Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.189
Filtrar
1.
J Agric Food Chem ; 72(19): 10981-10994, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38691768

RESUMEN

Arctium lappa L. is widely consumed for its various biological effects, and polysaccharides are its main functional components. The present study aimed to evaluate the immunoregulatory effects of the main polysaccharides from burdock (ALP-1) and reveal the underlying mechanisms. ALP-1 consisted of fructose and glucose (14.57:1) and had a molecular weight of 2757 Da, with typical characteristics of (1 → 2)-linked linear fructans. Oral intake of ALP-1 significantly increased the number of colonic goblet cells, serum immunoglobulin A and immunoglobulin G levels, and fecal secretory immunoglobulin A content as well as up-regulated antioxidant enzymes and increased short chain fatty acid production. In addition, ALP-1 administration regulated pro/anti-inflammatory cytokines (i.e., interleukin (IL)-1ß, IL-6, tumor necrosis factor-α, interferon-γ, and IL-10), intestinal microbiota structure, and the spatial information on key metabolites. Some gut-microbiota-mediated metabolic processes were also significantly altered. These results indicated that ALP-1 could exert beneficial effects on immune responses and intestinal health in healthy mice.


Asunto(s)
Arctium , Fructanos , Microbioma Gastrointestinal , Extractos Vegetales , Arctium/química , Animales , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Fructanos/farmacología , Fructanos/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/inmunología , Bacterias/aislamiento & purificación , Bacterias/genética , Masculino , Metabolómica , Humanos , Citocinas/metabolismo , Citocinas/inmunología , Inmunoglobulina A/inmunología
2.
Gut Microbes ; 16(1): 2350156, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726597

RESUMEN

Extensive research has explored the role of gut microbiota in colorectal cancer (CRC). Nonetheless, metatranscriptomic studies investigating the in situ functional implications of host-microbe interactions in CRC are scarce. Therefore, we characterized the influence of CRC core pathogens and biofilms on the tumor microenvironment (TME) in 40 CRC, paired normal, and healthy tissue biopsies using fluorescence in situ hybridization (FISH) and dual-RNA sequencing. FISH revealed that Fusobacterium spp. was associated with increased bacterial biomass and inflammatory response in CRC samples. Dual-RNA sequencing demonstrated increased expression of pro-inflammatory cytokines, defensins, matrix-metalloproteases, and immunomodulatory factors in CRC samples with high bacterial activity. In addition, bacterial activity correlated with the infiltration of several immune cell subtypes, including M2 macrophages and regulatory T-cells in CRC samples. Specifically, Bacteroides fragilis and Fusobacterium nucleatum correlated with the infiltration of neutrophils and CD4+ T-cells, respectively. The collective bacterial activity/biomass appeared to exert a more significant influence on the TME than core pathogens, underscoring the intricate interplay between gut microbiota and CRC. These results emphasize how biofilms and core pathogens shape the immune phenotype and TME in CRC while highlighting the need to extend the bacterial scope beyond CRC pathogens to advance our understanding and identify treatment targets.


Asunto(s)
Biopelículas , Neoplasias Colorrectales , Microbioma Gastrointestinal , Microambiente Tumoral , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Humanos , Biopelículas/crecimiento & desarrollo , Microambiente Tumoral/inmunología , Masculino , Femenino , Bacterias/clasificación , Bacterias/genética , Bacterias/inmunología , Persona de Mediana Edad , Hibridación Fluorescente in Situ , Anciano , Fusobacterium nucleatum/inmunología , Citocinas/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Fenotipo , Bacteroides fragilis/inmunología , Bacteroides fragilis/fisiología , Bacteroides fragilis/genética
4.
Front Immunol ; 15: 1323723, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38650928

RESUMEN

Introduction: The gut microbiota, T cell subsets, and cytokines participate in tuberculosis (TB) pathogenesis. To date, the mechanisms by which these factors interactively promote TB development at different time points remain largely unclear. In the context of this study, We looked into the microorganisms in the digestive tract, T cell types, and cytokines related to tuberculosis. Methods: According to QIIME2, we analyzed 16SrDNA sequencing of the gut microbiome on the Illumina MiSeq. Enzyme-linked immunosorbent assay was used to measure the concentrations of cytokines. Results: We showed the presence of 26 identifiable differential microbiomes in the gut and 44 metabolic pathways between healthy controls and the different time points in the development of TB in patients. Five bacterial genera (Bacteroides, Bifidobacterium, Faecalibacterium, Collinsella, and Clostridium) were most closely associated with CD4/CD8, whereas three bacterial taxa (Faecalibacterium, Collinsella, and Clostridium) were most closely associated with CD4. Three bacterial taxa (Faecalibacterium, Ruminococcus, and Dorea) were most closely associated with IL-4. Ruminococcus was most closely associated with IL-2 and IL-10. Conclusion: Diverse microorganisms, subsets of T cells, and cytokines, exhibiting varying relative abundances and structural compositions, were observed in both healthy controls and patients throughout distinct phases of tuberculosis. Gaining insight into the function of the gut microbiome, T cell subsets, and cytokines may help modulate therapeutic strategies for TB.


Asunto(s)
Biomarcadores , Citocinas , Microbioma Gastrointestinal , Subgrupos de Linfocitos T , Tuberculosis , Humanos , Microbioma Gastrointestinal/inmunología , Citocinas/metabolismo , Masculino , Femenino , Adulto , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Persona de Mediana Edad , Tuberculosis/inmunología , Tuberculosis/microbiología , Tuberculosis/diagnóstico , Bacterias/inmunología , Bacterias/clasificación , Mycobacterium tuberculosis/inmunología , Heces/microbiología
6.
Curr Opin Infect Dis ; 37(3): 164-169, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38527455

RESUMEN

PURPOSE OF REVIEW: Many cholesterol-dependent cytolysin (CDC)-producing pathogens pose a significant threat to human health. Herein, we review the pore-dependent and -independent properties CDCs possess to assist pathogens in evading the host immune response. RECENT FINDINGS: Within the last 5 years, exciting new research suggests CDCs can act to inhibit important immune functions, disrupt critical cell signaling pathways, and have tissue-specific effects. Additionally, recent studies have identified a key region of CDCs that generates robust immunity, providing resources for the development of CDC-based vaccines. SUMMARY: This review provides new information on how CDCs alter host immune responses to aid bacteria in pathogenesis. These studies can assist in the design of more efficient vaccines and therapeutics against CDCs that will enhance the immune response to CDC-producing pathogens while mitigating the dampening effects CDCs have on the host immune response.


Asunto(s)
Colesterol , Citotoxinas , Humanos , Colesterol/metabolismo , Citotoxinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Bacterias/inmunología , Evasión Inmune/inmunología
7.
Nature ; 627(8004): 620-627, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38448595

RESUMEN

The fungus Candida albicans frequently colonizes the human gastrointestinal tract, from which it can disseminate to cause systemic disease. This polymorphic species can transition between growing as single-celled yeast and as multicellular hyphae to adapt to its environment. The current dogma of C. albicans commensalism is that the yeast form is optimal for gut colonization, whereas hyphal cells are detrimental to colonization but critical for virulence1-3. Here, we reveal that this paradigm does not apply to multi-kingdom communities in which a complex interplay between fungal morphology and bacteria dictates C. albicans fitness. Thus, whereas yeast-locked cells outcompete wild-type cells when gut bacteria are absent or depleted by antibiotics, hyphae-competent wild-type cells outcompete yeast-locked cells in hosts with replete bacterial populations. This increased fitness of wild-type cells involves the production of hyphal-specific factors including the toxin candidalysin4,5, which promotes the establishment of colonization. At later time points, adaptive immunity is engaged, and intestinal immunoglobulin A preferentially selects against hyphal cells1,6. Hyphal morphotypes are thus under both positive and negative selective pressures in the gut. Our study further shows that candidalysin has a direct inhibitory effect on bacterial species, including limiting their metabolic output. We therefore propose that C. albicans has evolved hyphal-specific factors, including candidalysin, to better compete with bacterial species in the intestinal niche.


Asunto(s)
Candida albicans , Proteínas Fúngicas , Microbioma Gastrointestinal , Hifa , Intestinos , Micotoxinas , Simbiosis , Animales , Femenino , Humanos , Masculino , Ratones , Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Candida albicans/crecimiento & desarrollo , Candida albicans/inmunología , Candida albicans/metabolismo , Candida albicans/patogenicidad , Proteínas Fúngicas/metabolismo , Microbioma Gastrointestinal/inmunología , Hifa/crecimiento & desarrollo , Hifa/inmunología , Hifa/metabolismo , Inmunoglobulina A/inmunología , Intestinos/inmunología , Intestinos/microbiología , Micotoxinas/metabolismo , Virulencia
8.
Nature ; 628(8006): 171-179, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509360

RESUMEN

The myriad microorganisms that live in close association with humans have diverse effects on physiology, yet the molecular bases for these impacts remain mostly unknown1-3. Classical pathogens often invade host tissues and modulate immune responses through interactions with human extracellular and secreted proteins (the 'exoproteome'). Commensal microorganisms may also facilitate niche colonization and shape host biology by engaging host exoproteins; however, direct exoproteome-microbiota interactions remain largely unexplored. Here we developed and validated a novel technology, BASEHIT, that enables proteome-scale assessment of human exoproteome-microbiome interactions. Using BASEHIT, we interrogated more than 1.7 million potential interactions between 519 human-associated bacterial strains from diverse phylogenies and tissues of origin and 3,324 human exoproteins. The resulting interactome revealed an extensive network of transkingdom connectivity consisting of thousands of previously undescribed host-microorganism interactions involving 383 strains and 651 host proteins. Specific binding patterns within this network implied underlying biological logic; for example, conspecific strains exhibited shared exoprotein-binding patterns, and individual tissue isolates uniquely bound tissue-specific exoproteins. Furthermore, we observed dozens of unique and often strain-specific interactions with potential roles in niche colonization, tissue remodelling and immunomodulation, and found that strains with differing host interaction profiles had divergent interactions with host cells in vitro and effects on the host immune system in vivo. Overall, these studies expose a previously unexplored landscape of molecular-level host-microbiota interactions that may underlie causal effects of indigenous microorganisms on human health and disease.


Asunto(s)
Bacterias , Interacciones Microbiota-Huesped , Microbiota , Filogenia , Proteoma , Simbiosis , Animales , Femenino , Humanos , Ratones , Bacterias/clasificación , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/patogenicidad , Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/fisiología , Tropismo al Anfitrión , Microbiota/inmunología , Microbiota/fisiología , Especificidad de Órganos , Unión Proteica , Proteoma/inmunología , Proteoma/metabolismo , Reproducibilidad de los Resultados
10.
Nature ; 628(8006): 204-211, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38418880

RESUMEN

The eye, an anatomical extension of the central nervous system (CNS), exhibits many molecular and cellular parallels to the brain. Emerging research demonstrates that changes in the brain are often reflected in the eye, particularly in the retina1. Still, the possibility of an immunological nexus between the posterior eye and the rest of the CNS tissues remains unexplored. Here, studying immune responses to herpes simplex virus in the brain, we observed that intravitreal immunization protects mice against intracranial viral challenge. This protection extended to bacteria and even tumours, allowing therapeutic immune responses against glioblastoma through intravitreal immunization. We further show that the anterior and posterior compartments of the eye have distinct lymphatic drainage systems, with the latter draining to the deep cervical lymph nodes through lymphatic vasculature in the optic nerve sheath. This posterior lymphatic drainage, like that of meningeal lymphatics, could be modulated by the lymphatic stimulator VEGFC. Conversely, we show that inhibition of lymphatic signalling on the optic nerve could overcome a major limitation in gene therapy by diminishing the immune response to adeno-associated virus and ensuring continued efficacy after multiple doses. These results reveal a shared lymphatic circuit able to mount a unified immune response between the posterior eye and the brain, highlighting an understudied immunological feature of the eye and opening up the potential for new therapeutic strategies in ocular and CNS diseases.


Asunto(s)
Encéfalo , Ojo , Sistema Linfático , Animales , Femenino , Humanos , Masculino , Ratones , Conejos , Bacterias/inmunología , Encéfalo/anatomía & histología , Encéfalo/inmunología , Dependovirus/inmunología , Ojo/anatomía & histología , Ojo/inmunología , Glioblastoma/inmunología , Herpesvirus Humano 2/inmunología , Inyecciones Intravítreas , Sistema Linfático/anatomía & histología , Sistema Linfático/inmunología , Vasos Linfáticos/anatomía & histología , Vasos Linfáticos/inmunología , Macaca mulatta , Meninges/inmunología , Nervio Óptico/inmunología , Porcinos , Pez Cebra , Factor C de Crecimiento Endotelial Vascular/inmunología , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/farmacología
11.
Science ; 383(6686): eabm9903, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38422126

RESUMEN

All living organisms deploy cell-autonomous defenses to combat infection. In plants and animals, large supramolecular complexes often activate immune proteins for protection. In this work, we resolved the native structure of a massive host-defense complex that polymerizes 30,000 guanylate-binding proteins (GBPs) over the surface of gram-negative bacteria inside human cells. Construction of this giant nanomachine took several minutes and remained stable for hours, required guanosine triphosphate hydrolysis, and recruited four GBPs plus caspase-4 and Gasdermin D as a cytokine and cell death immune signaling platform. Cryo-electron tomography suggests that GBP1 can adopt an extended conformation for bacterial membrane insertion to establish this platform, triggering lipopolysaccharide release that activated coassembled caspase-4. Our "open conformer" model provides a dynamic view into how the human GBP1 defense complex mobilizes innate immunity to infection.


Asunto(s)
Bacterias , Infecciones Bacterianas , Membrana Celular , Proteínas de Unión al GTP , Reconocimiento de Inmunidad Innata , Humanos , Citocinas/química , Tomografía con Microscopio Electrónico , Proteínas de Unión al GTP/química , Guanosina Trifosfato/química , Hidrólisis , Inmunidad Celular , Microscopía por Crioelectrón , Gasderminas/química , Proteínas de Unión a Fosfato/química , Conformación Proteica , Membrana Celular/química , Membrana Celular/inmunología , Caspasas Iniciadoras/química , Infecciones Bacterianas/inmunología , Bacterias/inmunología
12.
Nature ; 625(7996): 750-759, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38200311

RESUMEN

Iron is critical during host-microorganism interactions1-4. Restriction of available iron by the host during infection is an important defence strategy, described as nutritional immunity5. However, this poses a conundrum for externally facing, absorptive tissues such as the gut epithelium or the plant root epidermis that generate environments that favour iron bioavailability. For example, plant roots acquire iron mostly from the soil and, when iron deficient, increase iron availability through mechanisms that include rhizosphere acidification and secretion of iron chelators6-9. Yet, the elevated iron bioavailability would also be beneficial for the growth of bacteria that threaten plant health. Here we report that microorganism-associated molecular patterns such as flagellin lead to suppression of root iron acquisition through a localized degradation of the systemic iron-deficiency signalling peptide Iron Man 1 (IMA1) in Arabidopsis thaliana. This response is also elicited when bacteria enter root tissues, but not when they dwell on the outer root surface. IMA1 itself has a role in modulating immunity in root and shoot, affecting the levels of root colonization and the resistance to a bacterial foliar pathogen. Our findings reveal an adaptive molecular mechanism of nutritional immunity that affects iron bioavailability and uptake, as well as immune responses.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Bacterias , Péptidos y Proteínas de Señalización Intracelular , Hierro , Moléculas de Patrón Molecular Asociado a Patógenos , Raíces de Plantas , Arabidopsis/inmunología , Arabidopsis/metabolismo , Arabidopsis/microbiología , Proteínas de Arabidopsis/metabolismo , Bacterias/inmunología , Bacterias/metabolismo , Flagelina/inmunología , Regulación de la Expresión Génica de las Plantas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hierro/metabolismo , Inmunidad de la Planta , Raíces de Plantas/inmunología , Raíces de Plantas/metabolismo , Raíces de Plantas/microbiología , Brotes de la Planta/inmunología , Brotes de la Planta/metabolismo , Brotes de la Planta/microbiología , Rizosfera , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo
14.
Nature ; 625(7994): 352-359, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37992756

RESUMEN

It was recently shown that bacteria use, apart from CRISPR-Cas and restriction systems, a considerable diversity of phage resistance systems1-4, but it is largely unknown how phages cope with this multilayered bacterial immunity. Here we analysed groups of closely related Bacillus phages that showed differential sensitivity to bacterial defence systems, and discovered four distinct families of anti-defence proteins that inhibit the Gabija, Thoeris and Hachiman systems. We show that these proteins Gad1, Gad2, Tad2 and Had1 efficiently cancel the defensive activity when co-expressed with the respective defence system or introduced into phage genomes. Homologues of these anti-defence proteins are found in hundreds of phages that infect taxonomically diverse bacterial species. We show that the anti-Gabija protein Gad1 blocks the ability of the Gabija defence complex to cleave phage-derived DNA. Our data further reveal that the anti-Thoeris protein Tad2 is a 'sponge' that sequesters the immune signalling molecules produced by Thoeris TIR-domain proteins in response to phage infection. Our results demonstrate that phages encode an arsenal of anti-defence proteins that can disable a variety of bacterial defence mechanisms.


Asunto(s)
Fagos de Bacillus , Bacterias , Proteínas Virales , Fagos de Bacillus/clasificación , Fagos de Bacillus/genética , Fagos de Bacillus/inmunología , Fagos de Bacillus/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/inmunología , Bacterias/virología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
15.
Nature ; 625(7994): 360-365, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37992757

RESUMEN

Bacteria encode hundreds of diverse defence systems that protect them from viral infection and inhibit phage propagation1-5. Gabija is one of the most prevalent anti-phage defence systems, occurring in more than 15% of all sequenced bacterial and archaeal genomes1,6,7, but the molecular basis of how Gabija defends cells from viral infection remains poorly understood. Here we use X-ray crystallography and cryo-electron microscopy (cryo-EM) to define how Gabija proteins assemble into a supramolecular complex of around 500 kDa that degrades phage DNA. Gabija protein A (GajA) is a DNA endonuclease that tetramerizes to form the core of the anti-phage defence complex. Two sets of Gabija protein B (GajB) dimers dock at opposite sides of the complex and create a 4:4 GajA-GajB assembly (hereafter, GajAB) that is essential for phage resistance in vivo. We show that a phage-encoded protein, Gabija anti-defence 1 (Gad1), directly binds to the Gabija GajAB complex and inactivates defence. A cryo-EM structure of the virally inhibited state shows that Gad1 forms an octameric web that encases the GajAB complex and inhibits DNA recognition and cleavage. Our results reveal the structural basis of assembly of the Gabija anti-phage defence complex and define a unique mechanism of viral immune evasion.


Asunto(s)
Bacterias , Proteínas Bacterianas , Bacteriófagos , Evasión Inmune , Multimerización de Proteína , Bacterias/genética , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/virología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/genética , Bacteriófagos/inmunología , Bacteriófagos/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Desoxirribonucleasas/química , Desoxirribonucleasas/metabolismo , Desoxirribonucleasas/ultraestructura , ADN Viral/química , ADN Viral/metabolismo , ADN Viral/ultraestructura
16.
Nature ; 623(7989): 1001-1008, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37968393

RESUMEN

Cyclic oligonucleotide-based antiphage signalling systems (CBASS) protect prokaryotes from viral (phage) attack through the production of cyclic oligonucleotides, which activate effector proteins that trigger the death of the infected host1,2. How bacterial cyclases recognize phage infection is not known. Here we show that staphylococcal phages produce a structured RNA transcribed from the terminase subunit genes, termed CBASS-activating bacteriophage RNA (cabRNA), which binds to a positively charged surface of the CdnE03 cyclase and promotes the synthesis of the cyclic dinucleotide cGAMP to activate the CBASS immune response. Phages that escape the CBASS defence harbour mutations that lead to the generation of a longer form of the cabRNA that cannot activate CdnE03. As the mammalian cyclase OAS1 also binds viral double-stranded RNA during the interferon response, our results reveal a conserved mechanism for the activation of innate antiviral defence pathways.


Asunto(s)
Bacterias , Nucleotidiltransferasas , ARN Viral , Fagos de Staphylococcus , Animales , 2',5'-Oligoadenilato Sintetasa/metabolismo , Bacterias/enzimología , Bacterias/inmunología , Evolución Molecular , Inmunidad Innata , Nucleotidiltransferasas/metabolismo , Oligonucleótidos/inmunología , Oligonucleótidos/metabolismo , ARN Viral/inmunología , ARN Viral/metabolismo , Transducción de Señal/inmunología , Fagos de Staphylococcus/genética , Fagos de Staphylococcus/inmunología
17.
Nature ; 623(7987): 601-607, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37853129

RESUMEN

Many bacteria use CRISPR-Cas systems to combat mobile genetic elements, such as bacteriophages and plasmids1. In turn, these invasive elements have evolved anti-CRISPR proteins to block host immunity2,3. Here we unveil a distinct type of CRISPR-Cas Inhibition strategy that is based on small non-coding RNA anti-CRISPRs (Racrs). Racrs mimic the repeats found in CRISPR arrays and are encoded in viral genomes as solitary repeat units4. We show that a prophage-encoded Racr strongly inhibits the type I-F CRISPR-Cas system by interacting specifically with Cas6f and Cas7f, resulting in the formation of an aberrant Cas subcomplex. We identified Racr candidates for almost all CRISPR-Cas types encoded by a diverse range of viruses and plasmids, often in the genetic context of other anti-CRISPR genes5. Functional testing of nine candidates spanning the two CRISPR-Cas classes confirmed their strong immune inhibitory function. Our results demonstrate that molecular mimicry of CRISPR repeats is a widespread anti-CRISPR strategy, which opens the door to potential biotechnological applications6.


Asunto(s)
Bacterias , Bacteriófagos , Sistemas CRISPR-Cas , Imitación Molecular , ARN Viral , Bacterias/genética , Bacterias/inmunología , Bacterias/virología , Bacteriófagos/genética , Bacteriófagos/inmunología , Biotecnología/métodos , Biotecnología/tendencias , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/inmunología , Plásmidos/genética , Profagos/genética , Profagos/inmunología , ARN Viral/genética
18.
Nature ; 621(7977): 162-170, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37587342

RESUMEN

Certain bacterial strains from the microbiome induce a potent, antigen-specific T cell response1-5. However, the specificity of microbiome-induced T cells has not been explored at the strain level across the gut community. Here, we colonize germ-free mice with complex defined communities (roughly 100 bacterial strains) and profile T cell responses to each strain. The pattern of responses suggests that many T cells in the gut repertoire recognize several bacterial strains from the community. We constructed T cell hybridomas from 92 T cell receptor (TCR) clonotypes; by screening every strain in the community against each hybridoma, we find that nearly all the bacteria-specific TCRs show a one-to-many TCR-to-strain relationship, including 13 abundant TCR clonotypes that each recognize 18 Firmicutes. By screening three pooled bacterial genomic libraries, we discover that these 13 clonotypes share a single target: a conserved substrate-binding protein from an ATP-binding cassette transport system. Peripheral regulatory T cells and T helper 17 cells specific for an epitope from this protein are abundant in community-colonized and specific pathogen-free mice. Our work reveals that T cell recognition of commensals is focused on widely conserved, highly expressed cell-surface antigens, opening the door to new therapeutic strategies in which colonist-specific immune responses are rationally altered or redirected.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , Linfocitos T , Animales , Ratones , Antígenos de Superficie/inmunología , Bacterias/clasificación , Bacterias/inmunología , Firmicutes/inmunología , Microbioma Gastrointestinal/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Linfocitos T/inmunología , Simbiosis/inmunología , Vida Libre de Gérmenes , Receptores de Antígenos de Linfocitos T/inmunología , Hibridomas/citología , Hibridomas/inmunología , Separación Celular
19.
Science ; 380(6649): eabo2296, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37289890

RESUMEN

Antibiotics (ABX) compromise the efficacy of programmed cell death protein 1 (PD-1) blockade in cancer patients, but the mechanisms underlying their immunosuppressive effects remain unknown. By inducing the down-regulation of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the ileum, post-ABX gut recolonization by Enterocloster species drove the emigration of enterotropic α4ß7+CD4+ regulatory T 17 cells into the tumor. These deleterious ABX effects were mimicked by oral gavage of Enterocloster species, by genetic deficiency, or by antibody-mediated neutralization of MAdCAM-1 and its receptor, α4ß7 integrin. By contrast, fecal microbiota transplantation or interleukin-17A neutralization prevented ABX-induced immunosuppression. In independent lung, kidney, and bladder cancer patient cohorts, low serum levels of soluble MAdCAM-1 had a negative prognostic impact. Thus, the MAdCAM-1-α4ß7 axis constitutes an actionable gut immune checkpoint in cancer immunosurveillance.


Asunto(s)
Antibacterianos , Moléculas de Adhesión Celular , Resistencia a Antineoplásicos , Microbioma Gastrointestinal , Inhibidores de Puntos de Control Inmunológico , Tolerancia Inmunológica , Vigilancia Inmunológica , Integrinas , Mucoproteínas , Neoplasias , Animales , Humanos , Ratones , Antibacterianos/efectos adversos , Bacterias/inmunología , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Tolerancia Inmunológica/efectos de los fármacos , Integrinas/metabolismo , Interleucina-17/metabolismo , Mucoproteínas/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Células Th17/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología
20.
Nature ; 620(7974): 660-668, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37380027

RESUMEN

RNA-guided systems, which use complementarity between a guide RNA and target nucleic acid sequences for recognition of genetic elements, have a central role in biological processes in both prokaryotes and eukaryotes. For example, the prokaryotic CRISPR-Cas systems provide adaptive immunity for bacteria and archaea against foreign genetic elements. Cas effectors such as Cas9 and Cas12 perform guide-RNA-dependent DNA cleavage1. Although a few eukaryotic RNA-guided systems have been studied, including RNA interference2 and ribosomal RNA modification3, it remains unclear whether eukaryotes have RNA-guided endonucleases. Recently, a new class of prokaryotic RNA-guided systems (termed OMEGA) was reported4,5. The OMEGA effector TnpB is the putative ancestor of Cas12 and has RNA-guided endonuclease activity4,6. TnpB may also be the ancestor of the eukaryotic transposon-encoded Fanzor (Fz) proteins4,7, raising the possibility that eukaryotes are also equipped with CRISPR-Cas or OMEGA-like programmable RNA-guided endonucleases. Here we report the biochemical characterization of Fz, showing that it is an RNA-guided DNA endonuclease. We also show that Fz can be reprogrammed for human genome engineering applications. Finally, we resolve the structure of Spizellomyces punctatus Fz at 2.7 Å using cryogenic electron microscopy, showing the conservation of core regions among Fz, TnpB and Cas12, despite diverse cognate RNA structures. Our results show that Fz is a eukaryotic OMEGA system, demonstrating that RNA-guided endonucleases are present in all three domains of life.


Asunto(s)
Quitridiomicetos , Endonucleasas , Eucariontes , Proteínas Fúngicas , Edición Génica , ARN , Humanos , Archaea/genética , Archaea/inmunología , Bacterias/genética , Bacterias/inmunología , Proteína 9 Asociada a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/química , Proteínas Asociadas a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/ultraestructura , Sistemas CRISPR-Cas , Elementos Transponibles de ADN/genética , Endonucleasas/química , Endonucleasas/metabolismo , Endonucleasas/ultraestructura , Eucariontes/enzimología , Edición Génica/métodos , ARN/genética , ARN/metabolismo , ARN Guía de Sistemas CRISPR-Cas/genética , ARN Guía de Sistemas CRISPR-Cas/metabolismo , Microscopía por Crioelectrón , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/ultraestructura , Evolución Molecular , Secuencia Conservada , Quitridiomicetos/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA