Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
2.
Diabetes ; 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39167681

RESUMO

Leptin is a homeostatic regulatory element that signals the presence of adipocyte energy stores, reduces food intake, and increases energy expenditure. Similarly, serotonin (5- HT), a signaling molecule found in both the central and peripheral nervous systems, also controls food intake. Using neuronal tract-tracing, pharmacological and optogenetics approaches, and in vivo microdialysis, combined with behavioral endpoints, we tested the hypothesis that leptin controls food intake not only by activating hypothalamic leptin receptors (LepRs), but also through activation of LepRs expressed by serotonergic raphe neurons that send projections to the arcuate (ARC). We show that microinjection of leptin directly into the dorsal raphe nucleus (DRN) reduces food intake in rats. This effect is mediated by LepR expressing neurons in the DRN as selective optogenetic activation of these neurons at either their DRN cell bodies or their ARC terminals reduces food intake. Anatomically, we identified a unique population of serotonergic raphe neurons expressing LepRs that send projections to the ARC. Finally, by utilizing in vivo microdialysis, we show that leptin administration to the DRN increases 5-HT efflux into the ARC, and specific antagonism of the 5-HT2C receptors in the ARC diminishes the leptin anorectic effect. Overall, this study identifies a novel circuit for leptin-mediated control of food intake through a DRN-ARC pathway, identifying a new level of interaction between leptin and serotonin to control food intake. Characterization of this new pathway creates opportunities for understanding how the brain controls eating behavior, as well as opens alternative routes for the treatment of eating disorders.

3.
Hum Pathol ; 152: 105639, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39151736

RESUMO

We present a series of 9 follicular lymphomas that progressed/transformed into classical Hodgkin lymphoma (CHL). Three cases of CHL showed a syncytial pattern (SCHL) making the differential diagnosis to Gray zone lymphoma (GZL) challenging. None of these three cases presented in the mediastinum. Based in all molecular data analyzed (BCL2/BCL6 FISH studies, IgH PCR and TNGS with a customized gene panel) we did find clonal relationship between the BCL2-positive FL cases and their CHL components in all cases. The three SCHL/GZL cases showed an activated phenotype according to Hans algorithm, presented the t(14; 18)(q32; q21), two out of three showed B cell markers and all expressed CD30 and p53. Interestingly, we identified three BCL2-negative FL cases with a further diagnosis of CHL expanding the spectrum of these association. In one of these three cases a different mutational profile was found in both the FL and the CHL components. All this data together suggests that CHL associated to BCL2-positive FL could be originated in a common progenitor cell (CPC) that give rise to both FL and CHL, acquiring this last component further genetic events in a linear fashion. On the other hand, no clonal relationship between CHL and BCL2-negative FL could be found, suggesting a fortuity association. Nevertheless, ample series of cases studied with more sensitive techniques are needed to confirm our hypothesis.


Assuntos
Biomarcadores Tumorais , Doença de Hodgkin , Linfoma Folicular , Proteínas Proto-Oncogênicas c-bcl-2 , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Linfoma Folicular/patologia , Linfoma Folicular/genética , Linfoma Folicular/diagnóstico , Doença de Hodgkin/patologia , Doença de Hodgkin/genética , Doença de Hodgkin/metabolismo , Doença de Hodgkin/diagnóstico , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Adulto , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Hibridização in Situ Fluorescente , Diagnóstico Diferencial , Mutação , Idoso de 80 Anos ou mais
4.
Nat Immunol ; 25(7): 1231-1244, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38898157

RESUMO

To understand the role of T cells in the pathogenesis of ulcerative colitis (UC), we analyzed colonic T cells isolated from patients with UC and controls. Here we identified colonic CD4+ and CD8+ T lymphocyte subsets with gene expression profiles resembling stem-like progenitors, previously reported in several mouse models of autoimmune disease. Stem-like T cells were increased in inflamed areas compared to non-inflamed regions from the same patients. Furthermore, TCR sequence analysis indicated stem-like T cells were clonally related to proinflammatory T cells, suggesting their involvement in sustaining effectors that drive inflammation. Using an adoptive transfer colitis model in mice, we demonstrated that CD4+ T cells deficient in either BCL-6 or TCF1, transcription factors that promote T cell stemness, had decreased colon T cells and diminished pathogenicity. Our results establish a strong association between stem-like T cell populations and UC pathogenesis, highlighting the potential of targeting this population to improve clinical outcomes.


Assuntos
Colite Ulcerativa , Fator 1-alfa Nuclear de Hepatócito , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Humanos , Animais , Camundongos , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 1-alfa Nuclear de Hepatócito/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/genética , Células-Tronco/imunologia , Células-Tronco/metabolismo , Feminino , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Camundongos Knockout , Colo/imunologia , Colo/patologia , Masculino , Camundongos Endogâmicos C57BL , Transferência Adotiva , Modelos Animais de Doenças , Adulto , Pessoa de Meia-Idade
5.
Medicina (Kaunas) ; 59(9)2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37763746

RESUMO

Anaplastic large cell lymphoma (ALCL) with leukaemic presentation (either ab initio or along the course of the disease) has been rarely reported. Irrespective of ALK expression in the neoplastic cells, it features a dismal prognosis. We report a rare case of leukaemic, small cell variant ALK-positive ALCL with 9-year survival in a young woman who was treated upfront with corticosteroids and standard chemotherapy, and review thoroughly the previously published cases. Such an unexpected, good outcome hints at the existence of different clinical subgroups in the leukaemic variant of ALK-positive ALCL.


Assuntos
Linfoma Anaplásico de Células Grandes , Feminino , Humanos , Linfoma Anaplásico de Células Grandes/diagnóstico , Linfoma Anaplásico de Células Grandes/tratamento farmacológico , Linfoma Anaplásico de Células Grandes/metabolismo , Quinase do Linfoma Anaplásico , Receptores Proteína Tirosina Quinases/uso terapêutico , Prognóstico
6.
Front Immunol ; 14: 1095132, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36875069

RESUMO

Introduction: Inflammation is a common unifying factor in experimental models of non-alcoholic fatty liver disease (NAFLD) progression. Recent evidence suggests that housing temperature-driven alterations in hepatic inflammation correlate with exacerbated hepatic steatosis, development of hepatic fibrosis, and hepatocellular damage in a model of high fat diet-driven NAFLD. However, the congruency of these findings across other, frequently employed, experimental mouse models of NAFLD has not been studied. Methods: Here, we examine the impact of housing temperature on steatosis, hepatocellular damage, hepatic inflammation, and fibrosis in NASH diet, methionine and choline deficient diet, and western diet + carbon tetrachloride experimental models of NAFLD in C57BL/6 mice. Results: We show that differences relevant to NAFLD pathology uncovered by thermoneutral housing include: (i) augmented NASH diet-driven hepatic immune cell accrual, exacerbated serum alanine transaminase levels and increased liver tissue damage as determined by NAFLD activity score; (ii) augmented methionine choline deficient diet-driven hepatic immune cell accrual and increased liver tissue damage as indicated by amplified hepatocellular ballooning, lobular inflammation, fibrosis and overall NAFLD activity score; and (iii) dampened western diet + carbon tetrachloride driven hepatic immune cell accrual and serum alanine aminotransferase levels but similar NAFLD activity score. Discussion: Collectively, our findings demonstrate that thermoneutral housing has broad but divergent effects on hepatic immune cell inflammation and hepatocellular damage across existing experimental NAFLD models in mice. These insights may serve as a foundation for future mechanistic interrogations focused on immune cell function in shaping NAFLD progression.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Camundongos Endogâmicos C57BL , Tetracloreto de Carbono , Habitação , Cirrose Hepática , Metionina , Alanina Transaminase , Colina , Modelos Animais de Doenças , Inflamação
7.
Disabil Rehabil ; 45(1): 9-26, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35068313

RESUMO

PURPOSE: To review the evidence around self-management interventions used to improve mobility post-stroke. MATERIALS AND METHODS: An integrative review was carried out. Eight databases were searched from 1992 to July 2021 using keywords based on the PICOS strategy. Two reviewers independently screened and extracted the relevant data. Quality of studies was assessed and a quantitatively led narrative synthesis of data, supported by qualitative evidence, was then conducted. RESULTS: Twenty-four studies with 823 participants were reviewed. Self-management strategies such as patient education, providing information, goal setting, problem-solving, action planning, self-monitoring, and social support were integrated with rehabilitation therapy to improve mobility post-stroke. The reviewed studies showed improvements in functional mobility and walking ability, self-efficacy, participation in physical activity, and quality of life to various extents. Participants in qualitative studies considered the self-management interventions as a valuable addition to their therapy and perceived the improvement in their mobility following them. CONCLUSION: There is some evidence that self-management interventions help to improve mobility outcomes post-stroke. Heterogeneity of data in the studies made meta-analysis impossible. Most of the identified studies examined the feasibility and fidelity of the interventions and further research is warranted to examine the efficacy of these interventions to improve functional mobility post-stroke.Implications for rehabilitationSelf-management interventions can improve mobility-related outcomes, which are considered a priority goal for many stroke survivors.Survivors valued their participation in self-management integrated care programmes and linked that to the perceived improvement in their rehabilitation outcomes.Self-management interventions such as patient education, goal-setting, self-monitoring, and professionals/carers support have been found to improve mobility outcomes for stroke survivors.The outcomes that benefited to a slight extent using self-management were; functional mobility, walking ability (speed, distance, and endurance), and a number of steps per day.


Assuntos
Autogestão , Reabilitação do Acidente Vascular Cerebral , Acidente Vascular Cerebral , Humanos , Qualidade de Vida , Caminhada
8.
Front Immunol ; 13: 1034648, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389671

RESUMO

Colitis is characterized by an exacerbated intestinal immune response, but the genetic and other mechanisms regulating immune activation remain incompletely understood. In order to identify new pathways leading to colitis, we sought to identify genes with increased expression in the colons of patients that also are near loci identified by genome wide association studies (GWAS) associated with IBD risk. One such SNP, rs9557195 was of particular interest because it is within an intron of G-protein-coupled receptor (GPR) 183, known to be important for lymphocyte migration. Furthermore, this SNP is in close proximity to the gene encoding another G-protein coupled receptor, GPR18. Analyzing publicly available datasets, we found transcripts of GPR183 and GPR18 to be increased in colon biopsies from ulcerative colitis and Crohn's disease patients, and GPR183 was even more increased in patients resistant to TNF treatment. Expression of both genes also was increased in mouse models of colitis. Therefore, our aim was to understand if increased expression of these GPRs in the intestine is related to disease severity in colitis models. Here we investigated the role of these receptors in the T cell transfer model and the dextran sulfate sodium model. In the T cell transfer model, GPR183 expression on donor T cells, as well as on other cell types in the Rag-/- recipients, was not essential for severe colitis induction. Furthermore, deficiency in Rag-/- mice for the enzyme that synthesizes a cholesterol metabolite that is a major ligand for GPR183 also did not affect disease. Similarly, lack of GPR18 expression in T cells or other cell types did not affect colitis pathogenesis in the T cell transfer or in the dextran sulfate sodium model. Therefore, despite increased expression of transcripts for these genes in the intestine during inflammation in humans and mice, they are not required for disease severity in mouse models of colitis induced by chemical injury or T cell cytokines, perhaps due to redundancy in mechanisms important for homing and survival of lymphocytes to the inflamed intestine.


Assuntos
Colite , Estudo de Associação Genômica Ampla , Camundongos , Humanos , Animais , Sulfato de Dextrana/efeitos adversos , Camundongos Endogâmicos C57BL , Colite/induzido quimicamente , Colite/genética , Modelos Animais de Doenças , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Linfócitos T CD4-Positivos/metabolismo
9.
Cell Metab ; 33(6): 1187-1204.e9, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34004162

RESUMO

Emerging evidence suggests a key contribution to non-alcoholic fatty liver disease (NAFLD) pathogenesis by Th17 cells. The pathogenic characteristics and mechanisms of hepatic Th17 cells, however, remain unknown. Here, we uncover and characterize a distinct population of inflammatory hepatic CXCR3+Th17 (ihTh17) cells sufficient to exacerbate NAFLD pathogenesis. Hepatic ihTh17 cell accrual was dependent on the liver microenvironment and CXCR3 axis activation. Mechanistically, the pathogenic potential of ihTh17 cells correlated with increased chromatin accessibility, glycolytic output, and concomitant production of IL-17A, IFNγ, and TNFα. Modulation of glycolysis using 2-DG or cell-specific PKM2 deletion was sufficient to reverse ihTh17-centric inflammatory vigor and NAFLD severity. Importantly, ihTh17 cell characteristics, CXCR3 axis activation, and hepatic expression of glycolytic genes were conserved in human NAFLD. Together, our data show that the steatotic liver microenvironment regulates Th17 cell accrual, metabolism, and competence toward an ihTh17 fate. Modulation of these pathways holds potential for development of novel therapeutic strategies for NAFLD.


Assuntos
Proteínas de Transporte/imunologia , Proteínas de Membrana/imunologia , Hepatopatia Gordurosa não Alcoólica/imunologia , Piruvato Quinase/imunologia , Receptores CXCR3/imunologia , Células Th17/imunologia , Hormônios Tireóideos/imunologia , Animais , Linhagem Celular , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th17/citologia , Proteínas de Ligação a Hormônio da Tireoide
10.
Mucosal Immunol ; 14(3): 679-690, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33568785

RESUMO

Inflammatory bowel disease is characterized by an exacerbated intestinal immune response, but the critical mechanisms regulating immune activation remain incompletely understood. We previously reported that the TNF-superfamily molecule TNFSF14 (LIGHT) is required for preventing severe disease in mouse models of colitis. In addition, deletion of lymphotoxin beta receptor (LTßR), which binds LIGHT, also led to aggravated colitis pathogenesis. Here, we aimed to determine the cell type(s) requiring LTßR and the mechanism critical for exacerbation of colitis. Specific deletion of LTßR in neutrophils (LTßRΔN), but not in several other cell types, was sufficient to induce aggravated colitis and colonic neutrophil accumulation. Mechanistically, RNA-Seq analysis revealed LIGHT-induced suppression of cellular metabolism, and mitochondrial function, that was dependent on LTßR. Functional studies confirmed increased mitochondrial mass and activity, associated with excessive mitochondrial ROS production and elevated glycolysis at steady-state and during colitis. Targeting these metabolic changes rescued exacerbated disease severity. Our results demonstrate that LIGHT signals to LTßR on neutrophils to suppress metabolic activation and thereby prevents exacerbated immune pathogenesis during colitis.


Assuntos
Colite/imunologia , Doenças Inflamatórias Intestinais/imunologia , Receptor beta de Linfotoxina/metabolismo , Mitocôndrias/metabolismo , Neutrófilos/metabolismo , Ativação Metabólica , Animais , Sulfato de Dextrana , Modelos Animais de Doenças , Progressão da Doença , Humanos , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
11.
Methods Mol Biol ; 2121: 129-140, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32147792

RESUMO

Innate lymphoid cells (ILCs) are important regulators of the early responses to infection at mucosal barriers, including the intestine. Recently, we have shown that specific ILC3 subsets protect against enteric bacterial pathogens. Here, we describe a mouse model of oral infection by Yersinia enterocolitica (Y. enterocolitica) and several different methodologies to assess the severity of the infection. We also detail how ILC3 subsets can be isolated from the mouse small intestine and transferred into recipient immune deficient mice to study the function of these ILCs in the small intestine.


Assuntos
Transferência Adotiva/métodos , Imunidade Inata , Intestino Delgado/imunologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Mucosa/imunologia , Yersiniose/imunologia , Animais , Proteínas de Homeodomínio/genética , Intestino Delgado/citologia , Intestino Delgado/microbiologia , Camundongos , Camundongos Knockout , Mucosa/citologia
12.
Mucosal Immunol ; 13(3): 399-412, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32047273

RESUMO

Innate lymphoid cells (ILCs) are a lymphocyte population that is mostly resident at mucosal surfaces. They help to induce an appropriate immune response to the microbiome at homeostasis. In healthy people, the mucosal immune system works symbiotically with organisms that make up the microbiota. ILCs play a critical role in orchestrating this balance, as they can both influence and in turn be influenced by the microbiome. ILCs also are important regulators of the early response to infections by diverse types of pathogenic microbes at mucosal barriers. Their rapid responses initiate inflammatory programs, production of antimicrobial products and repair processes. This review will focus on the role of ILCs in response to the microbiota and to microbial infections of the lung and intestine.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Microbiota/imunologia , Mucosa/imunologia , Mucosa/microbiologia , Animais , Biomarcadores , Citocinas/metabolismo , Homeostase , Interações Hospedeiro-Parasita/imunologia , Humanos
13.
Front Pharmacol ; 10: 212, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30930775

RESUMO

The inflammatory Bowel diseases (IBDs) are a chronic, relapsing inflammatory diseases of the gastrointestinal tract with heterogeneous behavior and prognosis. The introduction of biological therapies including anti-TNF, anti-IL-12/23, and anti-integrins, has revolutionized the treatment of IBD, but these drugs are not universally effective. Due to the complex molecular structures of biologics, they are uniformly immunogenic. New discoveries concerning the underlying mechanisms involved in the pathogenesis of IBD have allowed for progress in the development of new treatment options. The advantage of small molecules (SMs) over biological therapies includes their lack of immunogenicity, short half-life, oral administration, and low manufacturing cost. Among these, the Janus Kinases (JAKs) inhibition has emerged as a novel strategy to modulate downstream cytokine signaling during immune-mediated diseases. These drugs target various cytokine signaling pathways that participate in the pathogenesis of IBD. Tofacitinib, a JAK inhibitor targeting predominantly JAK1 and JAK3, has been approved for the treatment of ulcerative colitis (UC), and there are other specific JAK inhibitors under development that may be effective in Crohn's. Similarly, the traffic of lymphocytes can now be targeted by another SM. Sphingosine-1-phosphate receptor (S1PR) agonism is a novel strategy that acts, in part, by interfering with lymphocyte recirculation, through blockade of lymphocyte egress from lymph nodes. S1PR agonists are being studied in IBD and other immune-mediated disorders. This review will focus on SM drugs approved and under development, including JAK inhibitors (tofacitinib, filgotinib, upadacitinib, peficitinib) and S1PR agonists (KRP-203, fingolimod, ozanimod, etrasimod, amiselimod), and their mechanism of action.

14.
Front Immunol ; 9: 2585, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524422

RESUMO

Over 1.5 million individuals in the United States are afflicted with inflammatory bowel disease (IBD). While the progression of IBD is multifactorial, chronic, unresolved inflammation certainly plays a key role. Additionally, while multiple immune mediators have been shown to affect pathogenesis, a comprehensive understanding of disease progression is lacking. Previous work has demonstrated that a member of the TNF superfamily, TNFSF14 (LIGHT), which is pro-inflammatory in several contexts, surprisingly plays an important role in protection from inflammation in mouse models of colitis, with LIGHT deficient mice having more severe disease pathogenesis. However, LIGHT is a single member of a complex signaling network. It signals through multiple receptors, including herpes virus entry mediator (HVEM) and lymphotoxin beta receptor (LTßR); these two receptors in turn can bind to other ligands. It remains unknown which receptors and competing ligands can mediate or counteract the outcome of LIGHT-signaling during colitis. Here we demonstrate that LIGHT signaling through LTßR, rather than HVEM, plays a critical role in the progression of DSS-induced colitis, as LTßR deficient mice exhibit a more severe disease phenotype. Further, mice deficient in LTαß do not exhibit differential colitis progression compared to WT mice. However, deletion of both LIGHT and LTαß, but not deletion of both LTαß and LTßR, resulted in a reversal of the adverse effects associated with the loss of LIGHT. In sum, the LIGHT/LTαß/LTßR signaling network contributes to DSS colitis, but there may be additional receptors or indirect effects, and therefore, the relationships between these receptors and ligands remains enigmatic.


Assuntos
Colite/imunologia , Doenças Inflamatórias Intestinais/imunologia , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-beta/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Colite/induzido quimicamente , Sulfato de Dextrana , Modelos Animais de Doenças , Progressão da Doença , Humanos , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
Cell Host Microbe ; 24(2): 249-260.e4, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30092201

RESUMO

Innate lymphoid cells (ILCs) are important regulators of early infection at mucosal barriers. ILCs are divided into three groups based on expression profiles, and are activated by cytokines and neuropeptides. Yet, it remains unknown if ILCs integrate other signals in providing protection. We show that signaling through herpes virus entry mediator (HVEM), a member of the tumor necrosis factor (TNF) receptor superfamily, in ILC3 is important for host defense against oral infection with the bacterial pathogen Yersinia enterocolitica. HVEM stimulates protective interferon-γ (IFN-γ) secretion from ILCs, and mice with HVEM-deficient ILC3 exhibit reduced IFN-γ production, higher bacterial burdens and increased mortality. In addition, IFN-γ production is critical as adoptive transfer of wild-type but not IFN-γ-deficient ILC3 can restore protection to mice lacking ILCs. We identify the TNF superfamily member, LIGHT, as the ligand inducing HVEM signals in ILCs. Thus HVEM signaling mediated by LIGHT plays a critical role in regulating ILC3-derived IFN-γ production for protection following infection. VIDEO ABSTRACT.


Assuntos
Infecções por Enterobacteriaceae/prevenção & controle , Interferon gama/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Transferência Adotiva , Adulto , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/metabolismo , Transporte Proteico , Receptores CCR6/genética , Receptores CCR6/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Baço/microbiologia , Baço/patologia , Yersinia enterocolitica/patogenicidade
16.
Hepatol Commun ; 2(5): 546-560, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29761170

RESUMO

Nonalcoholic fatty liver disease (NAFLD) represents a disease spectrum ranging from benign steatosis to life-threatening cirrhosis and hepatocellular carcinoma. Elevated levels of reactive oxygen species (ROS) and exacerbated inflammatory responses have been implicated in NAFLD progression. Nicotinamide adenine dinucleotide phosphate (reduced) oxidase 2 (NOX2; also known as gp91Phox), the main catalytic subunit of the nicotinamide adenine dinucleotide phosphate (reduced) oxidase complex, modulates ROS production, immune responsiveness, and pathogenesis of obesity-associated metabolic derangements. However, the role of NOX2 in the regulation of immune cell function and inflammatory vigor in NAFLD remains underdefined. Here, we demonstrate that obesogenic diet feeding promoted ROS production by bone marrow, white adipose tissue, and liver immune cells. Genetic ablation of NOX2 impeded immune cell ROS synthesis and was sufficient to uncouple obesity from glucose dysmetabolism and NAFLD pathogenesis. Protection from hepatocellular damage in NOX2-deficient mice correlated with reduced hepatic neutrophil, macrophage, and T-cell infiltration, diminished production of key NAFLD-driving proinflammatory cytokines, and an inherent reduction in T-cell polarization toward Th17 phenotype. Conclusion: Current findings demonstrate a crucial role of the NOX2-ROS axis in immune cell effector function and polarization and consequent NAFLD progression in obesity. Pharmacologic targeting of NOX2 function in immune cells may represent a viable approach for reducing morbidity of obesity-associated NAFLD pathogenesis. (Hepatology Communications 2018;2:546-560).

17.
JCI Insight ; 3(6)2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29563328

RESUMO

Nonalcoholic fatty liver disease (NAFLD), a metabolic predisposition for development of hepatocellular carcinoma (HCC), represents a disease spectrum ranging from steatosis to steatohepatitis to cirrhosis. Acox1, a rate-limiting enzyme in peroxisomal fatty acid ß-oxidation, regulates metabolism, spontaneous hepatic steatosis, and hepatocellular damage over time. However, it is unknown whether Acox1 modulates inflammation relevant to NAFLD pathogenesis or if Acox1-associated metabolic and inflammatory derangements uncover and accelerate potential for NAFLD progression. Here, we show that mice with a point mutation in Acox1 (Acox1Lampe1) exhibited altered cellular metabolism, modified T cell polarization, and exacerbated immune cell inflammatory potential. Further, in context of a brief obesogenic diet stress, NAFLD progression associated with Acox1 mutation resulted in significantly accelerated and exacerbated hepatocellular damage via induction of profound histological changes in hepatocytes, hepatic inflammation, and robust upregulation of gene expression associated with HCC development. Collectively, these data demonstrate that ß-oxidation links metabolism and immune responsiveness and that a better understanding of peroxisomal ß-oxidation may allow for discovery of mechanisms central for NAFLD progression.


Assuntos
Acil-CoA Oxidase/metabolismo , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Acil-CoA Oxidase/genética , Tecido Adiposo Marrom/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Citocinas/metabolismo , Dieta , Modelos Animais de Doenças , Progressão da Doença , Ácidos Graxos/metabolismo , Regulação Neoplásica da Expressão Gênica , Hepatócitos/patologia , Inflamação , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática , Neoplasias Hepáticas/genética , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade , Mutação Puntual , Estresse Fisiológico , Linfócitos T
19.
Nat Med ; 23(7): 829-838, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28604704

RESUMO

Nonalcoholic fatty liver disease (NAFLD), a common prelude to cirrhosis and hepatocellular carcinoma, is the most common chronic liver disease worldwide. Defining the molecular mechanisms underlying the pathogenesis of NAFLD has been hampered by a lack of animal models that closely recapitulate the severe end of the disease spectrum in humans, including bridging hepatic fibrosis. Here we demonstrate that a novel experimental model employing thermoneutral housing, as opposed to standard housing, resulted in lower stress-driven production of corticosterone, augmented mouse proinflammatory immune responses and markedly exacerbated high-fat diet (HFD)-induced NAFLD pathogenesis. Disease exacerbation at thermoneutrality was conserved across multiple mouse strains and was associated with augmented intestinal permeability, an altered microbiome and activation of inflammatory pathways that are associated with the disease in humans. Depletion of Gram-negative microbiota, hematopoietic cell deletion of Toll-like receptor 4 (TLR4) and inactivation of the IL-17 axis resulted in altered immune responsiveness and protection from thermoneutral-housing-driven NAFLD amplification. Finally, female mice, typically resistant to HFD-induced obesity and NAFLD, develop full disease characteristics at thermoneutrality. Thus, thermoneutral housing provides a sex-independent model of exacerbated NAFLD in mice and represents a novel approach for interrogation of the cellular and molecular mechanisms underlying disease pathogenesis.


Assuntos
Dieta Hiperlipídica , Abrigo para Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo , Receptores de Interleucina-17/imunologia , Estresse Fisiológico/imunologia , Temperatura , Receptor 4 Toll-Like/metabolismo , Animais , Temperatura Baixa , Corticosterona/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Citometria de Fluxo , Microbioma Gastrointestinal/imunologia , Perfilação da Expressão Gênica , Bactérias Gram-Negativas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Inflamação , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Aprendizado de Máquina , Masculino , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/imunologia , Obesidade/imunologia , Permeabilidade , Receptores de Interleucina-17/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Receptor 4 Toll-Like/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA