Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.594
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(14): 3461-3495, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38906136

RESUMEN

Developmental biology-the study of the processes by which cells, tissues, and organisms develop and change over time-has entered a new golden age. After the molecular genetics revolution in the 80s and 90s and the diversification of the field in the early 21st century, we have entered a phase when powerful technologies provide new approaches and open unexplored avenues. Progress in the field has been accelerated by advances in genomics, imaging, engineering, and computational biology and by emerging model systems ranging from tardigrades to organoids. We summarize how revolutionary technologies have led to remarkable progress in understanding animal development. We describe how classic questions in gene regulation, pattern formation, morphogenesis, organogenesis, and stem cell biology are being revisited. We discuss the connections of development with evolution, self-organization, metabolism, time, and ecology. We speculate how developmental biology might evolve in an era of synthetic biology, artificial intelligence, and human engineering.


Asunto(s)
Biología Evolutiva , Animales , Humanos , Evolución Biológica , Genómica , Inteligencia Artificial
2.
Cell ; 185(5): 755-758, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35245477

RESUMEN

Support for basic science has been eclipsed by initiatives aimed at specific medical problems. The latest example is the dismantling of the Skirball Institute at NYU School of Medicine. Here, we reflect on the achievements and mission underlying the Skirball to gain insight into the dividends of maintaining a basic science vision within the academic enterprises.


Asunto(s)
Academias e Institutos , Investigación Biomédica , Facultades de Medicina
3.
Cell ; 184(23): 5728-5739.e16, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34644530

RESUMEN

The cyclic pyrimidines 3',5'-cyclic cytidine monophosphate (cCMP) and 3',5'-cyclic uridine monophosphate (cUMP) have been reported in multiple organisms and cell types. As opposed to the cyclic nucleotides 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP), which are second messenger molecules with well-established regulatory roles across all domains of life, the biological role of cyclic pyrimidines has remained unclear. Here we report that cCMP and cUMP are second messengers functioning in bacterial immunity against viruses. We discovered a family of bacterial pyrimidine cyclase enzymes that specifically synthesize cCMP and cUMP following phage infection and demonstrate that these molecules activate immune effectors that execute an antiviral response. A crystal structure of a uridylate cyclase enzyme from this family explains the molecular mechanism of selectivity for pyrimidines as cyclization substrates. Defense systems encoding pyrimidine cyclases, denoted here Pycsar (pyrimidine cyclase system for antiphage resistance), are widespread in prokaryotes. Our results assign clear biological function to cCMP and cUMP as immunity signaling molecules in bacteria.


Asunto(s)
Bacterias/inmunología , Bacterias/virología , Bacteriófagos/fisiología , CMP Cíclico/metabolismo , Nucleótidos Cíclicos/metabolismo , Uridina Monofosfato/metabolismo , Secuencia de Aminoácidos , Bacterias/genética , Burkholderia/enzimología , CMP Cíclico/química , Ciclización , Escherichia coli/enzimología , Modelos Moleculares , Mutación/genética , Nucleótidos Cíclicos/química , Liasas de Fósforo-Oxígeno/química , Liasas de Fósforo-Oxígeno/metabolismo , Pirimidinas/metabolismo , Uridina Monofosfato/química
4.
Cell ; 183(5): 1151-1161, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33068526

RESUMEN

Transcriptome studies reveal pervasive transcription of complex genomes, such as those of mammals. Despite popular arguments for functionality of most, if not all, of these transcripts, genome-wide analysis of selective constraints indicates that most of the produced RNA are junk. However, junk is not garbage. On the contrary, junk transcripts provide the raw material for the evolution of diverse long non-coding (lnc) RNAs by non-adaptive mechanisms, such as constructive neutral evolution. The generation of many novel functional entities, such as lncRNAs, that fuels organismal complexity does not seem to be driven by strong positive selection. Rather, the weak selection regime that dominates the evolution of most multicellular eukaryotes provides ample material for functional innovation with relatively little adaptation involved.


Asunto(s)
ARN Largo no Codificante/genética , ARN Mensajero/genética , Animales , ADN Intergénico/genética , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Humanos , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética
5.
Cell ; 182(1): 38-49.e17, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32544385

RESUMEN

cGAS/DncV-like nucleotidyltransferase (CD-NTase) enzymes are immune sensors that synthesize nucleotide second messengers and initiate antiviral responses in bacterial and animal cells. Here, we discover Enterobacter cloacae CD-NTase-associated protein 4 (Cap4) as a founding member of a diverse family of >2,000 bacterial receptors that respond to CD-NTase signals. Structures of Cap4 reveal a promiscuous DNA endonuclease domain activated through ligand-induced oligomerization. Oligonucleotide recognition occurs through an appended SAVED domain that is an unexpected fusion of two CRISPR-associated Rossman fold (CARF) subunits co-opted from type III CRISPR immunity. Like a lock and key, SAVED effectors exquisitely discriminate 2'-5'- and 3'-5'-linked bacterial cyclic oligonucleotide signals and enable specific recognition of at least 180 potential nucleotide second messenger species. Our results reveal SAVED CARF family proteins as major nucleotide second messenger receptors in CBASS and CRISPR immune defense and extend the importance of linkage specificity beyond mammalian cGAS-STING signaling.


Asunto(s)
Bacterias/virología , Bacteriófagos/metabolismo , Sistemas CRISPR-Cas , Inmunidad , Oligonucleótidos/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Desoxirribonucleasa I/metabolismo , Ligandos , Mutagénesis/genética , Nucleotidiltransferasas/metabolismo , Unión Proteica , Sistemas de Mensajero Secundario
6.
Cell ; 177(2): 478-491.e20, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30929901

RESUMEN

Genomic studies have identified hundreds of candidate genes near loci associated with risk for schizophrenia. To define candidates and their functions, we mutated zebrafish orthologs of 132 human schizophrenia-associated genes. We created a phenotype atlas consisting of whole-brain activity maps, brain structural differences, and profiles of behavioral abnormalities. Phenotypes were diverse but specific, including altered forebrain development and decreased prepulse inhibition. Exploration of these datasets identified promising candidates in more than 10 gene-rich regions, including the magnesium transporter cnnm2 and the translational repressor gigyf2, and revealed shared anatomical sites of activity differences, including the pallium, hypothalamus, and tectum. Single-cell RNA sequencing uncovered an essential role for the understudied transcription factor znf536 in the development of forebrain neurons implicated in social behavior and stress. This phenotypic landscape of schizophrenia-associated genes prioritizes more than 30 candidates for further study and provides hypotheses to bridge the divide between genetic association and biological mechanism.


Asunto(s)
Esquizofrenia/genética , Esquizofrenia/fisiopatología , Animales , Encéfalo , Corteza Cerebral , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Pez Cebra/genética
7.
Immunity ; 57(5): 973-986.e7, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38697117

RESUMEN

The ubiquitin-binding endoribonuclease N4BP1 potently suppresses cytokine production by Toll-like receptors (TLRs) that signal through the adaptor MyD88 but is inactivated via caspase-8-mediated cleavage downstream of death receptors, TLR3, or TLR4. Here, we examined the mechanism whereby N4BP1 limits inflammatory responses. In macrophages, deletion of N4BP1 prolonged activation of inflammatory gene transcription at late time points after TRIF-independent TLR activation. Optimal suppression of inflammatory cytokines by N4BP1 depended on its ability to bind polyubiquitin chains, as macrophages and mice-bearing inactivating mutations in a ubiquitin-binding motif in N4BP1 displayed increased TLR-induced cytokine production. Deletion of the noncanonical IκB kinases (ncIKKs), Tbk1 and Ikke, or their adaptor Tank phenocopied N4bp1 deficiency and enhanced macrophage responses to TLR1/2, TLR7, or TLR9 stimulation. Mechanistically, N4BP1 acted in concert with the ncIKKs to limit the duration of canonical IκB kinase (IKKα/ß) signaling. Thus, N4BP1 and the ncIKKs serve as an important checkpoint against over-exuberant innate immune responses.


Asunto(s)
Endorribonucleasas , Quinasa I-kappa B , Inflamación , Macrófagos , Ratones Noqueados , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Receptores Toll-Like , Animales , Ratones , Inflamación/inmunología , Inflamación/metabolismo , Receptores Toll-Like/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Ubiquitina/metabolismo , Citocinas/metabolismo , Ratones Endogámicos C57BL , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética
8.
Immunity ; 56(10): 2358-2372.e5, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37699392

RESUMEN

Lung-resident memory B cells (lung-BRMs) differentiate into plasma cells after reinfection, providing enhanced pulmonary protection. Here, we investigated the determinants of lung-BRM differentiation upon influenza infection. Kinetic analyses revealed that influenza nucleoprotein (NP)-specific BRMs preferentially differentiated early after infection and required T follicular helper (Tfh) cell help. BRM differentiation temporally coincided with transient interferon (IFN)-γ production by Tfh cells. Depletion of IFN-γ in Tfh cells prevented lung-BRM differentiation and impaired protection against heterosubtypic infection. IFN-γ was required for expression of the transcription factor T-bet by germinal center (GC) B cells, which promoted differentiation of a CXCR3+ GC B cell subset that were precursors of lung-BRMs and CXCR3+ memory B cells in the mediastinal lymph node. Absence of IFN-γ signaling or T-bet in GC B cells prevented CXCR3+ pre-memory precursor development and hampered CXCR3+ memory B cell differentiation and subsequent lung-BRM responses. Thus, Tfh-cell-derived IFN-γ is critical for lung-BRM development and pulmonary immunity, with implications for vaccination strategies targeting BRMs.


Asunto(s)
Gripe Humana , Linfocitos T Colaboradores-Inductores , Humanos , Interferón gamma/metabolismo , Células B de Memoria , Células T Auxiliares Foliculares/metabolismo , Centro Germinal , Diferenciación Celular , Receptores CXCR3/metabolismo
9.
Immunity ; 56(4): 847-863.e8, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36958335

RESUMEN

Seasonal influenza vaccination elicits hemagglutinin (HA)-specific memory B (Bmem) cells, and although multiple Bmem cell populations have been characterized, considerable heterogeneity exists. We found that HA-specific human Bmem cells differed in the expression of surface marker FcRL5 and transcriptional factor T-bet. FcRL5+T-bet+ Bmem cells were transcriptionally similar to effector-like memory cells, while T-betnegFcRL5neg Bmem cells exhibited stem-like central memory properties. FcRL5+ Bmem cells did not express plasma-cell-commitment factors but did express transcriptional, epigenetic, metabolic, and functional programs that poised these cells for antibody production. Accordingly, HA+ T-bet+ Bmem cells at day 7 post-vaccination expressed intracellular immunoglobulin, and tonsil-derived FcRL5+ Bmem cells differentiated more rapidly into antibody-secreting cells (ASCs) in vitro. The T-bet+ Bmem cell response positively correlated with long-lived humoral immunity, and clonotypes from T-bet+ Bmem cells were represented in the secondary ASC response to repeat vaccination, suggesting that this effector-like population predicts influenza vaccine durability and recall potential.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Humanos , Gripe Humana/prevención & control , Formación de Anticuerpos , Células B de Memoria , Vacunación , Memoria Inmunológica , Anticuerpos Antivirales
10.
Immunity ; 55(3): 494-511.e11, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35263568

RESUMEN

Interleukin (IL)-22 is central to immune defense at barrier sites. We examined the contributions of innate lymphoid cell (ILC) and T cell-derived IL-22 during Citrobacter rodentium (C.r) infection using mice that both report Il22 expression and allow lineage-specific deletion. ILC-derived IL-22 activated STAT3 in C.r-colonized surface intestinal epithelial cells (IECs) but only temporally restrained bacterial growth. T cell-derived IL-22 induced a more robust and extensive activation of STAT3 in IECs, including IECs lining colonic crypts, and T cell-specific deficiency of IL-22 led to pathogen invasion of the crypts and increased mortality. This reflected a requirement for T cell-derived IL-22 for the expression of a host-protective transcriptomic program that included AMPs, neutrophil-recruiting chemokines, and mucin-related molecules, and it restricted IFNγ-induced proinflammatory genes. Our findings demonstrate spatiotemporal differences in the production and action of IL-22 by ILCs and T cells during infection and reveal an indispensable role for IL-22-producing T cells in the protection of the intestinal crypts.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae , Animales , Antibacterianos , Inmunidad Innata , Interleucinas/metabolismo , Mucosa Intestinal , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Linfocitos T/metabolismo , Interleucina-22
11.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38355799

RESUMEN

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.


Asunto(s)
Animales Recién Nacidos , Embrión de Mamíferos , Desarrollo Embrionario , Gástrula , Análisis de la Célula Individual , Imagen de Lapso de Tiempo , Animales , Femenino , Ratones , Embarazo , Animales Recién Nacidos/embriología , Animales Recién Nacidos/genética , Diferenciación Celular/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Desarrollo Embrionario/genética , Gástrula/citología , Gástrula/embriología , Gastrulación/genética , Riñón/citología , Riñón/embriología , Mesodermo/citología , Mesodermo/enzimología , Neuronas/citología , Neuronas/metabolismo , Retina/citología , Retina/embriología , Somitos/citología , Somitos/embriología , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética , Especificidad de Órganos/genética
12.
Immunity ; 53(1): 172-186.e6, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32610078

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Subgrupos de Linfocitos B/inmunología , Polisacáridos Bacterianos/inmunología , Streptococcus pyogenes/inmunología , Acetilglucosamina/metabolismo , Animales , Animales Recién Nacidos/inmunología , Vida Libre de Gérmenes/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/inmunología
13.
Cell ; 159(7): 1698-710, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25497548

RESUMEN

Cells control dynamic transitions in transcript levels by regulating transcription, processing, and/or degradation through an integrated regulatory strategy. Here, we combine RNA metabolic labeling, rRNA-depleted RNA-seq, and DRiLL, a novel computational framework, to quantify the level; editing sites; and transcription, processing, and degradation rates of each transcript at a splice junction resolution during the LPS response of mouse dendritic cells. Four key regulatory strategies, dominated by RNA transcription changes, generate most temporal gene expression patterns. Noncanonical strategies that also employ dynamic posttranscriptional regulation control only a minority of genes, but provide unique signal processing features. We validate Tristetraprolin (TTP) as a major regulator of RNA degradation in one noncanonical strategy. Applying DRiLL to the regulation of noncoding RNAs and to zebrafish embryogenesis demonstrates its broad utility. Our study provides a new quantitative approach to discover transcriptional and posttranscriptional events that control dynamic changes in transcript levels using RNA sequencing data.


Asunto(s)
Simulación por Computador , Células Dendríticas/metabolismo , Análisis de Secuencia de ARN/métodos , Animales , Perfilación de la Expresión Génica/métodos , Cinética , Lipopolisacáridos/metabolismo , Ratones , Procesamiento Postranscripcional del ARN , Estabilidad del ARN , ARN no Traducido/metabolismo , Transcripción Genética , Tristetraprolina/metabolismo , Pez Cebra/embriología
14.
Immunity ; 50(5): 1172-1187.e7, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31076359

RESUMEN

Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.


Asunto(s)
Linfocitos B/inmunología , Interferón gamma/inmunología , Receptores de Interferón/metabolismo , Proteínas de Dominio T Box/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Células Productoras de Anticuerpos/inmunología , Linfocitos B/citología , Diferenciación Celular/inmunología , Células Cultivadas , Cromatina/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/biosíntesis , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Proteínas de Dominio T Box/genética , Receptor de Interferón gamma
15.
Cell ; 154(5): 955-956, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23993087

RESUMEN

Animals are often in discrete behavioral states, but it is unclear how one specific state is generated and opposes alternative states. Flavell et al. now identify molecular and neural components in C. elegans that are involved in the generation of dwelling and roaming states.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Neuropéptidos/metabolismo , Serotonina/metabolismo , Transducción de Señal , Animales
16.
Cell ; 153(3): 550-61, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23622240

RESUMEN

Sharply delineated domains of cell types arise in developing tissues under instruction of inductive signal (morphogen) gradients, which specify distinct cell fates at different signal levels. The translation of a morphogen gradient into discrete spatial domains relies on precise signal responses at stable cell positions. However, cells in developing tissues undergoing morphogenesis and proliferation often experience complex movements, which may affect their morphogen exposure, specification, and positioning. How is a clear pattern achieved with cells moving around? Using in toto imaging of the zebrafish neural tube, we analyzed specification patterns and movement trajectories of neural progenitors. We found that specified progenitors of different fates are spatially mixed following heterogeneous Sonic Hedgehog signaling responses. Cell sorting then rearranges them into sharply bordered domains. Ectopically induced motor neuron progenitors also robustly sort to correct locations. Our results reveal that cell sorting acts to correct imprecision of spatial patterning by noisy inductive signals.


Asunto(s)
Morfogénesis , Células-Madre Neurales/metabolismo , Tubo Neural/citología , Transducción de Señal , Pez Cebra/embriología , Animales , Movimiento Celular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Proteínas Hedgehog/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
17.
Nature ; 608(7924): 803-807, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35859168

RESUMEN

Stimulator of interferon genes (STING) is an antiviral signalling protein that is broadly conserved in both innate immunity in animals and phage defence in prokaryotes1-4. Activation of STING requires its assembly into an oligomeric filament structure through binding of a cyclic dinucleotide4-13, but the molecular basis of STING filament assembly and extension remains unknown. Here we use cryogenic electron microscopy to determine the structure of the active Toll/interleukin-1 receptor (TIR)-STING filament complex from a Sphingobacterium faecium cyclic-oligonucleotide-based antiphage signalling system (CBASS) defence operon. Bacterial TIR-STING filament formation is driven by STING interfaces that become exposed on high-affinity recognition of the cognate cyclic dinucleotide signal c-di-GMP. Repeating dimeric STING units stack laterally head-to-head through surface interfaces, which are also essential for human STING tetramer formation and downstream immune signalling in mammals5. The active bacterial TIR-STING structure reveals further cross-filament contacts that brace the assembly and coordinate packing of the associated TIR NADase effector domains at the base of the filament to drive NAD+ hydrolysis. STING interface and cross-filament contacts are essential for cell growth arrest in vivo and reveal a stepwise mechanism of activation whereby STING filament assembly is required for subsequent effector activation. Our results define the structural basis of STING filament formation in prokaryotic antiviral signalling.


Asunto(s)
Proteínas Bacterianas , Microscopía por Crioelectrón , Proteínas de la Membrana , Receptores de Interleucina-1 , Sphingobacterium , Receptores Toll-Like , Animales , Antivirales/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/inmunología , Fosfatos de Dinucleósidos/metabolismo , Humanos , Inmunidad Innata , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Operón/genética , Receptores de Interleucina-1/química , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/ultraestructura , Sphingobacterium/química , Sphingobacterium/genética , Sphingobacterium/ultraestructura , Sphingobacterium/virología , Receptores Toll-Like/química , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Receptores Toll-Like/ultraestructura
18.
Nature ; 602(7896): 328-335, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34933320

RESUMEN

Mutations in the protein kinase PINK1 lead to defects in mitophagy and cause autosomal recessive early onset Parkinson's disease1,2. PINK1 has many unique features that enable it to phosphorylate ubiquitin and the ubiquitin-like domain of Parkin3-9. Structural analysis of PINK1 from diverse insect species10-12 with and without ubiquitin provided snapshots of distinct structural states yet did not explain how PINK1 is activated. Here we elucidate the activation mechanism of PINK1 using crystallography and cryo-electron microscopy (cryo-EM). A crystal structure of unphosphorylated Pediculus humanus corporis (Ph; human body louse) PINK1 resolves an N-terminal helix, revealing the orientation of unphosphorylated yet active PINK1 on the mitochondria. We further provide a cryo-EM structure of a symmetric PhPINK1 dimer trapped during the process of trans-autophosphorylation, as well as a cryo-EM structure of phosphorylated PhPINK1 undergoing a conformational change to an active ubiquitin kinase state. Structures and phosphorylation studies further identify a role for regulatory PINK1 oxidation. Together, our research delineates the complete activation mechanism of PINK1, illuminates how PINK1 interacts with the mitochondrial outer membrane and reveals how PINK1 activity may be modulated by mitochondrial reactive oxygen species.


Asunto(s)
Proteínas de Insectos , Pediculus , Proteínas Quinasas , Animales , Microscopía por Crioelectrón , Proteínas de Insectos/metabolismo , Mitocondrias , Mitofagia , Fosforilación , Conformación Proteica , Proteínas Quinasas/metabolismo , Ubiquitina/metabolismo
19.
Nat Immunol ; 16(7): 755-65, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26006014

RESUMEN

Acute systemic lupus erythematosus (SLE) courses with surges of antibody-secreting cells (ASCs) whose origin, diversity and contribution to serum autoantibodies remain unknown. Here, deep sequencing, proteomic profiling of autoantibodies and single-cell analysis demonstrated highly diversified ASCs punctuated by clones expressing the variable heavy-chain region VH4-34 that produced dominant serum autoantibodies. A fraction of ASC clones contained autoantibodies without mutation, a finding consistent with differentiation outside the germinal centers. A substantial ASC segment was derived from a distinct subset of newly activated naive cells of considerable clonality that persisted in the circulation for several months. Thus, selection of SLE autoreactivities occurred during polyclonal activation, with prolonged recruitment of recently activated naive B cells. Our findings shed light on the pathogenesis of SLE, help explain the benefit of agents that target B cells and should facilitate the design of future therapies.


Asunto(s)
Diversidad de Anticuerpos/inmunología , Células Productoras de Anticuerpos/inmunología , Autoanticuerpos/inmunología , Proliferación Celular , Lupus Eritematoso Sistémico/inmunología , Enfermedad Aguda , Secuencia de Aminoácidos , Diversidad de Anticuerpos/genética , Células Productoras de Anticuerpos/metabolismo , Autoanticuerpos/genética , Autoanticuerpos/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Células Clonales/inmunología , Células Clonales/metabolismo , Citometría de Flujo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Pesadas de Inmunoglobulina/metabolismo , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/metabolismo , Vacunas contra la Influenza/inmunología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Datos de Secuencia Molecular , Proteoma/análisis , Proteoma/inmunología , Proteómica/métodos , Homología de Secuencia de Aminoácido , Análisis de la Célula Individual/métodos , Espectrometría de Masas en Tándem , Toxoide Tetánico/inmunología
20.
Nature ; 592(7853): 248-252, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33790469

RESUMEN

The archaeological record of Africa provides the earliest evidence for the emergence of the complex symbolic and technological behaviours that characterize Homo sapiens1-7. The coastal setting of many archaeological sites of the Late Pleistocene epoch, and the abundant shellfish remains recovered from them, has led to a dominant narrative in which modern human origins in southern Africa are intrinsically tied to the coast and marine resources8-12, and behavioural innovations in the interior lag behind. However, stratified Late Pleistocene sites with good preservation and robust chronologies are rare in the interior of southern Africa, and the coastal hypothesis therefore remains untested. Here we show that early human innovations that are similar to those dated to around 105 thousand years ago (ka) in coastal southern Africa existed at around the same time among humans who lived over 600 km inland. We report evidence for the intentional collection of non-utilitarian objects (calcite crystals) and ostrich eggshell from excavations of a stratified rockshelter deposit in the southern Kalahari Basin, which we date by optically stimulated luminescence to around 105 ka. Uranium-thorium dating of relict tufa deposits indicates sporadic periods of substantial volumes of fresh, flowing water; the oldest of these episodes is dated to between 110 and 100 ka and is coeval with the archaeological deposit. Our results suggest that behavioural innovations among humans in the interior of southern Africa did not lag behind those of populations near the coast, and that these innovations may have developed within a wet savannah environment. Models that tie the emergence of behavioural innovations to the exploitation of coastal resources by our species may therefore require revision.


Asunto(s)
Arqueología , Carbonato de Calcio/análisis , Cáscara de Huevo , Pradera , Invenciones/historia , Lluvia , Struthioniformes , África Austral , Animales , Carbonato de Calcio/química , Cuevas , Historia Antigua , Humanos , Magnesio , Torio , Uranio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA