Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.628
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(4): 897-913.e18, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38280374

RESUMEN

Canonically, the complement system is known for its rapid response to remove microbes in the bloodstream. However, relatively little is known about a functioning complement system on intestinal mucosal surfaces. Herein, we report the local synthesis of complement component 3 (C3) in the gut, primarily by stromal cells. C3 is expressed upon commensal colonization and is regulated by the composition of the microbiota in healthy humans and mice, leading to an individual host's specific luminal C3 levels. The absence of membrane attack complex (MAC) components in the gut ensures that C3 deposition does not result in the lysis of commensals. Pathogen infection triggers the immune system to recruit neutrophils to the infection site for pathogen clearance. Basal C3 levels directly correlate with protection against enteric infection. Our study reveals the gut complement system as an innate immune mechanism acting as a vigilant sentinel that combats pathogens and spares commensals.


Asunto(s)
Complemento C3 , Mucosa Intestinal , Microbiota , Animales , Humanos , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Neutrófilos , Complemento C3/metabolismo , Células del Estroma/metabolismo
2.
Cell ; 185(22): 4190-4205.e25, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36243004

RESUMEN

Neuroepithelial crosstalk is critical for gut physiology. However, the mechanisms by which sensory neurons communicate with epithelial cells to mediate gut barrier protection at homeostasis and during inflammation are not well understood. Here, we find that Nav1.8+CGRP+ nociceptor neurons are juxtaposed with and signal to intestinal goblet cells to drive mucus secretion and gut protection. Nociceptor ablation led to decreased mucus thickness and dysbiosis, while chemogenetic nociceptor activation or capsaicin treatment induced mucus growth. Mouse and human goblet cells expressed Ramp1, receptor for the neuropeptide CGRP. Nociceptors signal via the CGRP-Ramp1 pathway to induce rapid goblet cell emptying and mucus secretion. Notably, commensal microbes activated nociceptors to control homeostatic CGRP release. In the absence of nociceptors or epithelial Ramp1, mice showed increased epithelial stress and susceptibility to colitis. Conversely, CGRP administration protected nociceptor-ablated mice against colitis. Our findings demonstrate a neuron-goblet cell axis that orchestrates gut mucosal barrier protection.


Asunto(s)
Colitis , Células Caliciformes , Ratones , Humanos , Animales , Células Caliciformes/metabolismo , Nociceptores/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Colitis/metabolismo , Moco/metabolismo , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo
3.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32402238

RESUMEN

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Asunto(s)
Sistema Digestivo/inmunología , Linfocitos T Reguladores/inmunología , Animales , Susceptibilidad a Enfermedades/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Homeostasis/inmunología , Inmunoglobulina A/inmunología , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos NOD , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología
4.
Cell ; 180(4): 655-665.e18, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32004463

RESUMEN

Human endocannabinoid systems modulate multiple physiological processes mainly through the activation of cannabinoid receptors CB1 and CB2. Their high sequence similarity, low agonist selectivity, and lack of activation and G protein-coupling knowledge have hindered the development of therapeutic applications. Importantly, missing structural information has significantly held back the development of promising CB2-selective agonist drugs for treating inflammatory and neuropathic pain without the psychoactivity of CB1. Here, we report the cryoelectron microscopy structures of synthetic cannabinoid-bound CB2 and CB1 in complex with Gi, as well as agonist-bound CB2 crystal structure. Of important scientific and therapeutic benefit, our results reveal a diverse activation and signaling mechanism, the structural basis of CB2-selective agonists design, and the unexpected interaction of cholesterol with CB1, suggestive of its endogenous allosteric modulating role.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Receptor Cannabinoide CB1/química , Receptor Cannabinoide CB2/química , Transducción de Señal , Regulación Alostérica , Sitio Alostérico , Animales , Células CHO , Agonistas de Receptores de Cannabinoides/química , Cannabinoides/química , Cannabinoides/farmacología , Línea Celular Tumoral , Colesterol/química , Colesterol/farmacología , Cricetinae , Cricetulus , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Simulación de Dinámica Molecular , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Células Sf9 , Spodoptera
5.
Cell ; 176(3): 459-467.e13, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30639103

RESUMEN

The cannabinoid receptor CB2 is predominately expressed in the immune system, and selective modulation of CB2 without the psychoactivity of CB1 has therapeutic potential in inflammatory, fibrotic, and neurodegenerative diseases. Here, we report the crystal structure of human CB2 in complex with a rationally designed antagonist, AM10257, at 2.8 Å resolution. The CB2-AM10257 structure reveals a distinctly different binding pose compared with CB1. However, the extracellular portion of the antagonist-bound CB2 shares a high degree of conformational similarity with the agonist-bound CB1, which led to the discovery of AM10257's unexpected opposing functional profile of CB2 antagonism versus CB1 agonism. Further structural analysis using mutagenesis studies and molecular docking revealed the molecular basis of their function and selectivity for CB2 and CB1. Additional analyses of our designed antagonist and agonist pairs provide important insight into the activation mechanism of CB2. The present findings should facilitate rational drug design toward precise modulation of the endocannabinoid system.


Asunto(s)
Receptor Cannabinoide CB2/metabolismo , Receptor Cannabinoide CB2/ultraestructura , Animales , Antagonistas de Receptores de Cannabinoides/farmacología , Cannabinoides/farmacología , Diseño de Fármacos , Endocannabinoides , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB2/química , Receptores de Cannabinoides/química , Receptores de Cannabinoides/metabolismo , Receptores de Cannabinoides/ultraestructura , Receptores Acoplados a Proteínas G/metabolismo , Células Sf9 , Relación Estructura-Actividad
6.
Cell ; 170(6): 1247-1257.e12, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28844695

RESUMEN

The respiratory megacomplex represents the highest-order assembly of respiratory chain complexes, and it allows mitochondria to respond to energy-requiring conditions. To understand its architecture, we examined the human respiratory chain megacomplex-I2III2IV2 (MCI2III2IV2) with 140 subunits and a subset of associated cofactors using cryo-electron microscopy. The MCI2III2IV2 forms a circular structure with the dimeric CIII located in the center, where it is surrounded by two copies each of CI and CIV. Two cytochrome c (Cyt.c) molecules are positioned to accept electrons on the surface of the c1 state CIII dimer. Analyses indicate that CII could insert into the gaps between CI and CIV to form a closed ring, which we termed the electron transport chain supercomplex. The structure not only reveals the precise assignment of individual subunits of human CI and CIII, but also enables future in-depth analysis of the electron transport chain as a whole.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/química , Complejos Multienzimáticos/química , Microscopía por Crioelectrón , Proteínas del Complejo de Cadena de Transporte de Electrón/aislamiento & purificación , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/aislamiento & purificación , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/aislamiento & purificación , Complejo II de Transporte de Electrones/metabolismo , Humanos , Mitocondrias/química , Mitocondrias/metabolismo , Modelos Moleculares , Complejos Multienzimáticos/aislamiento & purificación , Complejos Multienzimáticos/metabolismo
7.
Cell ; 167(6): 1598-1609.e10, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27912063

RESUMEN

The mammalian respiratory chain complexes assemble into supercomplexes (SCs) and reside in the inner mitochondrial membrane to transfer electrons and establish the proton gradient for complex V to synthesize ATP. The precise arrangement of SCs is largely unknown. Here, we report a 4.0-Å cryo-electron microscopy (cryo-EM) structure of the major SC in porcine heart, the 1.7-MDa SCI1III2IV1. The complex III (CIII) dimer and complex IV (CIV) bind at the same side of the L-shaped complex I (CI). Several accessory or supernumerary subunits of CI, such as NDUFA11, NDUFB4, NDUFB8, and NDUFB9, directly contribute to the oligomerization of CI, CIII, and CIV. COX7C and COX7A of CIV attach CIV to the concave surface formed by CIII and the distal end of membrane arm of CI. The structure suggests a possible mechanism by which electrons are transferred from NADH to cytochrome c and provides a platform for future functional dissection of respiration.


Asunto(s)
Transporte de Electrón , Mitocondrias Cardíacas/química , Membranas Mitocondriales/química , Animales , Microscopía por Crioelectrón , Modelos Moleculares , Complejos Multienzimáticos/química , Bombas de Protones/química , Sus scrofa
8.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26046436

RESUMEN

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Asunto(s)
Descubrimiento de Drogas , Bibliotecas de Moléculas Pequeñas , Animales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , National Institutes of Health (U.S.) , Estados Unidos
9.
Nature ; 619(7971): 837-843, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380774

RESUMEN

The human gut microbiome constantly converts natural products derived from the host and diet into numerous bioactive metabolites1-3. Dietary fats are essential micronutrients that undergo lipolysis to release free fatty acids (FAs) for absorption in the small intestine4. Gut commensal bacteria modify some unsaturated FAs-for example, linoleic acid (LA)-into various intestinal FA isomers that regulate host metabolism and have anticarcinogenic properties5. However, little is known about how this diet-microorganism FA isomerization network affects the mucosal immune system of the host. Here we report that both dietary factors and microbial factors influence the level of gut LA isomers (conjugated LAs (CLAs)) and that CLAs in turn modulate a distinct population of CD4+ intraepithelial lymphocytes (IELs) that express CD8αα in the small intestine. Genetic abolition of FA isomerization pathways in individual gut symbionts significantly decreases the number of CD4+CD8αα+ IELs in gnotobiotic mice. Restoration of CLAs increases CD4+CD8αα+ IEL levels in the presence of the transcription factor hepatocyte nuclear factor 4γ (HNF4γ). Mechanistically, HNF4γ facilitates CD4+CD8αα+ IEL development by modulating interleukin-18 signalling. In mice, specific deletion of HNF4γ in T cells leads to early mortality from infection by intestinal pathogens. Our data reveal a new role for bacterial FA metabolic pathways in the control of host intraepithelial immunological homeostasis by modulating the relative number of CD4+ T cells that were CD4+CD8αα+.


Asunto(s)
Ácidos Grasos , Microbioma Gastrointestinal , Linfocitos Intraepiteliales , Animales , Humanos , Ratones , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Isomerismo , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Lipólisis , Ácido Linoleico/metabolismo , Inmunidad Mucosa
10.
Nature ; 617(7960): 377-385, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37138075

RESUMEN

The gut microbiota is a crucial regulator of anti-tumour immunity during immune checkpoint inhibitor therapy. Several bacteria that promote an anti-tumour response to immune checkpoint inhibitors have been identified in mice1-6. Moreover, transplantation of faecal specimens from responders can improve the efficacy of anti-PD-1 therapy in patients with melanoma7,8. However, the increased efficacy from faecal transplants is variable and how gut bacteria promote anti-tumour immunity remains unclear. Here we show that the gut microbiome downregulates PD-L2 expression and its binding partner repulsive guidance molecule b (RGMb) to promote anti-tumour immunity and identify bacterial species that mediate this effect. PD-L1 and PD-L2 share PD-1 as a binding partner, but PD-L2 can also bind RGMb. We demonstrate that blockade of PD-L2-RGMb interactions can overcome microbiome-dependent resistance to PD-1 pathway inhibitors. Antibody-mediated blockade of the PD-L2-RGMb pathway or conditional deletion of RGMb in T cells combined with an anti-PD-1 or anti-PD-L1 antibody promotes anti-tumour responses in multiple mouse tumour models that do not respond to anti-PD-1 or anti-PD-L1 alone (germ-free mice, antibiotic-treated mice and even mice colonized with stool samples from a patient who did not respond to treatment). These studies identify downregulation of the PD-L2-RGMb pathway as a specific mechanism by which the gut microbiota can promote responses to PD-1 checkpoint blockade. The results also define a potentially effective immunological strategy for treating patients who do not respond to PD-1 cancer immunotherapy.


Asunto(s)
Resistencia a Antineoplásicos , Inmunoterapia , Melanoma , Microbiota , Animales , Humanos , Ratones , Moléculas de Adhesión Celular Neuronal , Modelos Animales de Enfermedad , Regulación hacia Abajo , Resistencia a Antineoplásicos/efectos de los fármacos , Trasplante de Microbiota Fecal , Vida Libre de Gérmenes , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Melanoma/inmunología , Melanoma/microbiología , Melanoma/terapia , Unión Proteica/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
11.
Nature ; 622(7984): 850-862, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37794185

RESUMEN

Immune checkpoint blockade is effective for some patients with cancer, but most are refractory to current immunotherapies and new approaches are needed to overcome resistance1,2. The protein tyrosine phosphatases PTPN2 and PTPN1 are central regulators of inflammation, and their genetic deletion in either tumour cells or immune cells promotes anti-tumour immunity3-6. However, phosphatases are challenging drug targets; in particular, the active site has been considered undruggable. Here we present the discovery and characterization of ABBV-CLS-484 (AC484), a first-in-class, orally bioavailable, potent PTPN2 and PTPN1 active-site inhibitor. AC484 treatment in vitro amplifies the response to interferon and promotes the activation and function of several immune cell subsets. In mouse models of cancer resistant to PD-1 blockade, AC484 monotherapy generates potent anti-tumour immunity. We show that AC484 inflames the tumour microenvironment and promotes natural killer cell and CD8+ T cell function by enhancing JAK-STAT signalling and reducing T cell dysfunction. Inhibitors of PTPN2 and PTPN1 offer a promising new strategy for cancer immunotherapy and are currently being evaluated in patients with advanced solid tumours (ClinicalTrials.gov identifier NCT04777994 ). More broadly, our study shows that small-molecule inhibitors of key intracellular immune regulators can achieve efficacy comparable to or exceeding that of antibody-based immune checkpoint blockade in preclinical models. Finally, to our knowledge, AC484 represents the first active-site phosphatase inhibitor to enter clinical evaluation for cancer immunotherapy and may pave the way for additional therapeutics that target this important class of enzymes.


Asunto(s)
Inmunoterapia , Neoplasias , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 2 , Animales , Humanos , Ratones , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia/métodos , Interferones/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
12.
Immunity ; 50(3): 692-706.e7, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824326

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a severe form of lung fibrosis with a high mortality rate. However, the etiology of IPF remains unknown. Here, we report that alterations in lung microbiota critically promote pulmonary fibrosis pathogenesis. We found that lung microbiota was dysregulated, and the dysregulated microbiota in turn induced production of interleukin-17B (IL-17B) during bleomycin-induced mouse lung fibrosis. Either lung-microbiota depletion or IL-17B deficiency ameliorated the disease progression. IL-17B cooperated with tumor necrosis factor-α to induce expression of neutrophil-recruiting genes and T helper 17 (Th17)-cell-promoting genes. Three pulmonary commensal microbes, which belong to the genera Bacteroides and Prevotella, were identified to promote fibrotic pathogenesis through IL-17R signaling. We further defined that the outer membrane vesicles (OMVs) that were derived from the identified commensal microbes induced IL-17B production through Toll-like receptor-Myd88 adaptor signaling. Together our data demonstrate that specific pulmonary symbiotic commensals can promote lung fibrosis by regulating a profibrotic inflammatory cytokine network.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/microbiología , Interleucina-17/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Microbiota/fisiología , Animales , Bacteroides/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/metabolismo , Prevotella/metabolismo , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Proc Natl Acad Sci U S A ; 121(6): e2309243121, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38289950

RESUMEN

Staphylococcus aureus skin colonization and eosinophil infiltration are associated with many inflammatory skin disorders, including atopic dermatitis, bullous pemphigoid, Netherton's syndrome, and prurigo nodularis. However, whether there is a relationship between S. aureus and eosinophils and how this interaction influences skin inflammation is largely undefined. We show in a preclinical mouse model that S. aureus epicutaneous exposure induced eosinophil-recruiting chemokines and eosinophil infiltration into the skin. Remarkably, we found that eosinophils had a comparable contribution to the skin inflammation as T cells, in a manner dependent on eosinophil-derived IL-17A and IL-17F production. Importantly, IL-36R signaling induced CCL7-mediated eosinophil recruitment to the inflamed skin. Last, S. aureus proteases induced IL-36α expression in keratinocytes, which promoted infiltration of IL-17-producing eosinophils. Collectively, we uncovered a mechanism for S. aureus proteases to trigger eosinophil-mediated skin inflammation, which has implications in the pathogenesis of inflammatory skin diseases.


Asunto(s)
Dermatitis Atópica , Eosinofilia , Infecciones Estafilocócicas , Animales , Ratones , Eosinófilos/metabolismo , Staphylococcus aureus/metabolismo , Péptido Hidrolasas/metabolismo , Piel/metabolismo , Dermatitis Atópica/metabolismo , Infecciones Estafilocócicas/metabolismo , Celulitis (Flemón)/metabolismo , Celulitis (Flemón)/patología , Inflamación/metabolismo
14.
EMBO Rep ; 25(7): 2914-2949, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38783164

RESUMEN

Neutrophil extracellular traps (NETs) are a key antimicrobial feature of cellular innate immunity mediated by polymorphonuclear neutrophils (PMNs). NETs counteract microbes but are also linked to inflammation in atherosclerosis, arthritis, or psoriasis by unknown mechanisms. Here, we report that NET-associated RNA (naRNA) stimulates further NET formation in naive PMNs via a unique TLR8-NLRP3 inflammasome-dependent pathway. Keratinocytes respond to naRNA with expression of psoriasis-related genes (e.g., IL17, IL36) via atypical NOD2-RIPK signaling. In vivo, naRNA drives temporary skin inflammation, which is drastically ameliorated by genetic ablation of RNA sensing. Unexpectedly, the naRNA-LL37 'composite damage-associated molecular pattern (DAMP)' is pre-stored in resting neutrophil granules, defining sterile NETs as inflammatory webs that amplify neutrophil activation. However, the activity of the naRNA-LL37 DAMP is transient and hence supposedly self-limiting under physiological conditions. Collectively, upon dysregulated NET release like in psoriasis, naRNA sensing may represent both a potential cause of disease and a new intervention target.


Asunto(s)
Alarminas , Catelicidinas , Trampas Extracelulares , Inflamación , Neutrófilos , Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Neutrófilos/inmunología , Inflamación/metabolismo , Inflamación/genética , Animales , Humanos , Ratones , Alarminas/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Queratinocitos/metabolismo , ARN/genética , ARN/metabolismo , Psoriasis/genética , Psoriasis/metabolismo , Psoriasis/patología , Transducción de Señal , Activación Neutrófila/genética , Inmunidad Innata/genética
15.
Nature ; 577(7791): 543-548, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31915378

RESUMEN

Although maternal antibodies protect newborn babies from infection1,2, little is known about how protective antibodies are induced without prior pathogen exposure. Here we show that neonatal mice that lack the capacity to produce IgG are protected from infection with the enteric pathogen enterotoxigenic Escherichia coli by maternal natural IgG antibodies against the maternal microbiota when antibodies are delivered either across the placenta or through breast milk. By challenging pups that were fostered by either maternal antibody-sufficient or antibody-deficient dams, we found that IgG derived from breast milk was crucial for protection against mucosal disease induced by enterotoxigenic E. coli. IgG also provides protection against systemic infection by E. coli. Pups used the neonatal Fc receptor to transfer IgG from milk into serum. The maternal commensal microbiota can induce antibodies that recognize antigens expressed by enterotoxigenic E. coli and other Enterobacteriaceae species. Induction of maternal antibodies against a commensal Pantoea species confers protection against enterotoxigenic E. coli in pups. This role of the microbiota in eliciting protective antibodies to a specific neonatal pathogen represents an important host defence mechanism against infection in neonates.


Asunto(s)
Anticuerpos/inmunología , Escherichia coli Enterotoxigénica/inmunología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Inmunidad Materno-Adquirida/inmunología , Recién Nacido/inmunología , Microbiota/inmunología , Leche Humana/inmunología , Animales , Anticuerpos/sangre , Anticuerpos/metabolismo , Lactancia Materna , Reacciones Cruzadas/inmunología , Infecciones por Escherichia coli/microbiología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Masculino , Ratones , Madres , Pantoea/inmunología , Receptores Fc/inmunología , Receptores Fc/metabolismo , Simbiosis/inmunología
16.
Nature ; 577(7790): 410-415, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31875848

RESUMEN

The metabolic pathways encoded by the human gut microbiome constantly interact with host gene products through numerous bioactive molecules1. Primary bile acids (BAs) are synthesized within hepatocytes and released into the duodenum to facilitate absorption of lipids or fat-soluble vitamins2. Some BAs (approximately 5%) escape into the colon, where gut commensal bacteria convert them into various intestinal BAs2 that are important hormones that regulate host cholesterol metabolism and energy balance via several nuclear receptors and/or G-protein-coupled receptors3,4. These receptors have pivotal roles in shaping host innate immune responses1,5. However, the effect of this host-microorganism biliary network on the adaptive immune system remains poorly characterized. Here we report that both dietary and microbial factors influence the composition of the gut BA pool and modulate an important population of colonic FOXP3+ regulatory T (Treg) cells expressing the transcription factor RORγ. Genetic abolition of BA metabolic pathways in individual gut symbionts significantly decreases this Treg cell population. Restoration of the intestinal BA pool increases colonic RORγ+ Treg cell counts and ameliorates host susceptibility to inflammatory colitis via BA nuclear receptors. Thus, a pan-genomic biliary network interaction between hosts and their bacterial symbionts can control host immunological homeostasis via the resulting metabolites.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Microbioma Gastrointestinal , Homeostasis , Intestinos/inmunología , Intestinos/microbiología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Linfocitos T Reguladores/inmunología , Animales , Ácidos y Sales Biliares/química , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética
17.
Nature ; 585(7823): 135-140, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32610344

RESUMEN

Chemokines and their receptors mediate cell migration, which influences multiple fundamental biological processes and disease conditions such as inflammation and cancer1. Although ample effort has been invested into the structural investigation of the chemokine receptors and receptor-chemokine recognition2-4, less is known about endogenous chemokine-induced receptor activation and G-protein coupling. Here we present the cryo-electron microscopy structures of interleukin-8 (IL-8, also known as CXCL8)-activated human CXC chemokine receptor 2 (CXCR2) in complex with Gi protein, along with a crystal structure of CXCR2 bound to a designed allosteric antagonist. Our results reveal a unique shallow mode of binding between CXCL8 and CXCR2, and also show the interactions between CXCR2 and Gi protein. Further structural analysis of the inactive and active states of CXCR2 reveals a distinct activation process and the competitive small-molecule antagonism of chemokine receptors. In addition, our results provide insights into how a G-protein-coupled receptor is activated by an endogenous protein molecule, which will assist in the rational development of therapeutics that target the chemokine system for better pharmacological profiles.


Asunto(s)
Modelos Moleculares , Receptores de Interleucina-8B/química , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Regulación Alostérica , Sitio Alostérico , Quimiocinas/clasificación , Quimiocinas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Interleucina-8/metabolismo , Unión Proteica , Relación Estructura-Actividad , Especificidad por Sustrato
18.
PLoS Genet ; 19(1): e1010551, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36656838

RESUMEN

Human activities have precipitated a rise in the levels of introgressive gene flow among animals. The investigation of conspecific populations at different time points may shed light on the magnitude of human-mediated introgression. We used the red junglefowl Gallus gallus, the wild ancestral form of the chicken, as our study system. As wild junglefowl and domestic chickens readily admix, conservationists fear that domestic introgression into junglefowl may compromise their wild genotype. By contrasting the whole genomes of 51 chickens with 63 junglefowl from across their natural range, we found evidence of a loss of the wild genotype across the Anthropocene. When comparing against the genomes of junglefowl from approximately a century ago using rigorous ancient-DNA protocols, we discovered that levels of domestic introgression are not equal among and within modern wild populations, with the percentage of domestic ancestry around 20-50%. We identified a number of domestication markers in which chickens are deeply differentiated from historic junglefowl regardless of breed and/or geographic provenance, with eight genes under selection. The latter are involved in pathways dealing with development, reproduction and vision. The wild genotype is an allelic reservoir that holds most of the genetic diversity of G. gallus, a species which is immensely important to human society. Our study provides fundamental genomic infrastructure to assist in efforts to prevent a further loss of the wild genotype through introgression of domestic alleles.


Asunto(s)
Pollos , Genética de Población , Genoma , Animales , Pollos/genética , Flujo Génico , Genoma/genética , Genotipo , Filogenia
19.
N Engl J Med ; 386(2): 116-127, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34942067

RESUMEN

BACKGROUND: Population-based data from the United States on the effectiveness of the three coronavirus disease 2019 (Covid-19) vaccines currently authorized by the Food and Drug Administration are limited. Whether declines in effectiveness are due to waning immunity, the B.1.617.2 (delta) variant of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), or other causes is unknown. METHODS: We used data for 8,690,825 adults in New York State to assess the effectiveness of the BNT162b2, mRNA-1273, and Ad26.COV2.S vaccines against laboratory-confirmed Covid-19 and hospitalization with Covid-19 (i.e., Covid-19 diagnosed at or after admission). We compared cohorts defined according to vaccine product received, age, and month of full vaccination with age-specific unvaccinated cohorts by linking statewide testing, hospital, and vaccine registry databases. We assessed vaccine effectiveness against Covid-19 from May 1 through September 3, 2021, and against hospitalization with Covid-19 from May 1 through August 31, 2021. RESULTS: There were 150,865 cases of Covid-19 and 14,477 hospitalizations with Covid-19. During the week of May 1, 2021, when the delta variant made up 1.8% of the circulating variants, the median vaccine effectiveness against Covid-19 was 91.3% (range, 84.1 to 97.0) for BNT162b2, 96.9% (range, 93.7 to 98.0) for mRNA-1273, and 86.6% (range, 77.8 to 89.7) for Ad26.COV2.S. Subsequently, effectiveness declined contemporaneously in all cohorts, from a median of 93.4% (range, 77.8 to 98.0) during the week of May 1 to a nadir of 73.5% (range, 13.8 to 90.0) around July 10, when the prevalence of the delta variant was 85.3%. By the week of August 28, when the prevalence of the delta variant was 99.6%, the effectiveness was 74.2% (range, 63.4 to 86.8). Effectiveness against hospitalization with Covid-19 among adults 18 to 64 years of age remained almost exclusively greater than 86%, with no apparent time trend. Effectiveness declined from May through August among persons 65 years of age or older who had received BNT162b2 (from 94.8 to 88.6%) or mRNA-1273 (from 97.1 to 93.7%). The effectiveness of Ad26.COV2.S was lower than that of the other vaccines, with no trend observed over time (range, 80.0 to 90.6%). CONCLUSIONS: The effectiveness of the three vaccines against Covid-19 declined after the delta variant became predominant. The effectiveness against hospitalization remained high, with modest declines limited to BNT162b2 and mRNA-1273 recipients 65 years of age or older.


Asunto(s)
Vacuna nCoV-2019 mRNA-1273 , Ad26COVS1 , Vacuna BNT162 , COVID-19/prevención & control , Hospitalización/estadística & datos numéricos , Eficacia de las Vacunas , Adolescente , Adulto , Factores de Edad , Anciano , COVID-19/epidemiología , COVID-19/virología , Estudios de Cohortes , Humanos , Incidencia , Persona de Mediana Edad , New York/epidemiología , SARS-CoV-2 , Adulto Joven
20.
Arterioscler Thromb Vasc Biol ; 44(8): 1784-1798, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38868942

RESUMEN

BACKGROUND: The chromatin-remodeling enzymes BRG1 (brahma-related gene 1) and CHD4 (chromodomain helicase DNA-binding protein 4) independently regulate the transcription of genes critical for vascular development, but their coordinated impact on vessels in late-stage embryos has not been explored. METHODS: In this study, we genetically deleted endothelial Brg1 and Chd4 in mixed background mice (Brg1fl/fl;Chd4fl/fl;VE-Cadherin-Cre), and littermates that were negative for Cre recombinase were used as controls. Tissues were analyzed by immunostaining, immunoblot, and flow cytometry. Quantitative reverse transcription polymerase chain reaction was used to determine gene expression, and chromatin immunoprecipitation revealed gene targets of BRG1 and CHD4 in cultured endothelial cells. RESULTS: We found Brg1/Chd4 double mutants grew normally but died soon after birth with small and compact lungs. Despite having normal cellular composition, distal air sacs of the mutant lungs displayed diminished ECM (extracellular matrix) components and TGFß (transforming growth factor-ß) signaling, which typically promotes ECM synthesis. Transcripts for collagen- and elastin-related genes and the TGFß ligand Tgfb1 were decreased in mutant lung endothelial cells, but genetic deletion of endothelial Tgfb1 failed to recapitulate the small lungs and ECM defects seen in Brg1/Chd4 mutants. We instead found several ECM genes to be direct targets of BRG1 and CHD4 in cultured endothelial cells. CONCLUSIONS: Collectively, our data highlight essential roles for endothelial chromatin-remodeling enzymes in promoting ECM deposition in the distal lung tissue during the saccular stage of embryonic lung development.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas , Células Endoteliales , Regulación del Desarrollo de la Expresión Génica , Pulmón , Proteínas Nucleares , Factores de Transcripción , Animales , ADN Helicasas/metabolismo , ADN Helicasas/genética , ADN Helicasas/deficiencia , Pulmón/embriología , Pulmón/metabolismo , Pulmón/enzimología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Células Endoteliales/metabolismo , Células Endoteliales/enzimología , Ratones Noqueados , Transducción de Señal , Matriz Extracelular/metabolismo , Ratones , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones Endogámicos C57BL , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Fenotipo , Humanos , Organogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA