Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Obes (Lond) ; 48(7): 1036-1038, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38467728

RESUMO

Although the orchestrating role of Interleukin-36 cytokines in regulating inflammation at barrier tissue sites, is well established, whether they play a significant role in the settings of metabolic health and disease, has yet to be fully established. Several recent studies have demonstrated that IL-36 cytokine expression is elevated among adult patients with obesity, and can play roles in regulating both insulin sensitivity and driving inflammation. In this report, we have extended these analyses to paediatric patients and identified an association between elevated serum levels of expression of the specific Interleukin-36 subfamily member, IL-36ß, among children with obesity displaying insulin sensitivity, compared to children with obesity who are insulin resistant. While these data further indicate a possible protective role for IL-36 in metabolic health, they also differ with previous findings from an adult patient cohort, where elevated levels of the related cytokine, IL-36γ, were found to occur in association with improved metabolic health. While highlighting important differences between paediatric and adult patient cohorts in the context of metabolic disease associated with obesity, these data underscore the need for a deeper mechanistic analysis of the role of IL-36 cytokines in disease.


Assuntos
Resistência à Insulina , Interleucina-1 , Obesidade Infantil , Humanos , Resistência à Insulina/fisiologia , Criança , Masculino , Feminino , Interleucina-1/sangue , Obesidade Infantil/sangue , Obesidade Infantil/complicações , Adolescente , Inflamação/sangue
2.
J Immunol ; 207(2): 651-660, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34253575

RESUMO

SIGIRR has been described as a negative regulator of several IL-1R/TLR family members and has been implicated in several inflammatory disease conditions. However, it is unknown whether it can suppress IL-36 family cytokines, which are members of the broader IL-1 superfamily that have emerged as critical orchestrators of psoriatic inflammation in both humans and mice. In this study, we demonstrate that SIGIRR is downregulated in psoriatic lesions in humans and mice, and this correlates with increased expression of IL-36 family cytokines. Using Sigirr -/- mice, we identify, for the first time (to our knowledge), SIGIRR as a negative regulator of IL-36 responses in the skin. Mechanistically, we identify dendritic cells and keratinocytes as the primary cell subsets in which IL-36 proinflammatory responses are regulated by SIGIRR. Both cell types displayed elevated IL-36 responsiveness in absence of SIGIRR activity, characterized by enhanced expression of neutrophil chemoattractants, leading to increased neutrophil infiltration to the inflamed skin. Blockade of IL-36R signaling ameliorated exacerbated psoriasiform inflammation in Sigirr -/- mice and inhibited neutrophil infiltration. These data identify SIGIRR activity as an important regulatory node in suppressing IL-36-dependent psoriatic inflammation in humans and mice.


Assuntos
Inflamação/metabolismo , Interleucina-1/metabolismo , Infiltração de Neutrófilos/fisiologia , Receptores de Interleucina-1/metabolismo , Pele/metabolismo , Animais , Citocinas/metabolismo , Regulação para Baixo/fisiologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Psoríase/metabolismo , Transdução de Sinais/fisiologia
3.
Cytokine ; 154: 155890, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35462264

RESUMO

The interleukin-1 (IL-1) family of cytokines and receptors are implicated in the functioning of innate and adaptive immunity and the genesis of inflammation. They are widely expressed in structural and immune cells with marked expression within barrier mucosal surfaces. In the lung, gut and skin, which are common entry sites for pathogens, they play essential functions in maintaining the functional integrity of the barrier and manage innate and adaptive immunity in response to insult and infections. In tissue sites, the IL-1 cytokines are tightly regulated by mechanisms involving decoy receptors and protease degradation. Dysregulation of these processes are associated with aberrant tissue inflammation leading to a number of inflammatory diseases. This review will address the roles of the different IL-1 cytokines at the lung, gut and skin barrier surfaces at homeostasis, and their roles as inflammatory mediators in diseases such as asthma, chronic obstructive pulmonary disease, inflammatory bowel diseases, atopic dermatitis and psoriasis.


Assuntos
Citocinas , Doenças Inflamatórias Intestinais , Imunidade Adaptativa , Humanos , Imunidade Inata , Inflamação , Interleucina-1
4.
Eur J Immunol ; 49(9): 1306-1320, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31250428

RESUMO

Since the first description of interleukin-1 (IL-1) and the genesis of the field of cytokine biology, the understanding of how IL-1 and related cytokines play central orchestrating roles in the inflammatory response has been an area of intense investigation. As a consequence of these endeavours, specific strategies have been developed to target the function of the IL-1 family in human disease realizing significant impacts for patients. While the most significant advances to date have been associated with inhibition of the prototypical family members IL-1α/ß, approaches to target more recently identified family members such as IL-18, IL-33 and the IL-36 subfamily are now beginning to come to fruition. This review summarizes current knowledge surrounding the roles of the IL-1 family in human disease and describes the rationale and strategies which have been developed to target these cytokines to inhibit the pathogenesis of a wide range of diseases in which inflammation plays a centrally important role.


Assuntos
Inflamação/metabolismo , Interleucina-1/metabolismo , Animais , Citocinas/metabolismo , Humanos
5.
Allergy ; 74(10): 1920-1933, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30937919

RESUMO

BACKGROUND: Atopic dermatitis (AD) is one of the most common skin diseases with a multifactorial etiology. Mutations leading to loss of skin barrier function are associated with the development of AD with group 2 innate lymphoid cells (ILC2) promoting acute skin inflammation. Filaggrin-mutant (Flgft/ft ) mice develop spontaneous skin inflammation accompanied by an increase in skin ILC2 numbers, IL-1ß production, and other cytokines recapitulating human AD. Here, we investigated the role of ILC2, effector cytokines, inflammasome activation, and mast cell function on the development of chronic AD-like inflammation in mice. METHODS: Mice with a frameshift mutation in the filaggrin gene develop spontaneous dermatitis. Flgft/ft mice were crossed to cell- or cytokine-deficient mouse strains, or bred under germ-free conditions. Skin inflammation was scored, and microbiome composition was analyzed. Skin protein expression was measured by multiplex immunoassay. Infiltrating cells were analyzed by flow cytometry. RESULTS: Wild-type and Flgft/ft mice significantly differ in their microbiome composition. Furthermore, mutant mice do not develop skin inflammation under germ-free conditions. ILC2 deficiency did not ameliorate chronic dermatitis in Flgft/ft mice, which was also independent of IL-4, IL-5, IL-9, IL-13, IL-17A, and IL-22. Inflammation was independent of NLRP3 inflammasome activation but required IL-1ß and IL-1R1-signaling. Mechanistically, IL-1ß promoted hyperactivation of IL-1R1-expressing mast cells. Treatment with anti-IL-1ß-antibody alleviated dermatitis exacerbation, while antibiotic intervention ameliorated dermatitis in neonatal mice but not in adults with established inflammation. CONCLUSIONS: In summary, we identified a critical role for the microbiome and IL-1ß mediating chronic inflammation in mice with an impaired skin barrier.


Assuntos
Dermatite Atópica/imunologia , Dermatite Atópica/metabolismo , Imunidade Inata , Interleucina-1beta/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Animais , Biópsia , Citocinas/metabolismo , Dermatite Atópica/patologia , Modelos Animais de Doenças , Proteínas Filagrinas , Inflamassomos/metabolismo , Linfócitos/patologia , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Transgênicos , Microbiota , Fenótipo , Transdução de Sinais , Pele/imunologia , Pele/metabolismo , Pele/patologia
6.
J Immunol ; 199(2): 707-717, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28615416

RESUMO

Atopic dermatitis (AD) is a common inflammatory skin disease affecting up to 20% of children and 3% of adults worldwide and is associated with dysregulation of the skin barrier. Although type 2 responses are implicated in AD, emerging evidence indicates a potential role for the IL-17A signaling axis in AD pathogenesis. In this study we show that in the filaggrin mutant mouse model of spontaneous AD, IL-17RA deficiency (Il17ra-/- ) resulted in severe exacerbation of skin inflammation. Interestingly, Il17ra-/- mice without the filaggrin mutation also developed spontaneous progressive skin inflammation with eosinophilia, as well as increased levels of thymic stromal lymphopoietin (TSLP) and IL-5 in the skin. Il17ra-/- mice have a defective skin barrier with altered filaggrin expression. The barrier dysregulation and spontaneous skin inflammation in Il17ra-/- mice was dependent on TSLP, but not the other alarmins IL-25 and IL-33. The associated skin inflammation was mediated by IL-5-expressing pathogenic effector Th2 cells and was independent of TCRγδ T cells and IL-22. An absence of IL-17RA in nonhematopoietic cells, but not in the hematopoietic cells, was required for the development of spontaneous skin inflammation. Skin microbiome dysbiosis developed in the absence of IL-17RA, with antibiotic intervention resulting in significant amelioration of skin inflammation and reductions in skin-infiltrating pathogenic effector Th2 cells and TSLP. This study describes a previously unappreciated protective role for IL-17RA signaling in regulation of the skin barrier and maintenance of skin immune homeostasis.


Assuntos
Dermatite Atópica/imunologia , Receptores de Interleucina-17/imunologia , Receptores de Interleucina-17/metabolismo , Pele/crescimento & desenvolvimento , Pele/patologia , Animais , Citocinas/imunologia , Dermatite Atópica/patologia , Modelos Animais de Doenças , Disbiose , Eosinofilia/imunologia , Proteínas Filagrinas , Regulação da Expressão Gênica , Homeostase , Interleucina-33/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Interleucinas/genética , Interleucinas/imunologia , Proteínas de Filamentos Intermediários/deficiência , Proteínas de Filamentos Intermediários/genética , Camundongos , Microbiota , Mutação , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Interleucina-17/deficiência , Receptores de Interleucina-17/genética , Transdução de Sinais , Pele/imunologia , Pele/microbiologia , Células Th2/imunologia , Linfopoietina do Estroma do Timo , Interleucina 22
7.
Semin Thromb Hemost ; 44(2): 167-175, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29232721

RESUMO

Tissue injury prompts the initiation of host defense responses to limit blood loss, restrict pathogen entry, and promote repair. Biochemical and cellular pathways that lead to blood coagulation serve a fundamental role in generating a physical barrier at the wound site, but have also evolved to promote immune response to injury. Similarly, anticoagulant pathways that attenuate clot formation also regulate innate and adaptive immune responses. Of particular importance is activated protein C (APC) which serves as a principal regulator of thrombin generation, shapes the innate immune response to infection, and has been shown to contribute to the adaptive immune response in several preclinical models of autoimmune disease. APC controls blood coagulation by proteolytic degradation of procoagulant activated cofactors essential for fibrin clot development, but also cleaves multiple additional substrates and interacts with cell surface receptors to exert additional physiologically important roles. In this review, we focus on the molecular mechanisms utilized by APC to limit inflammation and, in particular, current understanding of the basis for APC anticoagulant and signaling activities. In particular, we provide an overview of established and emerging signaling pathways initiated by APC on endothelial cells, monocytes, neutrophils, dendritic cells, and T cells to control and regulate immune cell physiology. Finally, we consider the impact of APC activity in the context of both acute and chronic inflammatory disease, and the continuing efforts to harness the immunoregulatory properties of recombinant APC for therapeutic use.


Assuntos
Inflamação/sangue , Monócitos/metabolismo , Proteína C/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia
8.
J Immunol ; 196(2): 703-14, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26673140

RESUMO

Epidemiologic studies in humans have demonstrated that infection with helminth parasites is associated with a reduced risk of developing autoimmune diseases. Mechanistic studies in mice have linked the protective effect of helminths on autoimmunity to the suppressive activity of helminth-induced regulatory T cells (Tregs) or Th2 cells. In this study, we demonstrate that treatment of mice with Fasciola hepatica excretory-secretory products (FHES) attenuated the clinical signs of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. Protection was associated with a significant reduction in the infiltration of pathogenic Th1 and Th17 cells into the brain. Although FHES enhanced anti-inflammatory cytokine and Th2 responses, protection against EAE was independent of IL-4, IL-10, and Tregs. However, administration of FHES induced production of the type 2 cytokines IL-33 and IL-5, which promoted accumulation of eosinophils. FHES-induced expansion of eosinophils and protection against EAE was lost in IL-33(-/-) mice and upon neutralization of IL-5. Furthermore, transfer of FHES-induced or IL-33-induced eosinophils conferred protection against EAE. In addition, treatment of mice with recombinant IL-33 attenuated autoimmunity, and this was dependent on IL-5. To our knowledge, this study is the first to report a role for helminth-induced IL-5 and IL-33 in protection against autoimmunity.


Assuntos
Antígenos de Helmintos/imunologia , Autoimunidade/imunologia , Eosinofilia/imunologia , Interleucina-33/imunologia , Interleucina-5/imunologia , Células Th2/imunologia , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/microbiologia , Encefalomielite Autoimune Experimental/patologia , Citometria de Fluxo , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
Blood ; 126(7): 915-9, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26084674

RESUMO

Activated protein C (APC) is an anticoagulant protease that initiates cell signaling via protease-activated receptor 1 (PAR1) to regulate vascular integrity and inflammatory response. In this study, a recombinant APC variant (APC(N329Q)) mimicking the naturally occurring APC-ß plasma glycoform was found to exhibit superior PAR1 proteolysis at a cleavage site that selectively mediates cytoprotective signaling. APC(N329Q) also enhanced integrin αMß2-dependent PAR1 proteolysis to exert significantly improved antiinflammatory activity on macrophages compared with wild-type APC. Recent therapeutic applications of recombinant APC in ischemic stroke models have used APC variants with limited anticoagulant activity to negate potential bleeding side effects. Using a mouse model of ischemic stroke and late t-PA intervention, the neuroprotective activity of a murine APC variant with limited anticoagulant activity (mAPC(PS)) was compared with an identical APC variant except for the absence of glycosylation at the APC-ß sequon (mAPC(PS/N329Q)). Remarkably, mAPC(PS/N329Q) limited cerebral ischemic injury and reduced brain lesion volume significantly more effectively than mAPC(PS). Collectively, this study reveals the importance of APC glycosylation in controlling the efficacy of PAR1 proteolysis by APC and demonstrates the potential of novel APC variants with superior cytoprotective signaling function as enhanced therapeutic agents for the treatment of ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , Proteína C/metabolismo , Receptor PAR-1/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Isquemia Encefálica/terapia , Catepsinas/genética , Catepsinas/metabolismo , Modelos Animais de Doenças , Receptor de Proteína C Endotelial , Variação Genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos , Proteína C/genética , Proteína C/uso terapêutico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/uso terapêutico , Proteólise , Receptor PAR-1/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais
11.
Laterality ; 22(6): 641-653, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27666522

RESUMO

There is considerable debate about the pattern and origin of laterality in forelimb emergence and turning behaviour within amphibians, with the latter being poorly investigated in tadpoles around metamorphic climax. Using 6 species of metamorphosing anurans, we investigated the effect of asymmetrical spiracle location, and disturbance at the time of forelimb emergence, on the pattern of forelimb emergence. Turning behaviour was observed to assess whether motor lateralization occurred in non-neobatrachian anurans and was linked to patterns of forelimb emergence. Biases in forelimb emergence differed among species, supporting the hypothesis that asymmetrical spiracle position results in the same asymmetry in forelimb emergence. However, this pattern only occurred when individuals were undisturbed. Therefore, context at the time of the emergence of the forelimbs may be important, and might explain some discrepancies in the literature. Turning biases, unconnected to forelimb emergence, were found in Pipidae and Bombinatoridae, confirming the basal origin of lateralized behaviour among anurans. Turning direction in our metamorphs differed from the leftward bias commonly observed in tadpoles, but may be analogous to the prevalent right-"handedness" among adult anurans. Therefore, the transitions occurring during metamorphosis may affect lateralized behaviour and metamorphosis may be fruitful for understanding the development of lateralization.


Assuntos
Anuros/fisiologia , Membro Anterior , Lateralidade Funcional , Metamorfose Biológica , Atividade Motora , Animais , Anuros/crescimento & desenvolvimento , Comportamento Animal , Reação de Fuga , Membro Anterior/crescimento & desenvolvimento , Membro Anterior/fisiologia , Modelos Lineares , Atividade Motora/fisiologia , Especificidade da Espécie
12.
J Immunol ; 191(6): 3337-46, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23945140

RESUMO

Expression of the orphan receptor Toll IL-1R8/single Ig IL-1-related receptor has been reported to be reduced in the peripheral blood of psoriatic arthritis patients. However whether TIR8/SIGIRR activity plays a specific role in regulating psoriatic inflammation is unknown. We report that Tir8/Sigirr-deficient mice develop more severe psoriatic inflammation in both the chemical (Aldara)- and cytokine (rIL-23)-induced models of psoriasis. Increased disease severity was associated with enhanced infiltration of Vγ4⁺ γδ T cells that express significantly elevated levels of IL-17A. Critically, we also demonstrate that TIR8/SIGIRR activity directly suppressed innate IL-17A expression by γδ T cells in vitro and in vivo. Importantly, treatment of Tir8/Sigirr⁻/⁻ mice with an IL-17A neutralization Ab reversed the enhanced disease severity observed in these mice. This study identifies TIR8/SIGIRR as a novel intrinsic negative regulator of innate IL-17A expression and characterizes a novel mechanism involved in the regulation of psoriatic inflammation.


Assuntos
Artrite Psoriásica/imunologia , Interleucina-17/biossíntese , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Receptores de Interleucina-1/imunologia , Linfócitos T/imunologia , Animais , Artrite Psoriásica/metabolismo , Artrite Psoriásica/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Interleucina-1/metabolismo , Linfócitos T/metabolismo
13.
J Thromb Haemost ; 22(2): 394-409, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37865288

RESUMO

BACKGROUND: Myeloid cell metabolic reprogramming is a hallmark of inflammatory disease; however, its role in inflammation-induced hypercoagulability is poorly understood. OBJECTIVES: We aimed to evaluate the role of inflammation-associated metabolic reprogramming in regulating blood coagulation. METHODS: We used novel myeloid cell-based global hemostasis assays and murine models of immunometabolic disease. RESULTS: Glycolysis was essential for enhanced activated myeloid cell tissue factor expression and decryption, driving increased cell-dependent thrombin generation in response to inflammatory challenge. Similarly, inhibition of glycolysis enhanced activated macrophage fibrinolytic activity through reduced plasminogen activator inhibitor 1 activity. Macrophage polarization or activation markedly increased endothelial protein C receptor (EPCR) expression on monocytes and macrophages, leading to increased myeloid cell-dependent protein C activation. Importantly, inflammation-dependent EPCR expression on tissue-resident macrophages was also observed in vivo. Adipose tissue macrophages from obese mice fed a high-fat diet exhibited significantly enhanced EPCR expression and activated protein C generation compared with macrophages isolated from the adipose tissue of healthy mice. Similarly, the induction of colitis in mice prompted infiltration of EPCR+ innate myeloid cells within inflamed colonic tissue that were absent from the intestinal tissue of healthy mice. CONCLUSION: Collectively, this study identifies immunometabolic regulation of myeloid cell hypercoagulability, opening new therapeutic possibilities for targeted mitigation of thromboinflammatory disease.


Assuntos
Proteína C , Trombofilia , Animais , Camundongos , Proteína C/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Células Mieloides/metabolismo , Inflamação/metabolismo , Trombofilia/etiologia , Glicólise , Camundongos Endogâmicos C57BL
14.
Am J Physiol Lung Cell Mol Physiol ; 304(5): L312-23, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23292810

RESUMO

The role of T cells in chronic obstructive pulmonary disease (COPD) is not well understood. We have previously demonstrated that chronic cigarette smoke exposure can lead to the accumulation of CD4(+) and CD8(+) T cells in the alveolar airspaces in a mouse model of COPD, implicating these cells in disease pathogenesis. However, whether specific inhibition of T cell responses represents a therapeutic strategy has not been fully investigated. In this study inhibition of T cell responses through specific depleting antibodies, or the T cell immunosuppressant drug cyclosporin A, prevented airspace enlargement and neutrophil infiltration in a mouse model of chronic cigarette smoke exposure. Furthermore, individual inhibition of either CD4(+) T helper or CD8(+) T cytotoxic cells prevented airspace enlargement to a similar degree, implicating both T cell subsets as critical mediators of the adaptive immune response induced by cigarette smoke exposure. Importantly, T cell depletion resulted in significantly decreased levels of the Th17-associated cytokine IL-17A, and of caspase 3 and caspase 7 gene expression and activity, induced by cigarette smoke exposure. Finally, inhibition of T cell responses in a therapeutic manner also inhibited cigarette smoke-induced airspace enlargement, IL-17A expression, and neutrophil influx in mice. Together these data demonstrate for the first time that therapeutic inhibition of T cell responses may be efficacious in the treatment of COPD. Given that broad immunosuppression may be undesirable in COPD patients, this study provides proof-of-concept for more targeted approaches to inhibiting the role of T cells in emphysema development.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Alvéolos Pulmonares/patologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Fumar , Animais , Caspase 3/sangue , Caspase 7/biossíntese , Caspase 7/genética , Ciclosporina , Modelos Animais de Doenças , Feminino , Expressão Gênica , Terapia de Imunossupressão , Interleucina-17/sangue , Medidas de Volume Pulmonar , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/imunologia , Alvéolos Pulmonares/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia , Poluição por Fumaça de Tabaco
15.
Nat Med ; 12(9): 1088-92, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16921377

RESUMO

TRAF6 has a key role in the regulation of innate immune responses by mediating signals from both TNF receptor and interleukin-1 receptor/Toll-like receptor superfamilies. Here we show that T cell-specific deletion of TRAF6 unexpectedly results in multiorgan inflammatory disease. TRAF6-deficient T cells exhibit hyperactivation of the phosphatidylinositol 3-kinase (PI3K)-Akt pathway compared with wild-type T cells and, as a result, become resistant to suppression by CD4+ CD25+ regulatory T cells. These data identify a previously unrecognized role for TRAF6 in the maintenance of peripheral tolerance, and suggest the presence of a T cell-intrinsic control mechanism to render responder T cells susceptible to tolerizing signals.


Assuntos
Homeostase/imunologia , Tolerância Imunológica/fisiologia , Inflamação/imunologia , Linfócitos T/fisiologia , Fator 6 Associado a Receptor de TNF/fisiologia , Animais , Antígenos CD4/fisiologia , Subunidade alfa de Receptor de Interleucina-2/fisiologia , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T Reguladores/fisiologia , Fator 6 Associado a Receptor de TNF/deficiência
16.
FEBS J ; 2023 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-37300849

RESUMO

Recent advances in understanding how the microbiome can influence both the physiology and the pathogenesis of disease in humans have highlighted the importance of gaining a deeper insight into the complexities of the host-microbial dialogue. In tandem with this progress, has been a greater understanding of the biological pathways which regulate both homeostasis and inflammation at barrier tissue sites, such as the skin and the gut. In this regard, the Interleukin-1 family of cytokines, which can be segregated into IL-1, IL-18 and IL-36 subfamilies, have emerged as important custodians of barrier health and immunity. With established roles as orchestrators of various inflammatory diseases in both the skin and intestine, it is now becoming clear that IL-1 family cytokine activity is not only directly influenced by external microbes, but can also play important roles in shaping the composition of the microbiome at barrier sites. This review explores the current knowledge surrounding the evidence that places these cytokines as key mediators at the interface between the microbiome and human health and disease at the skin and intestinal barrier tissues.

17.
Heart ; 109(9): 674-685, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-36914250

RESUMO

BACKGROUND: The changes which typically occur in molecular causal risk factors and predictive biomarkers for cardiometabolic diseases across early life are not well characterised. METHODS: We quantified sex-specific trajectories of 148 metabolic trait concentrations including various lipoprotein subclasses from age 7 years to 25 years. Data were from 7065 to 7626 offspring (11 702 to14 797 repeated measures) of the Avon Longitudinal Study of Parents and Children birth cohort study. Outcomes were quantified using nuclear magnetic resonance spectroscopy at 7, 15, 18 and 25 years. Sex-specific trajectories of each trait were modelled using linear spline multilevel models. RESULTS: Females had higher very-low-density lipoprotein (VLDL) particle concentrations at 7 years. VLDL particle concentrations decreased from 7 years to 25 years with larger decreases in females, leading to lower VLDL particle concentrations at 25 years in females. For example, females had a 0.25 SD (95% CI 0.20 to 0.31) higher small VLDL particle concentration at 7 years; mean levels decreased by 0.06 SDs (95% CI -0.01 to 0.13) in males and 0.85 SDs (95% CI 0.79 to 0.90) in females from 7 years to 25 years, leading to 0.42 SDs (95% CI 0.35 to 0.48) lower small VLDL particle concentrations in females at 25 years. Females had lower high-density lipoprotein (HDL) particle concentrations at 7 years. HDL particle concentrations increased from 7 years to 25 years with larger increases among females leading to higher HDL particle concentrations in females at 25 years. CONCLUSION: Childhood and adolescence are important periods for the emergence of sex differences in atherogenic lipids and predictive biomarkers for cardiometabolic disease, mostly to the detriment of males.


Assuntos
Aterosclerose , Lipoproteínas , Adolescente , Humanos , Criança , Masculino , Feminino , Adulto Jovem , Adulto , Estudos de Coortes , Estudos Longitudinais , Biomarcadores
18.
Nat Commun ; 14(1): 670, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36810851

RESUMO

In the long-lived naked mole-rat (NMR), the entire process of oogenesis occurs postnatally. Germ cell numbers increase significantly in NMRs between postnatal days 5 (P5) and P8, and germs cells positive for proliferation markers (Ki-67, pHH3) are present at least until P90. Using pluripotency markers (SOX2 and OCT4) and the primordial germ cell (PGC) marker BLIMP1, we show that PGCs persist up to P90 alongside germ cells in all stages of female differentiation and undergo mitosis both in vivo and in vitro. We identified VASA+ SOX2+ cells at 6 months and at 3-years in subordinate and reproductively activated females. Reproductive activation was associated with proliferation of VASA+ SOX2+ cells. Collectively, our results suggest that highly desynchronized germ cell development and the maintenance of a small population of PGCs that can expand upon reproductive activation are unique strategies that could help to maintain the NMR's ovarian reserve for its 30-year reproductive lifespan.


Assuntos
Oogênese , Reserva Ovariana , Animais , Feminino , Diferenciação Celular , Células Germinativas , Mitose , Ovário , Ratos-Toupeira
19.
Mucosal Immunol ; 15(3): 491-503, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35177818

RESUMO

IL-36 cytokines are emerging as potent orchestrators of intestinal inflammation and are being implicated in the pathogenesis of inflammatory bowel diseases (IBD). However, the mechanisms through which these cytokines mediate these effects remain to be fully uncovered. Here, we report specifically elevated expression of IL-36α, and not IL-36ß or IL-36γ in the serum of newly diagnosed, treatment naïve, paediatric IBD patients and identify T cells as primary cellular mediators of IL-36 responses in the inflamed gut. IL-36R expression on CD4+ T cells was found to promote intestinal pathology in a murine model of colitis. Consistent with these effects, IL-36R can act as a potent instructor of CD4+ T cell differentiation in vivo, enhancing Th1 responses, while inhibiting the generation of Tregs. In addition, loss of IL-36 responsiveness significantly reduced the migration of pathogenic CD4+ T cells towards intestinal tissues and IL-36 was found to act, uniquely among IL-1 family members, to induce the expression of gut homing receptors in proinflammatory murine and human CD4+ T cells. These data reveal an important role for IL-36 cytokines in driving the colitogenic potential of CD4+ T cells and identify a new mechanism through which they may contribute to disease pathogenesis.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Interleucinas/imunologia , Animais , Criança , Colite/metabolismo , Citocinas/metabolismo , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Camundongos , Fenótipo , Linfócitos T Auxiliares-Indutores/metabolismo
20.
Sci Immunol ; 7(78): eade5728, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36525507

RESUMO

Interleukin-1 (IL-1) family cytokines are key barrier cytokines that are typically expressed as inactive, or partially active, precursors that require proteolysis within their amino termini for activation. IL-37 is an enigmatic member of the IL-1 family that has been proposed to be activated by caspase-1 and to exert anti-inflammatory activity through engagement of the IL-18R and SIGIRR. However, here we show that the longest IL-37 isoform, IL-37b, exhibits robust proinflammatory activity upon amino-terminal proteolysis by neutrophil elastase or cathepsin S. In sharp contrast, caspase-1 failed to process or activate IL-37 at concentrations that robustly activated its canonical substrate, IL-1ß. IL-37 and IL-36 exhibit high structural homology, and, consistent with this, a K53-truncated form of IL-37, mimicking the cathepsin S-processed form of this cytokine, was found to exert its proinflammatory effects via IL-36 receptor engagement and produced an inflammatory signature practically identical to IL-36. Administration of K53-truncated IL-37b intraperitoneally into wild-type mice also elicited an inflammatory response that was attenuated in IL-36R-/- animals. These data demonstrate that, in common with other IL-1 family members, mature IL-37 can also elicit proinflammatory effects upon processing by specific proteases.


Assuntos
Interleucina-1 , Peptídeo Hidrolases , Receptores de Interleucina , Animais , Camundongos , Caspases , Catepsinas , Citocinas , Interleucina-1/metabolismo , Células Mieloides , Receptores de Interleucina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA