Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.079
Filtrar
Más filtros

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 ; 183(5): 1312-1324.e10, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33212011

RESUMEN

Interferon (IFN)-Is are crucial mediators of antiviral immunity and homeostatic immune system regulation. However, the source of IFN-I signaling under homeostatic conditions is unclear. We discovered that commensal microbes regulate the IFN-I response through induction of IFN-ß by colonic DCs. Moreover, the mechanism by which a specific commensal microbe induces IFN-ß was identified. Outer membrane (OM)-associated glycolipids of gut commensal microbes belonging to the Bacteroidetes phylum induce expression of IFN-ß. Using Bacteroides fragilis and its OM-associated polysaccharide A, we determined that IFN-ß expression was induced via TLR4-TRIF signaling. Antiviral activity of this purified microbial molecule against infection with either vesicular stomatitis virus (VSV) or influenza was demonstrated to be dependent on the induction of IFN-ß. In a murine VSV infection model, commensal-induced IFN-ß regulated natural resistance to virus infection. Due to the physiological importance of IFN-Is, discovery of an IFN-ß-inducing microbial molecule represents a potential approach for the treatment of some human diseases.


Asunto(s)
Inmunidad Innata , Microbiota , Virosis/microbiología , Animales , Bacteroides fragilis/fisiología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colon/patología , Colon/virología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucolípidos/metabolismo , Inmunidad Innata/efectos de los fármacos , Interferón beta/sangre , Interferón beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Receptor Toll-Like 4/metabolismo , Vesiculovirus/fisiología , Virosis/genética
4.
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
5.
Immunity ; 57(4): 876-889.e11, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38479384

RESUMEN

Concentrations of the secondary bile acid, deoxycholic acid (DCA), are aberrantly elevated in colorectal cancer (CRC) patients, but the consequences remain poorly understood. Here, we screened a library of gut microbiota-derived metabolites and identified DCA as a negative regulator for CD8+ T cell effector function. Mechanistically, DCA suppressed CD8+ T cell responses by targeting plasma membrane Ca2+ ATPase (PMCA) to inhibit Ca2+-nuclear factor of activated T cells (NFAT)2 signaling. In CRC patients, CD8+ T cell effector function negatively correlated with both DCA concentration and expression of a bacterial DCA biosynthetic gene. Bacteria harboring DCA biosynthetic genes suppressed CD8+ T cells effector function and promoted tumor growth in mice. This effect was abolished by disrupting bile acid metabolism via bile acid chelation, genetic ablation of bacterial DCA biosynthetic pathway, or specific bacteriophage. Our study demonstrated causation between microbial DCA metabolism and anti-tumor CD8+ T cell response in CRC, suggesting potential directions for anti-tumor therapy.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Ácidos y Sales Biliares , Ácido Desoxicólico/farmacología , Linfocitos T CD8-positivos
6.
Cell ; 175(6): 1546-1560.e17, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30500537

RESUMEN

Mammalian folate metabolism is comprised of cytosolic and mitochondrial pathways with nearly identical core reactions, yet the functional advantages of such an organization are not well understood. Using genome-editing and biochemical approaches, we find that ablating folate metabolism in the mitochondria of mammalian cell lines results in folate degradation in the cytosol. Mechanistically, we show that QDPR, an enzyme in tetrahydrobiopterin metabolism, moonlights to repair oxidative damage to tetrahydrofolate (THF). This repair capacity is overwhelmed when cytosolic THF hyperaccumulates in the absence of mitochondrially produced formate, leading to THF degradation. Unexpectedly, we also find that the classic antifolate methotrexate, by inhibiting its well-known target DHFR, causes even more extensive folate degradation in nearly all tested cancer cell lines. These findings shed light on design features of folate metabolism, provide a biochemical basis for clinically observed folate deficiency in QDPR-deficient patients, and reveal a hitherto unknown and unexplored cellular effect of methotrexate.


Asunto(s)
Carbono/metabolismo , Citosol/metabolismo , Formiatos/metabolismo , Mitocondrias/metabolismo , Neoplasias/metabolismo , Tetrahidrofolatos/metabolismo , Citosol/patología , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , Metotrexato/farmacocinética , Metotrexato/farmacología , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Tetrahidrofolato Deshidrogenasa/metabolismo
7.
Annu Rev Immunol ; 28: 107-30, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19968562

RESUMEN

The capsular polysaccharides (CPSs) of most pathogenic bacteria are T cell-independent antigens whose conjugation to carrier proteins evokes a carbohydrate-specific response eliciting T cell help. However, certain bacterial CPSs, known as zwitterionic polysaccharides (ZPSs), activate the adaptive immune system through processing by antigen-presenting cells and presentation by the major histocompatibility complex class II pathway to CD4(+) T cells. This discovery was the first mechanistic insight into how carbohydrates-a class of biological molecules previously thought to be T cell independent-can in fact activate T cells. Through their ability to activate CD4(+) T cells, ZPSs direct the cellular and physical maturation of the developing immune system. In this review, we explore the still-enigmatic relations between CPSs and the adaptive immune machinery at the cellular and molecular levels, and we discuss how new insights into the biological impact of ZPSs expand our concepts of the role of carbohydrates in microbial interactions with the adaptive immune system.


Asunto(s)
Inmunidad Adaptativa , Antígenos Bacterianos/inmunología , Bacterias/inmunología , Polisacáridos Bacterianos/inmunología , Animales , Antígenos Bacterianos/química , Bacterias/química , Vacunas Bacterianas/inmunología , Humanos , Polisacáridos Bacterianos/química , Linfocitos T/inmunología
8.
Cell ; 168(6): 1135-1148.e12, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28262351

RESUMEN

Investigation of host-environment interactions in the gut would benefit from a culture system that maintained tissue architecture yet allowed tight experimental control. We devised a microfabricated organ culture system that viably preserves the normal multicellular composition of the mouse intestine, with luminal flow to control perturbations (e.g., microbes, drugs). It enables studying short-term responses of diverse gut components (immune, neuronal, etc.). We focused on the early response to bacteria that induce either Th17 or RORg+ T-regulatory (Treg) cells in vivo. Transcriptional responses partially reproduced in vivo signatures, but these microbes elicited diametrically opposite changes in expression of a neuronal-specific gene set, notably nociceptive neuropeptides. We demonstrated activation of sensory neurons by microbes, correlating with RORg+ Treg induction. Colonic RORg+ Treg frequencies increased in mice lacking TAC1 neuropeptide precursor and decreased in capsaicin-diet fed mice. Thus, differential engagement of the enteric nervous system may partake in bifurcating pro- or anti-inflammatory responses to microbes.


Asunto(s)
Clostridium/crecimiento & desarrollo , Intestinos/crecimiento & desarrollo , Intestinos/microbiología , Técnicas de Cultivo de Órganos , Animales , Clostridium/clasificación , Clostridium/fisiología , Intestinos/citología , Ratones , Simbiosis
9.
Cell ; 168(5): 928-943.e11, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28215708

RESUMEN

Within the human gut reside diverse microbes coexisting with the host in a mutually advantageous relationship. Evidence has revealed the pivotal role of the gut microbiota in shaping the immune system. To date, only a few of these microbes have been shown to modulate specific immune parameters. Herein, we broadly identify the immunomodulatory effects of phylogenetically diverse human gut microbes. We monocolonized mice with each of 53 individual bacterial species and systematically analyzed host immunologic adaptation to colonization. Most microbes exerted several specialized, complementary, and redundant transcriptional and immunomodulatory effects. Surprisingly, these were independent of microbial phylogeny. Microbial diversity in the gut ensures robustness of the microbiota's ability to generate a consistent immunomodulatory impact, serving as a highly important epigenetic system. This study provides a foundation for investigation of gut microbiota-host mutualism, highlighting key players that could identify important therapeutics.


Asunto(s)
Bacterias/clasificación , Microbioma Gastrointestinal , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Inmunidad Adaptativa , Animales , Fenómenos Fisiológicos Bacterianos , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/fisiología , Vida Libre de Gérmenes , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Simbiosis
10.
Cell ; 171(7): 1573-1588.e28, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29224777

RESUMEN

There is considerable evidence that chromosome structure plays important roles in gene control, but we have limited understanding of the proteins that contribute to structural interactions between gene promoters and their enhancer elements. Large DNA loops that encompass genes and their regulatory elements depend on CTCF-CTCF interactions, but most enhancer-promoter interactions do not employ this structural protein. Here, we show that the ubiquitously expressed transcription factor Yin Yang 1 (YY1) contributes to enhancer-promoter structural interactions in a manner analogous to DNA interactions mediated by CTCF. YY1 binds to active enhancers and promoter-proximal elements and forms dimers that facilitate the interaction of these DNA elements. Deletion of YY1 binding sites or depletion of YY1 protein disrupts enhancer-promoter looping and gene expression. We propose that YY1-mediated enhancer-promoter interactions are a general feature of mammalian gene control.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Factor de Transcripción YY1/metabolismo , Animales , Factor de Unión a CCCTC/metabolismo , Células Madre Embrionarias/metabolismo , Humanos , Ratones
11.
Cell ; 167(5): 1161-1162, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863236

RESUMEN

In this issue of Cell, Desai et al. compare how dietary fiber affects the gut microbiota and susceptibility to disease. They find that a fiber-free diet promotes mucus-degrading bacteria and susceptibility to Citrobacter rodentium infection.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae/microbiología , Bacterias , Dieta , Susceptibilidad a Enfermedades , Humanos
12.
Cell ; 156(1-2): 123-33, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439373

RESUMEN

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


Asunto(s)
Bacteroides fragilis/metabolismo , Colitis/inmunología , Glicoesfingolípidos/metabolismo , Células T Asesinas Naturales/inmunología , Animales , Animales Recién Nacidos , Proliferación Celular , Colitis/inducido químicamente , Colitis/prevención & control , Colon/crecimiento & desarrollo , Colon/microbiología , Ratones , Ratones Endogámicos C57BL , Células T Asesinas Naturales/citología , Oxazolona
13.
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
14.
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
15.
Cell ; 149(7): 1578-93, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22726443

RESUMEN

Gut microbial induction of host immune maturation exemplifies host-microbe mutualism. We colonized germ-free (GF) mice with mouse microbiota (MMb) or human microbiota (HMb) to determine whether small intestinal immune maturation depends on a coevolved host-specific microbiota. Gut bacterial numbers and phylum abundance were similar in MMb and HMb mice, but bacterial species differed, especially the Firmicutes. HMb mouse intestines had low levels of CD4(+) and CD8(+) T cells, few proliferating T cells, few dendritic cells, and low antimicrobial peptide expression--all characteristics of GF mice. Rat microbiota also failed to fully expand intestinal T cell numbers in mice. Colonizing GF or HMb mice with mouse-segmented filamentous bacteria (SFB) partially restored T cell numbers, suggesting that SFB and other MMb organisms are required for full immune maturation in mice. Importantly, MMb conferred better protection against Salmonella infection than HMb. A host-specific microbiota appears to be critical for a healthy immune system.


Asunto(s)
Inmunidad Innata , Intestinos/inmunología , Intestinos/microbiología , Metagenoma , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/metabolismo , Proliferación Celular , Femenino , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Infecciones por Salmonella/inmunología , Especificidad de la Especie , Organismos Libres de Patógenos Específicos , Simbiosis , Linfocitos T/citología , Linfocitos T/inmunología
16.
Nature ; 600(7888): 302-307, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34759313

RESUMEN

Small molecules derived from symbiotic microbiota critically contribute to intestinal immune maturation and regulation1. However, little is known about the molecular mechanisms that control immune development in the host-microbiota environment. Here, using a targeted lipidomic analysis and synthetic approach, we carried out a multifaceted investigation of immunomodulatory α-galactosylceramides from the human symbiont Bacteroides fragilis (BfaGCs). The characteristic terminal branching of BfaGCs is the result of incorporation of branched-chain amino acids taken up in the host gut by B. fragilis. A B. fragilis knockout strain that cannot metabolize branched-chain amino acids showed reduced branching in BfaGCs, and mice monocolonized with this mutant strain had impaired colonic natural killer T (NKT) cell regulation, implying structure-specific immunomodulatory activity. The sphinganine chain branching of BfaGCs is a critical determinant of NKT cell activation, which induces specific immunomodulatory gene expression signatures and effector functions. Co-crystal structure and affinity analyses of CD1d-BfaGC-NKT cell receptor complexes confirmed the interaction of BfaGCs as CD1d-restricted ligands. We present a structural and molecular-level paradigm of immunomodulatory control by interactions of endobiotic metabolites with diet, microbiota and the immune system.


Asunto(s)
Aminoácidos de Cadena Ramificada/inmunología , Aminoácidos de Cadena Ramificada/metabolismo , Bacteroides fragilis/metabolismo , Galactosilceramidas/inmunología , Galactosilceramidas/metabolismo , Microbioma Gastrointestinal/inmunología , Simbiosis/inmunología , Aminoácidos de Cadena Ramificada/química , Animales , Antígenos CD1d/inmunología , Bacteroides fragilis/genética , Humanos , Ratones , Modelos Animales , Modelos Moleculares , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología
17.
Blood ; 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39316768

RESUMEN

A metronomic, low-dose schedule of decitabine and Venetoclax was safe and effective in myeloid malignancies with few dose reductions or interruptions in an older diverse population. Median OS for AML and TP53 mutated patients was 16.1 and 11.3 months respectively.

18.
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
19.
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
20.
J Cell Sci ; 136(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36606487

RESUMEN

53BP1 (also known as TP53BP1) is a key mediator of the non-homologous end joining (NHEJ) DNA repair pathway, which is the primary repair pathway in interphase cells. However, the mitotic functions of 53BP1 are less well understood. Here, we describe 53BP1 mitotic stress bodies (MSBs) formed in cancer cell lines in response to delayed mitosis. These bodies displayed liquid-liquid phase separation characteristics, were close to centromeres, and included lamin A/C and the DNA repair protein RIF1. After release from mitotic arrest, 53BP1 MSBs decreased in number and moved away from the chromatin. Using GFP fusion constructs, we found that the 53BP1 oligomerization domain region was required for MSB formation, and that inclusion of the 53BP1 N terminus increased MSB size. Exogenous expression of 53BP1 did not increase MSB size or number but did increase levels of MSB-free 53BP1. This was associated with slower mitotic progression, elevated levels of DNA damage and increased apoptosis, which is consistent with MSBs suppressing a mitotic surveillance by 53BP1 through sequestration. The 53BP1 MSBs, which were also found spontaneously in a subset of normally dividing cancer cells but not in non-transformed cells (ARPE-19), might facilitate the survival of cancer cells following aberrant mitoses. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Reparación del ADN , Neoplasias , Proteína 1 de Unión al Supresor Tumoral P53 , Humanos , Cromatina , Daño del ADN , Reparación del ADN por Unión de Extremidades , Mitosis , Neoplasias/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Línea Celular Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA