpopulações de Linfócitos B/imunologia , Polissacarídeos Bacterianos/imunologia , Streptococcus pyogenes/imunologia , Acetilglucosamina/metabolismo , Animais , Animais Recém-Nascidos/imunologia , Vida Livre de Germes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota/imunologia
18.
Immunity ; 52(2): 275-294.e9, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32075728

RESUMO

Type 3 innate lymphoid cells (ILC3s) are critical for lung defense against bacterial pneumonia in the neonatal period, but the signals that guide pulmonary ILC3 development remain unclear. Here, we demonstrated that pulmonary ILC3s descended from ILC precursors that populated a niche defined by fibroblasts in the developing lung. Alveolar fibroblasts produced insulin-like growth factor 1 (IGF1), which instructed expansion and maturation of pulmonary ILC precursors. Conditional ablation of IGF1 in alveolar fibroblasts or deletion of the IGF-1 receptor from ILC precursors interrupted ILC3 biogenesis and rendered newborn mice susceptible to pneumonia. Premature infants with bronchopulmonary dysplasia, characterized by interrupted postnatal alveolar development and increased morbidity to respiratory infections, had reduced IGF1 concentrations and pulmonary ILC3 numbers. These findings indicate that the newborn period is a critical window in pulmonary immunity development, and disrupted lung development in prematurely born infants may have enduring effects on host resistance to respiratory infections.


Assuntos
Imunidade Inata , Fator de Crescimento Insulin-Like I/metabolismo , Pulmão/imunologia , Linfócitos/citologia , Células Epiteliais Alveolares/metabolismo , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/imunologia , Diferenciação Celular , Proliferação de Células , Suscetibilidade a Doenças/imunologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Fator de Crescimento Insulin-Like I/deficiência , Interleucinas/metabolismo , Pulmão/citologia , Pulmão/crescimento & desenvolvimento , Linfócitos/metabolismo , Camundongos , Pneumonia/imunologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Interleucina 22
19.
Cell ; 156(1-2): 123-33, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24439373

RESUMO

Coevolution of beneficial microorganisms with the mammalian intestine fundamentally shapes mammalian physiology. Here, we report that the intestinal microbe Bacteroides fragilis modifies the homeostasis of host invariant natural killer T (iNKT) cells by supplementing the host's endogenous lipid antigen milieu with unique inhibitory sphingolipids. The process occurs early in life and effectively impedes iNKT cell proliferation during neonatal development. Consequently, total colonic iNKT cell numbers are restricted into adulthood, and hosts are protected against experimental iNKT cell-mediated, oxazolone-induced colitis. In studies with neonatal mice lacking access to bacterial sphingolipids, we found that treatment with B. fragilis glycosphingolipids-exemplified by an isolated peak (MW = 717.6) called GSL-Bf717-reduces colonic iNKT cell numbers and confers protection against oxazolone-induced colitis in adulthood. Our results suggest that the distinctive inhibitory capacity of GSL-Bf717 and similar molecules may prove useful in the treatment of autoimmune and allergic disorders in which iNKT cell activation is destructive.


Assuntos
Bacteroides fragilis/metabolismo , Colite/imunologia , Glicoesfingolipídeos/metabolismo , Células T Matadoras Naturais/imunologia , Animais , Animais Recém-Nascidos , Proliferação de Células , Colite/induzido quimicamente , Colite/prevenção & controle , Colo/crescimento & desenvolvimento , Colo/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Células T Matadoras Naturais/citologia , Oxazolona
20.
Cell ; 157(5): 1175-88, 2014 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-24813849

RESUMO

Upon ligand binding, RIPK1 is recruited to tumor necrosis factor receptor superfamily (TNFRSF) and Toll-like receptor (TLR) complexes promoting prosurvival and inflammatory signaling. RIPK1 also directly regulates caspase-8-mediated apoptosis or, if caspase-8 activity is blocked, RIPK3-MLKL-dependent necroptosis. We show that C57BL/6 Ripk1(-/-) mice die at birth of systemic inflammation that was not transferable by the hematopoietic compartment. However, Ripk1(-/-) progenitors failed to engraft lethally irradiated hosts properly. Blocking TNF reversed this defect in emergency hematopoiesis but, surprisingly, Tnfr1 deficiency did not prevent inflammation in Ripk1(-/-) neonates. Deletion of Ripk3 or Mlkl, but not Casp8, prevented extracellular release of the necroptotic DAMP, IL-33, and reduced Myd88-dependent inflammation. Reduced inflammation in the Ripk1(-/-)Ripk3(-/-), Ripk1(-/-)Mlkl(-/-), and Ripk1(-/-)Myd88(-/-) mice prevented neonatal lethality, but only Ripk1(-/-)Ripk3(-/-)Casp8(-/-) mice survived past weaning. These results reveal a key function for RIPK1 in inhibiting necroptosis and, thereby, a role in limiting, not only promoting, inflammation.


Assuntos
Genes Letais , Hematopoese , Inflamação/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Animais Recém-Nascidos , Caspase 8/metabolismo , Morte Celular , Fígado/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA