Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunol ; 212(9): 1442-1449, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38436421

RESUMEN

Protein arginine methyltransferases (PRMTs) modify diverse protein targets and regulate numerous cellular processes; yet, their contributions to individual effector T cell responses during infections are incompletely understood. In this study, we identify PRMT5 as a critical regulator of CD4+ T follicular helper cell (Tfh) responses during influenza virus infection in mice. Conditional PRMT5 deletion in murine T cells results in an almost complete ablation of both Tfh and T follicular regulatory populations and, consequently, reduced B cell activation and influenza-specific Ab production. Supporting a potential mechanism, we observe elevated surface expression of IL-2Rα on non-T regulatory effector PRMT5-deficient T cells. Notably, IL-2 signaling is known to negatively impact Tfh differentiation. Collectively, our findings identify PRMT5 as a prominent regulator of Tfh programming, with potential causal links to IL-2 signaling.


Asunto(s)
Gripe Humana , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Animales , Humanos , Ratones , Diferenciación Celular , Centro Germinal , Interleucina-2/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Células T Auxiliares Foliculares
2.
Mol Cancer Ther ; 23(4): 421-435, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38030380

RESUMEN

IL12 is a proinflammatory cytokine, that has shown promising antitumor activity in humans by promoting the recruitment and activation of immune cells in tumors. However, the systemic administration of IL12 has been accompanied by considerable toxicity, prompting interest in researching alternatives to drive preferential IL12 bioactivity in the tumor. Here, we have generated XTX301, a tumor-activated IL12 linked to the human Fc protein via a protease cleavable linker that is pharmacologically inactivated by an IL12 receptor subunit beta 2 masking domain. In vitro characterization demonstrates multiple matrix metalloproteases, as well as human primary tumors cultured as cell suspensions, can effectively activate XTX301. Intravenous administration of a mouse surrogate mXTX301 demonstrated significant tumor growth inhibition (TGI) in inflamed and non-inflamed mouse models without causing systemic toxicities. The superiority of mXTX301 in mediating TGI compared with non-activatable control molecules and the greater percentage of active mXTX301 in tumors versus other organs further confirms activation by the tumor microenvironment-associated proteases in vivo. Pharmacodynamic characterization shows tumor selective increases in inflammation and upregulation of immune-related genes involved in IFNγ cell signaling, antigen processing, presentation, and adaptive immune response. XTX301 was tolerated following four repeat doses up to 2.0 mg/kg in a nonhuman primate study; XTX301 exposures were substantially higher than those at the minimally efficacious dose in mice. Thus, XTX301 has the potential to achieve potent antitumor activity while widening the therapeutic index of IL12 treatment and is currently being evaluated in a phase I clinical trial.


Asunto(s)
Interleucina-12 , Neoplasias , Humanos , Ratones , Animales , Interleucina-12/metabolismo , Neoplasias/tratamiento farmacológico , Citocinas , Transducción de Señal , Índice Terapéutico , Microambiente Tumoral
3.
NAR Cancer ; 5(3): zcad039, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37519629

RESUMEN

CCNE1 amplification is a common alteration in high-grade serous ovarian cancer and occurs in 15-20% of these tumors. These amplifications are mutually exclusive with homologous recombination deficiency, and, as they have intact homologous recombination, are intrinsically resistant to poly (ADP-ribose) polymerase inhibitors or chemotherapy agents. Understanding the molecular mechanisms that lead to this mutual exclusivity may reveal therapeutic vulnerabilities that could be leveraged in the clinic in this still underserved patient population. Here, we demonstrate that CCNE1-amplified high-grade serous ovarian cancer cells rely on homologous recombination to repair collapsed replication forks. Cyclin-dependent kinase 2, the canonical partner of cyclin E1, uniquely regulates homologous recombination in this genetic context, and as such cyclin-dependent kinase 2 inhibition synergizes with DNA damaging agents in vitro and in vivo. We demonstrate that combining a selective cyclin-dependent kinase 2 inhibitor with a DNA damaging agent could be a powerful tool in the clinic for high-grade serous ovarian cancer.

4.
Nat Metab ; 4(10): 1322-1335, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36192601

RESUMEN

γ-Aminobutyrate (GAB), the biochemical form of (GABA) γ-aminobutyric acid, participates in shaping physiological processes, including the immune response. How GAB metabolism is controlled to mediate such functions remains elusive. Here we show that GAB is one of the most abundant metabolites in CD4+ T helper 17 (TH17) and induced T regulatory (iTreg) cells. GAB functions as a bioenergetic and signalling gatekeeper by reciprocally controlling pro-inflammatory TH17 cell and anti-inflammatory iTreg cell differentiation through distinct mechanisms. 4-Aminobutyrate aminotransferase (ABAT) funnels GAB into the tricarboxylic acid (TCA) cycle to maximize carbon allocation in promoting TH17 cell differentiation. By contrast, the absence of ABAT activity in iTreg cells enables GAB to be exported to the extracellular environment where it acts as an autocrine signalling metabolite that promotes iTreg cell differentiation. Accordingly, ablation of ABAT activity in T cells protects against experimental autoimmune encephalomyelitis (EAE) progression. Conversely, ablation of GABAA receptor in T cells worsens EAE. Our results suggest that the cell-autonomous control of GAB on CD4+ T cells is bimodal and consists of the sequential action of two processes, ABAT-dependent mitochondrial anaplerosis and the receptor-dependent signalling response, both of which are required for T cell-mediated inflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Células Th17 , Animales , Células Th17/metabolismo , 4-Aminobutirato Transaminasa/metabolismo , Receptores de GABA-A/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Inflamación/metabolismo , Antiinflamatorios/farmacología , Metabolismo Energético , Aminobutiratos/metabolismo , Carbono/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Ácidos Tricarboxílicos/metabolismo
5.
Infect Immun ; 90(4): e0059721, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35311543

RESUMEN

Streptococcus pneumoniae is a Gram-positive, encapsulated bacterium that is a significant cause of disease burden in pediatric and elderly populations. The rise in unencapsulated disease-causing strains and antimicrobial resistance in S. pneumoniae has increased the need for developing new antimicrobial strategies. Recent work by our laboratory has identified N,N-dimethyldithiocarbamate (DMDC) as a copper-dependent antimicrobial against bacterial, fungal, and parasitic pathogens. As a bactericidal antibiotic against S. pneumoniae, DMDC's ability to work as a copper-dependent antibiotic and its ability to work in vivo warranted further investigation. Here, our group studied the mechanisms of action of DMDC under various medium and excess-metal conditions and investigated DMDC's interactions with the innate immune system in vitro and in vivo. Of note, we found that DMDC plus copper significantly increased the internal copper concentration, hydrogen peroxide stress, nitric oxide stress, and the in vitro macrophage killing efficiency and decreased capsule. Furthermore, we found that in vivo DMDC treatment increased the quantity of innate immune cells in the lung during infection. Taken together, this study provides mechanistic insights regarding DMDC's activity as an antibiotic at the host-pathogen interface.


Asunto(s)
Antiinfecciosos , Infecciones Neumocócicas , Anciano , Antibacterianos , Antiinfecciosos/farmacología , Niño , Cobre , Dimetilditiocarbamato , Humanos , Macrófagos , Streptococcus pneumoniae
6.
J Neurosci ; 42(2): 325-348, 2022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-34819339

RESUMEN

Globally, more than 67 million people are living with the effects of ischemic stroke. Importantly, many stroke survivors develop a chronic inflammatory response that may contribute to cognitive impairment, a common and debilitating sequela of stroke that is insufficiently studied and currently untreatable. 2-Hydroxypropyl-ß-cyclodextrin (HPßCD) is an FDA-approved cyclic oligosaccharide that can solubilize and entrap lipophilic substances. The goal of the present study was to determine whether the repeated administration of HPßCD curtails the chronic inflammatory response to stroke by reducing lipid accumulation within stroke infarcts in a distal middle cerebral artery occlusion mouse model of stroke. To achieve this goal, we subcutaneously injected young adult and aged male mice with vehicle or HPßCD 3 times per week, with treatment beginning 1 week after stroke. We evaluated mice at 7 weeks following stroke using immunostaining, RNA sequencing, lipidomic, and behavioral analyses. Chronic stroke infarct and peri-infarct regions of HPßCD-treated mice were characterized by an upregulation of genes involved in lipid metabolism and a downregulation of genes involved in innate and adaptive immunity, reactive astrogliosis, and chemotaxis. Correspondingly, HPßCD reduced the accumulation of lipid droplets, T lymphocytes, B lymphocytes, and plasma cells in stroke infarcts. Repeated administration of HPßCD also preserved NeuN immunoreactivity in the striatum and thalamus and c-Fos immunoreactivity in hippocampal regions. Additionally, HPßCD improved recovery through the protection of hippocampal-dependent spatial working memory and reduction of impulsivity. These results indicate that systemic HPßCD treatment following stroke attenuates chronic inflammation and secondary neurodegeneration and prevents poststroke cognitive decline.SIGNIFICANCE STATEMENT Dementia is a common and debilitating sequela of stroke. Currently, there are no available treatments for poststroke dementia. Our study shows that lipid metabolism is disrupted in chronic stroke infarcts, which causes an accumulation of uncleared lipid debris and correlates with a chronic inflammatory response. To our knowledge, these substantial changes in lipid homeostasis have not been previously recognized or investigated in the context of ischemic stroke. We also provide a proof of principle that solubilizing and entrapping lipophilic substances using HPßCD could be an effective strategy for treating chronic inflammation after stroke and other CNS injuries. We propose that using HPßCD for the prevention of poststroke dementia could improve recovery and increase long-term quality of life in stroke sufferers.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/uso terapéutico , Encéfalo/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Factores de Edad , Animales , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Resultado del Tratamiento
7.
Cell Host Microbe ; 29(3): 325-326, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33705703

RESUMEN

In this COVID era, when many struggle to work remotely, methotrexate (MTX) has been doing so all along. In this issue of Cell Host & Microbe, Nayak et al. discover that MTX reduces immune activation due to off-target effects on the gut microbiota, potentially explaining MTX's anti-inflammatory effects.


Asunto(s)
COVID-19 , Microbioma Gastrointestinal , Bacterias , Humanos , Metotrexato , SARS-CoV-2
8.
Nat Rev Rheumatol ; 17(4): 224-237, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33674813

RESUMEN

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder that primarily affects the joints. One hypothesis for the pathogenesis of RA is that disease begins at mucosal sites as a consequence of interactions between the mucosal immune system and an aberrant local microbiota, and then transitions to involve the synovial joints. Alterations in the composition of the microbial flora in the lungs, mouth and gut in individuals with preclinical and established RA suggest a role for mucosal dysbiosis in the development and perpetuation of RA, although establishing whether these alterations are the specific consequence of intestinal involvement in the setting of a systemic inflammatory process, or whether they represent a specific localization of disease, is an ongoing challenge. Data from mouse models of RA and investigations into the preclinical stages of disease also support the hypothesis that these alterations to the microbiota predate the onset of disease. In addition, several therapeutic options widely used for the treatment of RA are associated with alterations in intestinal microbiota, suggesting that modulation of intestinal microbiota and/or intestinal barrier function might be useful in preventing or treating RA.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/microbiología , Articulaciones/patología , Microbiota/inmunología , Membrana Mucosa/inmunología , Animales , Antirreumáticos/efectos adversos , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Disbiosis/complicaciones , Disbiosis/inmunología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Humanos , Articulaciones/inmunología , Masculino , Ratones , Microbiota/efectos de los fármacos , Membrana Mucosa/microbiología , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Líquido Sinovial/inmunología
9.
J Immunol ; 206(5): 941-952, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33462137

RESUMEN

Autoantibodies play a major pathogenic role in rheumatoid arthritis. T follicular helper (Tfh) cells promote germinal center B cell and Ab responses. Excessive Tfh cell responses lead to autoimmunity, and therefore, counterregulation is crucial. T follicular regulatory (Tfr) cells, mainly differentiated from T regulatory cells, can negatively regulate Tfh and germinal center B cells. Dysbiosis is involved in rheumatoid arthritis's pathogenesis. We previously demonstrated that the gut microbiota, segmented filamentous bacteria (SFB), promote autoimmune arthritis by inducing Tfh cells. However, little is known regarding whether gut microbiota influence systemic (nongut) Tfr cells, impacting gut-distal autoimmunity. In this study, using SFB in autoimmune arthritic K/BxN mice, we demonstrated that SFB-induced arthritis is linked to the reduction of Tfr cells' CTLA-4, the key regulatory molecule of Tfr cells. This SFB-mediated CTLA-4 reduction is associated with increased Tfr glycolytic activity, and glycolytic inhibition increases Tfr cells' CTLA-4 levels and reduces arthritis. The surface expression of CTLA-4 is tied to TCR signaling strength, and we discovered that SFB-reduced CTLA-4 is associated with a reduction of Nur77, an indicator of TCR signaling strength. Nur77 is known for repressing glycolytic activity. Using a loss-of-function study, we demonstrated that Nur77+/- haplodeficiency increases glycolysis and reduces CTLA-4 on Tfr cells, which is associated with increased arthritis and anti-glucose-6-phosphate isomerase titers. Tfr-specific deletion (KRN.Foxp3CreBcl-6fl/fl) in autoimmune condition reveals that Tfr cells repress arthritis, Tfh cells, and autoantibody responses and that SFB can mitigate this repression. Overall, these findings demonstrated that gut microbiota distally impact systemic autoimmunity by fine-tuning Tfr cells.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/microbiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Autoinmunidad/inmunología , Microbioma Gastrointestinal/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoanticuerpos/inmunología , Bacterias/inmunología , Antígeno CTLA-4/inmunología , Diferenciación Celular/inmunología , Centro Germinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
10.
JCI Insight ; 5(5)2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32161196

RESUMEN

CD137 (4-1BB) is a member of the TNFR superfamily that represents a promising target for cancer immunotherapy. Recent insights into the function of TNFR agonist antibodies implicate epitope, affinity, and IgG subclass as critical features, and these observations help explain the limited activity and toxicity seen with clinically tested CD137 agonists. Here, we describe the preclinical characterization of CTX-471, a fully human IgG4 agonist of CD137 that engages a unique epitope that is shared by human, cynomolgus monkey, and mouse and is associated with a differentiated pharmacology and toxicology profile. In vitro, CTX-471 increased IFN-γ production by human T cells in an Fcγ receptor-dependent (FcγR-dependent) manner, displaying an intermediate level of activity between 2 clinical-stage anti-CD137 antibodies. In mice, CTX-471 exhibited curative monotherapy activity in various syngeneic tumor models and showed a unique ability to cure mice of very large (~500 mm3) tumors compared with validated antibodies against checkpoints and TNFR superfamily members. Extremely high doses of CTX-471 were well tolerated, with no signs of hepatic toxicity. Collectively, these data demonstrate that CTX-471 is a unique CD137 agonist that displays an excellent safety profile and an unprecedented level of monotherapy efficacy against very large tumors.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Mapeo Epitopo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Inmunoterapia/efectos adversos , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/metabolismo , Macaca fascicularis , Ratones , Ratones Desnudos , Neoplasias/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/química , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Oncogene ; 39(12): 2539-2549, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31988451

RESUMEN

Cancer-associated fibroblasts (CAFs) are activated fibroblasts that constitute the major components of tumor microenvironment (TME) and play crucial roles in tumor development and metastasis. Here, we generated fibroblast-specific inducible focal adhesion kinase (FAK) knockout (cKO) mice in a breast cancer model to study potential role and mechanisms of FAK signaling in CAF to promote breast cancer metastasis in vivo. While not affecting primary tumor development and growth, FAK deletion significantly suppressed breast cancer metastasis in vivo. Analyses of CAFs derived from cKO mice as well as human CAFs showed that FAK is required for their activity to promote mammary tumor cell migration. We further showed that FAK ablation in CAFs decreased exosome functions to promote tumor cell migration and other activities, which could contribute to the reduced metastasis observed in cKO mice. Lastly, profiling of miRs from CAF exosomes showed alterations of several exosomal miRs in FAK-null CAFs, and further analysis suggested that miR-16 and miR-148a enriched in exosomes from FAK-null CAFs contribute to the reduced tumor cell activities and metastasis. Together, these results identify a new role for FAK signaling in CAFs that regulate their intercellular communication with tumor cells to promote breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Movimiento Celular , Quinasa 1 de Adhesión Focal/metabolismo , MicroARNs/metabolismo , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/metabolismo , Animales , Neoplasias de la Mama/patología , Comunicación Celular , Línea Celular Tumoral , Exosomas , Femenino , Quinasa 1 de Adhesión Focal/genética , Humanos , Ratones , Ratones Noqueados , Transducción de Señal
12.
Nanomaterials (Basel) ; 9(9)2019 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-31484362

RESUMEN

A hollow nanostructure is attractive and important in different fields of applications, for instance, solar cells, sensors, supercapacitors, electronics, and biomedical, due to their unique structure, large available interior space, low bulk density, and stable physicochemical properties. Hence, the need to prepare hollow nanotubes is more important. In this present study, we have prepared CuCrO2 hollow nanotubes by simple approach. The CuCrO2 hollow nanotubes were prepared by applying electrospun Al2O3 fibers as a template for the first time. Copper chromium ions were dip-coated on the surface of electrospun-derived Al2O3 fibers and annealed at 600 °C in vacuum to form Al2O3-CuCrO2 core-shell nanofibers. The CuCrO2 hollow nanotubes were obtained by removing Al2O3 cores by sulfuric acid wet etching while preserving the rest of original structures. The structures of the CuCrO2-coated Al2O3 core-shell nanofibers and CuCrO2 hollow nanotubes were identified side-by-side by X-ray diffraction, field emission scanning electron microscopy, and transmission electron microscopy. The CuCrO2 hollow nanotubes may find applications in electrochemistry, catalysis, and biomedical application. This hollow nanotube preparation method could be extended to the preparation of other hollow nanotubes, fibers, and spheres.

13.
Front Immunol ; 10: 411, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30949163

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease that affects ~1% of the world's population. B cells and autoantibodies play an important role in the pathogenesis of RA. The P2RX7 receptor is an ATP-gated cation channel and its activation results in the release of pro-inflammatory molecules. Thus, antagonists of P2RX7 have been considered to have potential as novel anti-inflammatory therapies. Although originally identified for its role in innate immunity, P2RX7 has recently been found to negatively control Peyer's patches (PP) T follicular helper cells (Tfh), which specialize in helping B cells, under homeostatic conditions. We have previously demonstrated that PP Tfh cells are required for the augmentation of autoimmune arthritis mediated by gut commensal segmented filamentous bacteria (SFB). Thus, we hypothesized that P2RX7 is required to control autoimmune disease by keeping the Tfh cell response in check. To test our hypothesis, we analyzed the impact of P2RX7 deficiency in vivo using both the original K/BxN autoimmune arthritis model and T cell transfers in the K/BxN system. We also examined the impact of P2RX7 ablation on autoimmune development in the presence of the gut microbiota SFB. Our data illustrate that contrary to exerting an anti-inflammatory effect, P2RX7 deficiency actually enhances autoimmune arthritis. Interestingly, SFB colonization can negate the difference in disease severity between WT and P2RX7-deficient mice. We further demonstrated that P2RX7 ablation in the absence of SFB caused reduced apoptotic Tfh cells and enhanced the Tfh response, leading to an increase in autoantibody production. It has been shown that activation of TIGIT, a well-known T cell exhaustion marker, up-regulates anti-apoptotic molecules and promotes T cell survival. We demonstrated that the reduced apoptotic phenotype of P2rx7-/- Tfh cells is associated with their increased expression of TIGIT. This suggested that while P2RX7 was regulating the Tfh population by promoting cell death, TIGIT may have been opposing P2RX7 by inhibiting cell death. Together, these results demonstrated that systemic administration of general P2RX7 antagonists may have detrimental effects in autoimmune therapies, especially in Tfh cell-dependent autoimmune diseases, and cell-specific targeting of P2RX7 should be considered in order to achieve efficacy for P2RX7-related therapy.


Asunto(s)
Artritis Reumatoide/inmunología , Microbioma Gastrointestinal/inmunología , Receptores Purinérgicos P2X7/deficiencia , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Apoptosis/inmunología , Artritis Reumatoide/microbiología , Autoanticuerpos/inmunología , Femenino , Masculino , Ratones , Ratones Transgénicos , Ganglios Linfáticos Agregados/inmunología , Receptores Inmunológicos/metabolismo , Receptores Purinérgicos P2X7/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
14.
Immunology ; 156(4): 305-318, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30560993

RESUMEN

The immune system is essential for maintaining a delicate balance between eliminating pathogens and maintaining tolerance to self-tissues to avoid autoimmunity. An enormous and complex community of gut microbiota provides essential health benefits to the host, particularly by regulating immune homeostasis. Many of the metabolites derived from commensals can impact host health by directly regulating the immune system. Many autoimmune diseases arise from an imbalance between pathogenic effector T cells and regulatory T (Treg) cells. Recent interest has emerged in understanding how cross-talk between gut microbiota and the host immune system promotes autoimmune development by controlling the differentiation and plasticity of T helper and Treg cells. At the molecular level, our recent study, along with others, demonstrates that asymptomatic colonization by commensal bacteria in the gut is capable of triggering autoimmune disease by molecular mimicking self-antigen and skewing the expression of dual T-cell receptors on T cells. Dysbiosis, an imbalance of the gut microbiota, is involved in autoimmune development in both mice and humans. Although it is well known that dysbiosis can impact diseases occurring within the gut, growing literature suggests that dysbiosis also causes the development of gut-distal/non-gut autoimmunity. In this review, we discuss recent advances in understanding the potential molecular mechanisms whereby gut microbiota induces autoimmunity, and the evidence that the gut microbiota triggers gut-distal autoimmune diseases.


Asunto(s)
Autoinmunidad/inmunología , Microbioma Gastrointestinal/inmunología , Linfocitos T/inmunología , Animales , Disbiosis/inmunología , Humanos
15.
Artículo en Inglés | MEDLINE | ID: mdl-29755958

RESUMEN

Streptococcus pneumoniae causes infection-related mortality worldwide. Immunocompromised individuals, including young children, the elderly, and those with immunodeficiency, are especially vulnerable, yet little is known regarding S. pneumoniae-related pathogenesis and protection in immunocompromised hosts. Recently, strong interest has emerged in the gut microbiota's impact on lung diseases, or the "gut-lung axis." However, the mechanisms of gut microbiota protection against gut-distal lung diseases like pneumonia remain unclear. We investigated the role of the gut commensal, segmented filamentous bacteria (SFB), against pneumococcal pneumonia in immunocompetent and immunocompromised mouse models. For the latter, we chose the Rag-/- model, with adaptive immune deficiency. Immunocompetent adaptive protection against S. pneumoniae infection is based on antibodies against pneumococcal capsular polysaccharides, prototypical T cell independent-II (TI-II) antigens. Although SFB colonization enhanced TI-II antibodies in C57BL/6 mice, our data suggest that SFB did not further protect these immunocompetent animals. Indeed, basal B cell activity in hosts without SFB is sufficient for essential protection against S. pneumoniae. However, in immunocompromised Rag-/- mice, we demonstrate a gut-lung axis of communication, as SFB influenced lung protection by regulating innate immunity. Neutrophil resolution is crucial to recovery, since an unchecked neutrophil response causes severe tissue damage. We found no early neutrophil recruitment differences between hosts with or without SFB; however, we observed a significant drop in lung neutrophils in the resolution phase of S. pneumoniae infection, which corresponded with lower CD47 expression, a molecule that inhibits phagocytosis of apoptotic cells, in SFB-colonized Rag-/- mice. SFB promoted a shift in lung neutrophil phenotype from inflammatory neutrophils expressing high levels of CD18 and low levels of CD62L, to pro-resolution neutrophils with low CD18 and high CD62L. Blocking CD47 in SFB(-) mice increased pro-resolution neutrophils, suggesting CD47 down-regulation may be one neutrophil-modulating mechanism SFB utilizes. The SFB-induced lung neutrophil phenotype remained similar with heat-inactivated S. pneumoniae treatment, indicating these SFB-induced changes in neutrophil phenotype during the resolution phase are not simply secondary to better bacterial clearance in SFB(+) than SFB(-) mice. Together, these data demonstrate that the gut commensal SFB may provide much-needed protection in immunocompromised hosts in part by promoting neutrophil resolution post lung infection.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Microbioma Gastrointestinal/inmunología , Neutrófilos/inmunología , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/inmunología , Animales , Cápsulas Bacterianas/inmunología , Antígeno CD47/metabolismo , Modelos Animales de Enfermedad , Huésped Inmunocomprometido , Selectina L , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Fagocitosis/inmunología , Polisacáridos Bacterianos/inmunología
16.
Sci Rep ; 8(1): 2550, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416084

RESUMEN

Focal adhesion kinase (FAK) is essential in embryonic angiogenesis by regulating endothelial cell (EC) survival and barrier functions through its kinase-independent and -dependent activities. Here, we generated EC-specific tamoxifen-inducible FAK knockout and FAK kinase-defective (KD) mutant knockin mice to investigate the role of FAK and its kinase activity in angiogenesis of adult animals. Unlike previous observations of their differential defects in embryonic vascular development, both FAK ablation and inactivation of its kinase activity resulted in deficient angiogenesis in wound-healing as well as retinal angiogenesis models. Consistent with these phenotypes, loss of FAK or its kinase activity decreased EC proliferation and migration to similar extents, suggesting FAK primarily acts as a kinase for the regulation of adult EC-mediated angiogenesis. Further mechanistic analyses were carried out using an established mouse EC line MS1 cells. Interestingly, we found that FAK regulated the expression of VEGFR2, a central mediator of various EC functions and angiogenesis, which requires both FAK kinase activity and its translocation into the nucleus. Moreover, nuclear FAK was detected in the RNA polymerase II complex associated with VEGFR2 promoter, suggesting its direct participation in the transcriptional regulation of VEGFR2. Together, our results provide significant insights into the signaling mechanisms of FAK in angiogenesis that may contribute to future design of more effective angiogenesis related therapy.


Asunto(s)
Células Endoteliales/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Neovascularización Retiniana/genética , Transcripción Genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Movimiento Celular , Núcleo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/fisiología , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
17.
Ann N Y Acad Sci ; 1417(1): 57-70, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28984367

RESUMEN

To maintain health, the immune system must maintain a delicate balance between eliminating invading pathogens and avoiding immune disorders such as autoimmunity and allergies. The gut microbiota provide essential health benefits to the host, particularly by regulating immune homeostasis. Dysbiosis, an alteration and imbalance of the gut microbiota, is associated with the development of several autoimmune diseases in both mice and humans. In this review, we discuss recent advances in understanding how certain factors, such as age and gender, affect the gut microbiota, which in turn can influence the development of autoimmune diseases. The age factor in microbiota-dependent immune disorders indicates a window of opportunity for future diagnostic and therapeutic approaches. We also discuss unique commensal bacteria with strong immunomodulatory activity. Finally, we provide an overview of the potential molecular mechanisms whereby gut microbiota induce autoimmunity, as well as the evidence that gut microbiota trigger extraintestinal diseases by inducing the migration of gut-derived immune cells. Elucidating the interaction of gut microbiota and the host immune system will help us understand the pathogenesis of immune disorders, and provide us with new foundations to develop novel immuno- or microbe-targeted therapies.


Asunto(s)
Interacciones Microbiota-Huesped/inmunología , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/microbiología , Factores de Edad , Animales , Autoinmunidad , Disbiosis/inmunología , Disbiosis/microbiología , Epigénesis Genética , Ácidos Grasos Volátiles/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped/genética , Humanos , Enfermedades del Sistema Inmune/genética , Inmunidad Innata , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino , Ratones , Modelos Inmunológicos , Imitación Molecular/inmunología , Factores Sexuales , Tretinoina/inmunología , Ácido Úrico/inmunología
18.
Cell Host Microbe ; 22(5): 697-704.e4, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29120746

RESUMEN

Lung complications are a major cause of rheumatoid arthritis-related mortality. Involvement of gut microbiota in lung diseases by the gut-lung axis has been widely observed, but the underlying mechanism remains mostly unknown. Using an autoimmune arthritis model, we show that a constituent of the gut microbiota, segmented filamentous bacteria (SFB), distantly provoke lung pathology. SFB induce autoantibodies in lung during the pre-arthritic phase, and SFB-dependent lung pathology requires the T helper 17 (Th17) responses. SFB-induced gut Th17 cells are preferentially recruited to lung over spleen due to robust expression in the lung of the Th17 chemoattractant, CCL20. Additionally, we found that in peripheral tissues, SFB selectively expand dual T cell receptor (TCR)-expressing Th17 cells recognizing both an SFB epitope and self-antigen, thus augmenting autoimmunity. This study reveals mechanisms for commensal-mediated gut-lung crosstalk and dual TCR-based autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Autoinmunidad , Bacterias/inmunología , Microbioma Gastrointestinal/inmunología , Pulmón/inmunología , Células Th17/inmunología , Animales , Apoptosis/inmunología , Artritis Reumatoide/inmunología , Artritis Reumatoide/microbiología , Autoanticuerpos , Bacterias/patogenicidad , Diferenciación Celular/inmunología , Proliferación Celular , Quimiocina CCL20/metabolismo , Modelos Animales de Enfermedad , Heces/microbiología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Bazo , Simbiosis , Células Th17/metabolismo
19.
Arthritis Res Ther ; 19(1): 188, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28810929

RESUMEN

BACKGROUND: Age is an important risk factor for rheumatoid arthritis (RA), which often develops in middle age. However, how age-associated changes in immunity impact RA is poorly understood. Gut microbiota are known to be involved in the pathogenesis of RA, but the effects of microbiota in older subjects remain mostly unknown. METHODS: We used segmented filamentous bacteria (SFB), a gut commensal species with immunomodulatory effects, and K/BxN mice, a T cell receptor (TCR) transgenic model, to study the effect of age and microbiota on autoimmune arthritis. Comparing young and middle-aged K/BxN T cells of the same TCR specificity allows us to study T cells with an age focus eliminating a key variable: TCR repertoire alteration with age. In addition to joints, we also studied pathological changes in the lung, an important extra-articular RA manifestation. We used flow cytometry to evaluate T follicular helper (Tfh) and T helper 17 (Th17) cells, as they both contribute to autoantibody production, a key disease index in both RA and K/BxN arthritis. RESULTS: Middle-aged K/BxN mice had aggravated arthritis and pathological changes in the lung compared to young mice. Middle-aged mice displayed a strong accumulation of Tfh but not Th17 cells, and had defective Th17 differentiation and low expression of interleukin-23, a critical cytokine for Th17 maintenance. Although a soaring Tfh cell population accompanied by robust germinal center B cell responses were found in middle-aged mice, there was decreased cycling of Tfh cells, and SFB only induced the non-Tfh cells to upregulate Bcl-6, the Tfh master transcription factor, in the young but not the middle-aged group. Finally, the accumulated Tfh cells in middle-aged mice had an effector phenotype (CD62LloCD44hi). CONCLUSION: Age-dependent Tfh cell accumulation may play a crucial role in the increased autoimmune disease phenotype in middle-age. SFB, a potent stimulus for inducing Tfh differentiation, fails to promote Tfh differentiation in middle-aged K/BxN mice, suggesting that most of the middle-aged Tfh cells with an effector phenotype are Tfh effector memory cells induced at an earlier age. Our results also indicate that exposure to immunomodulatory commensals may allow the young host to develop an overactive immune system reminiscent of that found in the middle-aged host.


Asunto(s)
Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Microbioma Gastrointestinal/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Células Th17/inmunología , Factores de Edad , Animales , Artritis Reumatoide/genética , Enfermedades Autoinmunes/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/genética , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Células Th17/metabolismo
20.
Sci Transl Med ; 9(376)2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28179509

RESUMEN

Peripheral spondyloarthritis (SpA) is a common extraintestinal manifestation in patients with active inflammatory bowel disease (IBD) characterized by inflammatory enthesitis, dactylitis, or synovitis of nonaxial joints. However, a mechanistic understanding of the link between intestinal inflammation and SpA has yet to emerge. We evaluated and functionally characterized the fecal microbiome of IBD patients with or without peripheral SpA. Coupling the sorting of immunoglobulin A (IgA)-coated microbiota with 16S ribosomal RNA-based analysis (IgA-seq) revealed a selective enrichment in IgA-coated Escherichia coli in patients with Crohn's disease-associated SpA (CD-SpA) compared to CD alone. E. coli isolates from CD-SpA-derived IgA-coated bacteria were similar in genotype and phenotype to an adherent-invasive E. coli (AIEC) pathotype. In comparison to non-AIEC E. coli, colonization of germ-free mice with CD-SpA E. coli isolates induced T helper 17 cell (TH17) mucosal immunity, which required the virulence-associated metabolic enzyme propanediol dehydratase (pduC). Modeling the increase in mucosal and systemic TH17 immunity we observed in CD-SpA patients, colonization of interleukin-10-deficient or K/BxN mice with CD-SpA-derived E. coli lead to more severe colitis or inflammatory arthritis, respectively. Collectively, these data reveal the power of IgA-seq to identify immunoreactive resident pathosymbionts that link mucosal and systemic TH17-dependent inflammation and offer microbial and immunophenotype stratification of CD-SpA that may guide medical and biologic therapy.


Asunto(s)
Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Escherichia coli/metabolismo , Inmunoglobulina A/metabolismo , Inflamación/patología , Espondiloartritis/inmunología , Espondiloartritis/microbiología , Células Th17/inmunología , Animales , Biomarcadores/metabolismo , Colitis/inducido químicamente , Colitis/inmunología , Colitis/microbiología , Enfermedad de Crohn/complicaciones , Sulfato de Dextran , Epitelio/inmunología , Escherichia coli/aislamiento & purificación , Humanos , Inmunidad Mucosa , Inmunofenotipificación , Inflamación/complicaciones , Interleucina-10/metabolismo , Interleucina-23/metabolismo , Intestinos/microbiología , Articulaciones/patología , Ratones Endogámicos C57BL , Espondiloartritis/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...