Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Nat Immunol ; 25(7): 1183-1192, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38872000

RESUMEN

Natural killer (NK) cells function by eliminating virus-infected or tumor cells. Here we identified an NK-lineage-biased progenitor population, referred to as early NK progenitors (ENKPs), which developed into NK cells independently of common precursors for innate lymphoid cells (ILCPs). ENKP-derived NK cells (ENKP_NK cells) and ILCP-derived NK cells (ILCP_NK cells) were transcriptionally different. We devised combinations of surface markers that identified highly enriched ENKP_NK and ILCP_NK cell populations in wild-type mice. Furthermore, Ly49H+ NK cells that responded to mouse cytomegalovirus infection primarily developed from ENKPs, whereas ILCP_NK cells were better IFNγ producers after infection with Salmonella and herpes simplex virus. Human CD56dim and CD56bright NK cells were transcriptionally similar to ENKP_NK cells and ILCP_NK cells, respectively. Our findings establish the existence of two pathways of NK cell development that generate functionally distinct NK cell subsets in mice and further suggest these pathways may be conserved in humans.


Asunto(s)
Diferenciación Celular , Células Asesinas Naturales , Células Asesinas Naturales/inmunología , Animales , Ratones , Humanos , Diferenciación Celular/inmunología , Ratones Endogámicos C57BL , Inmunidad Innata , Antígeno CD56/metabolismo , Muromegalovirus/inmunología , Linaje de la Célula/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Ratones Noqueados , Células Cultivadas
2.
Nat Commun ; 15(1): 4696, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824133

RESUMEN

Age-related microangiopathy, also known as small vessel disease (SVD), causes damage to the brain, retina, liver, and kidney. Based on the DNA damage theory of aging, we reasoned that genomic instability may underlie an SVD caused by dominant C-terminal variants in TREX1, the most abundant 3'-5' DNA exonuclease in mammals. C-terminal TREX1 variants cause an adult-onset SVD known as retinal vasculopathy with cerebral leukoencephalopathy (RVCL or RVCL-S). In RVCL, an aberrant, C-terminally truncated TREX1 mislocalizes to the nucleus due to deletion of its ER-anchoring domain. Since RVCL pathology mimics that of radiation injury, we reasoned that nuclear TREX1 would cause DNA damage. Here, we show that RVCL-associated TREX1 variants trigger DNA damage in humans, mice, and Drosophila, and that cells expressing RVCL mutant TREX1 are more vulnerable to DNA damage induced by chemotherapy and cytokines that up-regulate TREX1, leading to depletion of TREX1-high cells in RVCL mice. RVCL-associated TREX1 mutants inhibit homology-directed repair (HDR), causing DNA deletions and vulnerablility to PARP inhibitors. In women with RVCL, we observe early-onset breast cancer, similar to patients with BRCA1/2 variants. Our results provide a mechanistic basis linking aberrant TREX1 activity to the DNA damage theory of aging, premature senescence, and microvascular disease.


Asunto(s)
Daño del ADN , Exodesoxirribonucleasas , Fosfoproteínas , Animales , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratones , Reparación del ADN por Recombinación , Fenotipo , Mutación , Drosophila/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Femenino , Drosophila melanogaster/genética , Masculino , Enfermedades de la Retina , Enfermedades Vasculares , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias
3.
Immunity ; 57(4): 859-875.e11, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38513665

RESUMEN

At mucosal surfaces, epithelial cells provide a structural barrier and an immune defense system. However, dysregulated epithelial responses can contribute to disease states. Here, we demonstrated that epithelial cell-intrinsic production of interleukin-23 (IL-23) triggers an inflammatory loop in the prevalent oral disease periodontitis. Epithelial IL-23 expression localized to areas proximal to the disease-associated microbiome and was evident in experimental models and patients with common and genetic forms of disease. Mechanistically, flagellated microbial species of the periodontitis microbiome triggered epithelial IL-23 induction in a TLR5 receptor-dependent manner. Therefore, unlike other Th17-driven diseases, non-hematopoietic-cell-derived IL-23 served as an initiator of pathogenic inflammation in periodontitis. Beyond periodontitis, analysis of publicly available datasets revealed the expression of epithelial IL-23 in settings of infection, malignancy, and autoimmunity, suggesting a broader role for epithelial-intrinsic IL-23 in human disease. Collectively, this work highlights an important role for the barrier epithelium in the induction of IL-23-mediated inflammation.


Asunto(s)
Interleucina-23 , Periodontitis , Humanos , Células Epiteliales , Inflamación , Receptor Toll-Like 5/metabolismo
4.
Science ; 382(6674): 1073-1079, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38033053

RESUMEN

Skin-resident CD8+ T cells include distinct interferon-γ-producing [tissue-resident memory T type 1 (TRM1)] and interleukin-17 (IL-17)-producing (TRM17) subsets that differentially contribute to immune responses. However, whether these populations use common mechanisms to establish tissue residence is unknown. In this work, we show that TRM1 and TRM17 cells navigate divergent trajectories to acquire tissue residency in the skin. TRM1 cells depend on a T-bet-Hobit-IL-15 axis, whereas TRM17 cells develop independently of these factors. Instead, c-Maf commands a tissue-resident program in TRM17 cells parallel to that induced by Hobit in TRM1 cells, with an ICOS-c-Maf-IL-7 axis pivotal to TRM17 cell commitment. Accordingly, by targeting this pathway, skin TRM17 cells can be ablated without compromising their TRM1 counterparts. Thus, skin-resident T cells rely on distinct molecular circuitries, which can be exploited to strategically modulate local immunity.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Células T de Memoria , Piel , Linfocitos T CD8-positivos/inmunología , Células T de Memoria/inmunología , Piel/inmunología , Humanos , Células Th17/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Interleucina-7/metabolismo
5.
Science ; 380(6641): 203-210, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37053311

RESUMEN

Certain bacterial colonists induce a highly specific T cell response. A hallmark of this encounter is that adaptive immunity develops preemptively, in the absence of an infection. However, the functional properties of colonist-induced T cells are not well defined, limiting our ability to understand anticommensal immunity and harness it therapeutically. We addressed both challenges by engineering the skin bacterium Staphylococcus epidermidis to express tumor antigens anchored to secreted or cell-surface proteins. Upon colonization, engineered S. epidermidis elicits tumor-specific T cells that circulate, infiltrate local and metastatic lesions, and exert cytotoxic activity. Thus, the immune response to a skin colonist can promote cellular immunity at a distal site and can be redirected against a target of therapeutic interest by expressing a target-derived antigen in a commensal.


Asunto(s)
Antígenos de Neoplasias , Melanoma , Neoplasias Cutáneas , Piel , Staphylococcus epidermidis , Humanos , Inmunidad Celular , Melanoma/inmunología , Melanoma/terapia , Piel/microbiología , Ingeniería Genética , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/terapia , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología
6.
Ann N Y Acad Sci ; 1523(1): 38-50, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36960914

RESUMEN

Immunometabolism considers the relationship between metabolism and immunity. Typically, researchers focus on either the metabolic pathways within immune cells that affect their function or the impact of immune cells on systemic metabolism. A more holistic approach that considers both these viewpoints is needed. On September 5-8, 2022, experts in the field of immunometabolism met for the Keystone symposium "Immunometabolism at the Crossroads of Obesity and Cancer" to present recent research across the field of immunometabolism, with the setting of obesity and cancer as an ideal example of the complex interplay between metabolism, immunity, and cancer. Speakers highlighted new insights on the metabolic links between tumor cells and immune cells, with a focus on leveraging unique metabolic vulnerabilities of different cell types in the tumor microenvironment as therapeutic targets and demonstrated the effects of diet, the microbiome, and obesity on immune system function and cancer pathogenesis and therapy. Finally, speakers presented new technologies to interrogate the immune system and uncover novel metabolic pathways important for immunity.


Asunto(s)
Neoplasias , Humanos , Neoplasias/metabolismo , Sistema Inmunológico , Redes y Vías Metabólicas , Obesidad/terapia , Obesidad/metabolismo , Microambiente Tumoral
7.
Proc Natl Acad Sci U S A ; 119(26): e2200348119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35727974

RESUMEN

Immune checkpoint inhibitors (ICIs) are essential components of the cancer therapeutic armamentarium. While ICIs have demonstrated remarkable clinical responses, they can be accompanied by immune-related adverse events (irAEs). These inflammatory side effects are of unclear etiology and impact virtually all organ systems, with the most common being sites colonized by the microbiota such as the skin and gastrointestinal tract. Here, we establish a mouse model of commensal bacteria-driven skin irAEs and demonstrate that immune checkpoint inhibition unleashes commensal-specific inflammatory T cell responses. These aberrant responses were dependent on production of IL-17 by commensal-specific T cells and induced pathology that recapitulated the cutaneous inflammation seen in patients treated with ICIs. Importantly, aberrant T cell responses unleashed by ICIs were sufficient to perpetuate inflammatory memory responses to the microbiota months following the cessation of treatment. Altogether, we have established a mouse model of skin irAEs and reveal that ICIs unleash aberrant immune responses against skin commensals, with long-lasting inflammatory consequences.


Asunto(s)
Dermatitis , Inhibidores de Puntos de Control Inmunológico , Microbiota , Animales , Dermatitis/inmunología , Dermatitis/microbiología , Modelos Animales de Enfermedad , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunidad/efectos de los fármacos , Interleucina-17/metabolismo , Ratones , Microbiota/efectos de los fármacos , Microbiota/inmunología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/inmunología , Simbiosis/efectos de los fármacos , Linfocitos T/inmunología
8.
Cell Host Microbe ; 30(7): 1020-1033.e6, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35568028

RESUMEN

Antibiotics are a modifiable iatrogenic risk factor for the most common human nosocomial fungal infection, invasive candidiasis, yet the underlying mechanisms remain elusive. We found that antibiotics enhanced the susceptibility to murine invasive candidiasis due to impaired lymphocyte-dependent IL-17A- and GM-CSF-mediated antifungal immunity within the gut. This led to non-inflammatory bacterial escape and systemic bacterial co-infection, which could be ameliorated by IL-17A or GM-CSF immunotherapy. Vancomycin alone similarly enhanced the susceptibility to invasive fungal infection and systemic bacterial co-infection. Mechanistically, vancomycin reduced the frequency of gut Th17 cells associated with impaired proliferation and RORγt expression. Vancomycin's effects on Th17 cells were indirect, manifesting only in vivo in the presence of dysbiosis. In humans, antibiotics were associated with an increased risk of invasive candidiasis and death after invasive candidiasis. Our work highlights the importance of antibiotic stewardship in protecting vulnerable patients from life-threatening infections and provides mechanistic insights into a controllable iatrogenic risk factor for invasive candidiasis.


Asunto(s)
Antibacterianos , Candidiasis Invasiva , Coinfección , Animales , Antibacterianos/administración & dosificación , Antibacterianos/efectos adversos , Bacterias/efectos de los fármacos , Bacterias/inmunología , Candida albicans/inmunología , Candidiasis Invasiva/inmunología , Candidiasis Invasiva/microbiología , Coinfección/inmunología , Coinfección/microbiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Enfermedad Iatrogénica , Inmunoterapia , Interleucina-17/inmunología , Interleucina-17/uso terapéutico , Ratones , Células Th17/metabolismo , Vancomicina/farmacología
9.
Mucosal Immunol ; 15(4): 629-641, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35501356

RESUMEN

The nervous system and the immune system both rely on an extensive set of modalities to perceive and act on perturbations in the internal and external environments. During feeding, the intestine is exposed to nutrients that may contain noxious substances and pathogens. Here we show that Vasoactive Intestinal Peptide (VIP), produced by the nervous system in response to feeding, potentiates the production of effector cytokines by intestinal type 2 and type 3 innate lymphoid cells (ILC2s and ILC3s). Exposure to VIP alone leads to modest activation of ILCs, but strongly potentiates ILCs to concomitant or subsequent activation by the inducer cytokines IL-33 or IL-23, via mobilization of cAMP and energy by glycolysis. Consequently, VIP increases resistance to intestinal infection by the helminth Trichuris muris and the enterobacteria Citrobacter rodentium. These findings uncover a functional neuro-immune crosstalk unfolding during feeding that increases the reactivity of innate immunity necessary to face potential threats associated with food intake.


Asunto(s)
Neuropéptidos , Péptido Intestinal Vasoactivo , Citocinas/metabolismo , Inmunidad Innata , Intestinos , Linfocitos , Neuropéptidos/metabolismo
10.
Nat Immunol ; 23(4): 594-604, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35354951

RESUMEN

While T cell receptor (TCR) αß+CD8α+CD8ß- intraepithelial lymphocytes (CD8αα+ IELs) differentiate from thymic IEL precursors (IELps) and contribute to gut homeostasis, the transcriptional control of their development remains poorly understood. In the present study we showed that mouse thymocytes deficient for the transcription factor leukemia/lymphoma-related factor (LRF) failed to generate TCRαß+CD8αα+ IELs and their CD8ß-expressing counterparts, despite giving rise to thymus and spleen CD8αß+ T cells. LRF-deficient IELps failed to migrate to the intestine and to protect against T cell-induced colitis, and had impaired expression of the gut-homing integrin α4ß7. Single-cell RNA-sequencing found that LRF was necessary for the expression of genes characteristic of the most mature IELps, including Itgb7, encoding the ß7 subunit of α4ß7. Chromatin immunoprecipitation and gene-regulatory network analyses both defined Itgb7 as an LRF target. Our study identifies LRF as an essential transcriptional regulator of IELp maturation in the thymus and subsequent migration to the intestinal epithelium.


Asunto(s)
Linfocitos Intraepiteliales , Leucemia , Linfoma , Animales , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/metabolismo , Cadenas beta de Integrinas , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/metabolismo , Leucemia/metabolismo , Linfoma/metabolismo , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Factores de Transcripción/metabolismo
11.
Nat Immunol ; 23(1): 75-85, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937930

RESUMEN

We report a pleiotropic disease due to loss-of-function mutations in RHBDF2, the gene encoding iRHOM2, in two kindreds with recurrent infections in different organs. One patient had recurrent pneumonia but no colon involvement, another had recurrent infectious hemorrhagic colitis but no lung involvement and the other two experienced recurrent respiratory infections. Loss of iRHOM2, a rhomboid superfamily member that regulates the ADAM17 metalloproteinase, caused defective ADAM17-dependent cleavage and release of cytokines, including tumor-necrosis factor and amphiregulin. To understand the diverse clinical phenotypes, we challenged Rhbdf2-/- mice with Pseudomonas aeruginosa by nasal gavage and observed more severe pneumonia, whereas infection with Citrobacter rodentium caused worse inflammatory colitis than in wild-type mice. The fecal microbiota in the colitis patient had characteristic oral species that can predispose to colitis. Thus, a human immunodeficiency arising from iRHOM2 deficiency causes divergent disease phenotypes that can involve the local microbial environment.


Asunto(s)
Proteína ADAM17/genética , Proteínas Portadoras/genética , Enfermedades de Inmunodeficiencia Primaria/genética , Células A549 , Animales , Niño , Preescolar , Citrobacter rodentium/patogenicidad , Colitis/genética , Citocinas/genética , Infecciones por Enterobacteriaceae/genética , Femenino , Células HEK293 , Humanos , Recién Nacido , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Infecciones por Pseudomonas/genética , Pseudomonas aeruginosa/patogenicidad , Transducción de Señal/genética
12.
Science ; 373(6558)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34446580

RESUMEN

The immune system has evolved in the face of microbial exposure. How maternal infection experienced at distinct developmental stages shapes the offspring immune system remains poorly understood. Here, we show that during pregnancy, maternally restricted infection can have permanent and tissue-specific impacts on offspring immunity. Mechanistically, maternal interleukin-6 produced in response to infection can directly impose epigenetic changes on fetal intestinal epithelial stem cells, leading to long-lasting impacts on intestinal immune homeostasis. As a result, offspring of previously infected dams develop enhanced protective immunity to gut infection and increased inflammation in the context of colitis. Thus, maternal infection can be coopted by the fetus to promote long-term, tissue-specific fitness, a phenomenon that may come at the cost of predisposition to inflammatory disorders.


Asunto(s)
Colitis/inmunología , Inmunidad , Interleucina-6/inmunología , Intestinos/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Células Th17/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Animales , Candidiasis/inmunología , Cromatina/metabolismo , Epigénesis Genética , Epigenoma , Femenino , Desarrollo Fetal , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Interleucina-6/sangre , Interleucina-6/farmacología , Mucosa Intestinal/citología , Mucosa Intestinal/embriología , Mucosa Intestinal/inmunología , Intestinos/embriología , Intestinos/microbiología , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal , Salmonelosis Animal/inmunología , Células Madre/inmunología , Células Madre/fisiología , Subgrupos de Linfocitos T/inmunología
13.
Science ; 371(6529): 595-602, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33542131

RESUMEN

Anti-programmed cell death protein 1 (PD-1) therapy provides long-term clinical benefits to patients with advanced melanoma. The composition of the gut microbiota correlates with anti-PD-1 efficacy in preclinical models and cancer patients. To investigate whether resistance to anti-PD-1 can be overcome by changing the gut microbiota, this clinical trial evaluated the safety and efficacy of responder-derived fecal microbiota transplantation (FMT) together with anti-PD-1 in patients with PD-1-refractory melanoma. This combination was well tolerated, provided clinical benefit in 6 of 15 patients, and induced rapid and durable microbiota perturbation. Responders exhibited increased abundance of taxa that were previously shown to be associated with response to anti-PD-1, increased CD8+ T cell activation, and decreased frequency of interleukin-8-expressing myeloid cells. Responders had distinct proteomic and metabolomic signatures, and transkingdom network analyses confirmed that the gut microbiome regulated these changes. Collectively, our findings show that FMT and anti-PD-1 changed the gut microbiome and reprogrammed the tumor microenvironment to overcome resistance to anti-PD-1 in a subset of PD-1 advanced melanoma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Resistencia a Antineoplásicos , Trasplante de Microbiota Fecal , Melanoma/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Neoplasias Cutáneas/terapia , Linfocitos T CD8-positivos/inmunología , Microbioma Gastrointestinal , Humanos , Interleucina-8/inmunología , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Células Mieloides/inmunología , Microambiente Tumoral/inmunología
14.
J Infect Dis ; 224(8): 1405-1409, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33606018

RESUMEN

Unbiased plasma proteomics in a matched case-control study of treated people with human immunodeficiency virus (PWH) revealed the complement cascade as being among the top pathways enriched in PWH. Specific complement components, namely C5, associated significantly with non-AIDS comorbidity prevalence, and did so more strongly than previously established predictive biomarkers.


Asunto(s)
Complemento C5/análisis , Infecciones por VIH/epidemiología , Envejecimiento , Biomarcadores/sangre , Estudios de Casos y Controles , Comorbilidad , VIH , Seronegatividad para VIH , Humanos , Factores Inmunológicos
15.
Hepatology ; 73(3): 1088-1104, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32557834

RESUMEN

BACKGROUND AND AIMS: Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), caused by autoimmune regulator (AIRE) mutations, manifests with chronic mucocutaneous candidiasis (CMC) and multisystem autoimmunity, most often hypoparathyroidism (HP) and adrenal insufficiency (AI). European cohorts previously reported a ~10% prevalence of APECED-associated hepatitis (APAH) with presentations ranging from asymptomatic laboratory derangements to fatal fulminant hepatic failure. Herein, we characterized APAH in a large APECED cohort from the Americas. APPROACH AND RESULTS: Forty-five consecutive patients with APECED were evaluated (2013-2015) at the National Institutes of Health (NIH; NCT01386437). Hepatology consultation assessed hepatic and autoimmune biomarkers and liver ultrasound in all patients. Liver biopsies evaluated autoimmune features and fibrosis. The 16S ribosomal RNA (rRNA) sequencing was performed in 35 patients' stools (12 with and 23 without APAH). Among 43 evaluable patients, 18 (42%) had APAH; in 33.3% of those with APAH, APAH occurred before developing classic APECED diagnostic criteria. At APAH diagnosis, the median age was 7.8 years, and patients manifested with aminotransferase elevation and/or hyperbilirubinemia. All patients with APAH were in clinical remission during their NIH evaluation while receiving immunomodulatory treatment. We found no difference in age, sex, or prevalence of CMC, AI, or HP between patients with or without APAH. Autoantibody positivity against aromatic L-amino acid decarboxylase, cytochrome P450 family 1 subfamily A member 2, histidine decarboxylase (HDC), bactericidal/permeability-increasing fold-containing B1, tryptophan hydroxlase, and 21-hydroxylase (21-OH), and the homozygous c.967_979del13 AIRE mutation were associated with APAH development. Classical serological biomarkers of autoimmune hepatitis (AIH) were only sporadically positive. AIH-like lymphoplasmacytic inflammation with mild fibrosis was the predominant histological feature. Stool microbiome analysis found Slackia and Acidaminococcus in greater abundance in patients with APAH. CONCLUSIONS: APAH is more common than previously described, may present early before classic APECED manifestations, and most often manifests with milder, treatment-responsive disease. Several APECED-associated autoantibodies, but not standard AIH-associated biomarkers, correlate with APAH.


Asunto(s)
Hepatitis Autoinmune/etiología , Poliendocrinopatías Autoinmunes/complicaciones , Adolescente , Adulto , Américas , Autoanticuerpos/inmunología , Biomarcadores/sangre , Biopsia , Femenino , Eliminación de Gen , Hepatitis Autoinmune/patología , Hepatitis Autoinmune/terapia , Humanos , Inmunoterapia , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Masculino , Poliendocrinopatías Autoinmunes/genética , Poliendocrinopatías Autoinmunes/patología , Poliendocrinopatías Autoinmunes/terapia , Adulto Joven
16.
Nature ; 587(7834): 448-454, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33149306

RESUMEN

Low concordance between studies that examine the role of microbiota in human diseases is a pervasive challenge that limits the capacity to identify causal relationships between host-associated microorganisms and pathology. The risk of obtaining false positives is exacerbated by wide interindividual heterogeneity in microbiota composition1, probably due to population-wide differences in human lifestyle and physiological variables2 that exert differential effects on the microbiota. Here we infer the greatest, generalized sources of heterogeneity in human gut microbiota profiles and also identify human lifestyle and physiological characteristics that, if not evenly matched between cases and controls, confound microbiota analyses to produce spurious microbial associations with human diseases. We identify alcohol consumption frequency and bowel movement quality as unexpectedly strong sources of gut microbiota variance that differ in distribution between healthy participants and participants with a disease and that can confound study designs. We demonstrate that for numerous prevalent, high-burden human diseases, matching cases and controls for confounding variables reduces observed differences in the microbiota and the incidence of spurious associations. On this basis, we present a list of host variables that we recommend should be captured in human microbiota studies for the purpose of matching comparison groups, which we anticipate will increase robustness and reproducibility in resolving the members of the gut microbiota that are truly associated with human disease.


Asunto(s)
Factores de Confusión Epidemiológicos , Análisis de Datos , Dieta , Enfermedad , Microbioma Gastrointestinal/fisiología , Estilo de Vida , Aprendizaje Automático , Adulto , Anciano , Anciano de 80 o más Años , Consumo de Bebidas Alcohólicas , Área Bajo la Curva , Índice de Masa Corporal , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2 , Heces/microbiología , Femenino , Motilidad Gastrointestinal , Humanos , Masculino , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Curva ROC , Características de la Residencia , Adulto Joven
17.
Cell ; 178(5): 1088-1101.e15, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442402

RESUMEN

Mammals evolved in the face of fluctuating food availability. How the immune system adapts to transient nutritional stress remains poorly understood. Here, we show that memory T cells collapsed in secondary lymphoid organs in the context of dietary restriction (DR) but dramatically accumulated within the bone marrow (BM), where they adopted a state associated with energy conservation. This response was coordinated by glucocorticoids and associated with a profound remodeling of the BM compartment, which included an increase in T cell homing factors, erythropoiesis, and adipogenesis. Adipocytes, as well as CXCR4-CXCL12 and S1P-S1P1R interactions, contributed to enhanced T cell accumulation in BM during DR. Memory T cell homing to BM during DR was associated with enhanced protection against infections and tumors. Together, this work uncovers a fundamental host strategy to sustain and optimize immunological memory during nutritional challenges that involved a temporal and spatial reorganization of the memory pool within "safe haven" compartments.


Asunto(s)
Médula Ósea/metabolismo , Memoria Inmunológica , Animales , Médula Ósea/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Restricción Calórica/veterinaria , Línea Celular Tumoral , Quimiocina CXCL12/metabolismo , Dieta Reductora/veterinaria , Metabolismo Energético , Regulación de la Expresión Génica , Glucocorticoides , Melanoma Experimental/mortalidad , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR4/metabolismo , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
18.
J Immunol ; 203(3): 686-695, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31243087

RESUMEN

The thymus is critical for the establishment of the adaptive immune system and the development of a diverse T cell repertoire. T cell development depends upon cell-cell interactions with epithelial cells in the thymus. The thymus is composed of two different types of epithelial cells: cortical and medullary epithelial cells. Both of these express and critically depend on the transcription factor Foxn1 Foxn1 is also expressed in the hair follicle, and disruption of Foxn1 function in mice results in severe thymic developmental defects and the hairless (nude) phenotype. Despite its importance, little is known about the direct regulation of Foxn1 expression. In this study, we identify a cis-regulatory element (RE) critical for expression of Foxn1 in mouse thymic epithelial cells but dispensable for expression in hair follicles. Analysis of chromatin accessibility, histone modifications, and sequence conservation identified regions within the first intron of Foxn1 that possessed the characteristics of REs. Systematic knockout of candidate regions lead us to identify a 1.6 kb region that, when deleted, results in a near total disruption of thymus development. Interestingly, Foxn1 expression and function in the hair follicle were unaffected. RNA fluorescent in situ hybridization showed a near complete loss of Foxn1 mRNA expression in the embryonic thymic bud. Our studies have identified a genomic RE with thymic-specific control of Foxn1 gene expression.


Asunto(s)
Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Elementos Reguladores de la Transcripción/genética , Linfocitos T/inmunología , Timo/metabolismo , Animales , Factores de Transcripción Forkhead/biosíntesis , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Folículo Piloso/metabolismo , Ratones , Ratones Noqueados , Ratones Desnudos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Linfocitos T/citología , Timo/citología
19.
Sci Transl Med ; 10(463)2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30333238

RESUMEN

Periodontitis is one of the most common human inflammatory diseases, yet the mechanisms that drive immunopathology and could be therapeutically targeted are not well defined. Here, we demonstrate an expansion of resident memory T helper 17 (TH17) cells in human periodontitis. Phenocopying humans, TH17 cells expanded in murine experimental periodontitis through local proliferation. Unlike homeostatic oral TH17 cells, which accumulate in a commensal-independent and interleukin-6 (IL-6)-dependent manner, periodontitis-associated expansion of TH17 cells was dependent on the local dysbiotic microbiome and required both IL-6 and IL-23. TH17 cells and associated neutrophil accumulation were necessary for inflammatory tissue destruction in experimental periodontitis. Genetic or pharmacological inhibition of TH17 cell differentiation conferred protection from immunopathology. Studies in a unique patient population with a genetic defect in TH17 cell differentiation established human relevance for our murine experimental studies. In the oral cavity, human TH17 cell defects were associated with diminished periodontal inflammation and bone loss, despite increased prevalence of recurrent oral fungal infections. Our study highlights distinct functions of TH17 cells in oral immunity and inflammation and paves the way to a new targeted therapeutic approach for the treatment of periodontitis.


Asunto(s)
Disbiosis/inmunología , Disbiosis/microbiología , Microbiota , Mucosa Bucal/inmunología , Mucosa Bucal/patología , Células Th17/inmunología , Animales , Bacterias/metabolismo , Resorción Ósea/microbiología , Resorción Ósea/patología , Resorción Ósea/prevención & control , Diferenciación Celular , Humanos , Inflamación/inmunología , Inflamación/patología , Interleucina-23/metabolismo , Interleucina-6/metabolismo , Ratones , Neutrófilos/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Periodontitis/inmunología , Periodontitis/microbiología , Periodontitis/patología
20.
Nat Immunol ; 19(9): 986-1000, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30127432

RESUMEN

Gain-of-function mutations in the gene encoding the phosphatidylinositol-3-OH kinase catalytic subunit p110δ (PI3Kδ) result in a human primary immunodeficiency characterized by lymphoproliferation, respiratory infections and inefficient responses to vaccines. However, what promotes these immunological disturbances at the cellular and molecular level remains unknown. We generated a mouse model that recapitulated major features of this disease and used this model and patient samples to probe how hyperactive PI3Kδ fosters aberrant humoral immunity. We found that mutant PI3Kδ led to co-stimulatory receptor ICOS-independent increases in the abundance of follicular helper T cells (TFH cells) and germinal-center (GC) B cells, disorganized GCs and poor class-switched antigen-specific responses to immunization, associated with altered regulation of the transcription factor FOXO1 and pro-apoptotic and anti-apoptotic members of the BCL-2 family. Notably, aberrant responses were accompanied by increased reactivity to gut bacteria and a broad increase in autoantibodies that were dependent on stimulation by commensal microbes. Our findings suggest that proper regulation of PI3Kδ is critical for ensuring optimal host-protective humoral immunity despite tonic stimulation from the commensal microbiome.


Asunto(s)
Linfocitos B/fisiología , Microbioma Gastrointestinal/inmunología , Centro Germinal/fisiología , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Linfocitos T Colaboradores-Inductores/fisiología , Animales , Autoanticuerpos/sangre , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I/genética , Modelos Animales de Enfermedad , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Humanos , Inmunidad Humoral/genética , Cambio de Clase de Inmunoglobulina/genética , Síndromes de Inmunodeficiencia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA