Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 289
Filtrar
1.
Nat Commun ; 12(1): 7041, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34873175

RESUMO

Despite unprecedented responses of some cancers to immune checkpoint blockade (ICB) therapies, the application of checkpoint inhibitors in pancreatic cancer has been unsuccessful. Glucocorticoids and glucocorticoid receptor (GR) signaling are long thought to suppress immunity by acting on immune cells. Here we demonstrate a previously undescribed tumor cell-intrinsic role for GR in activating PD-L1 expression and repressing the major histocompatibility complex class I (MHC-I) expression in pancreatic ductal adenocarcinoma (PDAC) cells through transcriptional regulation. In mouse models of PDAC, either tumor cell-specific depletion or pharmacologic inhibition of GR leads to PD-L1 downregulation and MHC-I upregulation in tumor cells, which in turn promotes the infiltration and activity of cytotoxic T cells, enhances anti-tumor immunity, and overcomes resistance to ICB therapy. In patients with PDAC, GR expression correlates with high PD-L1 expression, low MHC-I expression, and poor survival. Our results reveal GR signaling in cancer cells as a tumor-intrinsic mechanism of immunosuppression and suggest that therapeutic targeting of GR is a promising way to sensitize pancreatic cancer to immunotherapy.


Assuntos
Antígeno B7-H1/imunologia , Carcinoma Ductal Pancreático/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Neoplasias Pancreáticas/imunologia , Receptores de Glucocorticoides/imunologia , Evasão Tumoral/imunologia , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Immunoblotting , Imunoterapia/métodos , Estimativa de Kaplan-Meier , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Evasão Tumoral/genética
2.
Nat Commun ; 12(1): 6664, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795263

RESUMO

Crohn's disease is an inflammatory disease of the gastrointestinal tract characterized by an aberrant response to microbial and environmental triggers. This includes an altered microbiome dominated by Enterobacteriaceae and in particular adherent-invasive E. coli (AIEC). Clinical evidence implicates periods of psychological stress in Crohn's disease exacerbation, and disturbances in the gut microbiome might contribute to the pathogenic mechanism. Here we show that stress-exposed mice develop ileal dysbiosis, dominated by the expansion of Enterobacteriaceae. In an AIEC colonisation model, stress-induced glucocorticoids promote apoptosis of CD45+CD90+ cells that normally produce IL-22, a cytokine that is essential for the maintenance of ileal mucosal barrier integrity. Blockade of glucocorticoid signaling or administration of recombinant IL-22 restores mucosal immunity, prevents ileal dysbiosis, and blocks AIEC expansion. We conclude that psychological stress impairs IL-22-driven protective immunity in the gut, which creates a favorable niche for the expansion of pathobionts that have been implicated in Crohn's disease. Importantly, this work also shows that immunomodulation can counteract the negative effects of psychological stress on gut immunity and hence disease-associated dysbiosis.


Assuntos
Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Estresse Psicológico/imunologia , Animais , Aderência Bacteriana/imunologia , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Disbiose/microbiologia , Enterobacteriaceae/classificação , Enterobacteriaceae/genética , Enterobacteriaceae/imunologia , Escherichia coli/imunologia , Escherichia coli/fisiologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Íleo/imunologia , Íleo/microbiologia , Íleo/patologia , Interleucinas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Receptores de Glucocorticoides/imunologia , Receptores de Glucocorticoides/metabolismo , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismo , Interleucina 22
3.
Sci Rep ; 11(1): 22913, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824310

RESUMO

Inflammation is a physiological process whose deregulation causes some diseases including cancer. Nuclear Factor kB (NF-kB) is a family of ubiquitous and inducible transcription factors, in which the p65/p50 heterodimer is the most abundant complex, that play critical roles mainly in inflammation. Glucocorticoid Receptor (GR) is a ligand-activated transcription factor and acts as an anti-inflammatory agent and immunosuppressant. Thus, NF-kB and GR are physiological antagonists in the inflammation process. Here we show that in mice and humans there is a spliced variant of p65, named p65 iso5, which binds the corticosteroid hormone dexamethasone amplifying the effect of the glucocorticoid receptor and is expressed in the liver of patients with hepatic cirrhosis and hepatocellular carcinoma (HCC). Furthermore, we have quantified the gene expression level of p65 and p65 iso5 in the PBMC of patients affected by SARS-CoV-2 disease. The results showed that in these patients the p65 and p65 iso5 mRNA levels are higher than in healthy subjects. The ability of p65 iso5 to bind dexamethasone and the regulation of the glucocorticoid (GC) response in the opposite way of the wild type improves our knowledge and understanding of the anti-inflammatory response and identifies it as a new therapeutic target to control inflammation and related diseases.


Assuntos
Inflamação/imunologia , Receptores de Glucocorticoides/metabolismo , Fator de Transcrição RelA/metabolismo , Corticosteroides/metabolismo , Adulto , Processamento Alternativo , Animais , COVID-19/imunologia , Carcinoma Hepatocelular/metabolismo , Dexametasona/metabolismo , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Glucocorticoides/metabolismo , Hepatite/metabolismo , Humanos , Inflamação/metabolismo , Leucócitos Mononucleares/metabolismo , Fígado/metabolismo , Hepatopatias/imunologia , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Isoformas de Proteínas , Receptores de Glucocorticoides/imunologia , SARS-CoV-2/patogenicidade , Fator de Transcrição RelA/imunologia , Fator de Transcrição RelA/fisiologia
4.
Viruses ; 13(11)2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34835102

RESUMO

Following acute infection, herpes simplex virus 1 (HSV-1) establishes lifelong latency in neurons, including sensory neurons within trigeminal ganglia. During latency, lytic cycle viral gene expression is silenced. However, stressful stimuli can trigger reactivation from latency. The viral tegument protein, VP-16, transactivates all immediate early (IE) promoters during productive infection. Conversely, cellular factors are expected to trigger viral gene expression during early stages of reactivation from latency and in non-neuronal cells that do not support high levels of productive infection. The glucocorticoid receptor (GR), synthetic corticosteroid dexamethasone, and certain stress-induced transcription factors cooperatively transactivate infected cell protein 0 (ICP0) and ICP4 promoters. Since ICP27 protein expression is required for productive infection, we hypothesized that the ICP27 promoter is transactivated by stress-induced transcription factors. New studies have demonstrated that ICP27 enhancer sequences were transactivated by GR and Krüppel-like factor 15 (KLF15). Mutation of a consensus Sp1 binding site within ICP27 enhancer sequences impaired transactivation by GR and KLF15. Chromatin immunoprecipitation studies have demonstrated that GR and KLF15 occupy ICP27 promoter sequences during productive infection. Cells transfected with an ICP27 enhancer fragment revealed the GR and KLF15 occupancy of ICP27 enhancer sequences required the intact Sp1 binding site. Notably, GR and KLF15 form a feed-forward transcription loop in response to stress, suggesting these cellular factors promote viral replication following stressful stimuli.


Assuntos
Herpes Simples/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/imunologia , Receptores de Glucocorticoides/imunologia , Latência Viral , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Imunoprecipitação da Cromatina , Regulação Viral da Expressão Gênica , Fatores de Transcrição Kruppel-Like/imunologia , Camundongos , Fatores de Transcrição , Células Vero , Ativação Viral
5.
J Steroid Biochem Mol Biol ; 213: 105952, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34274458

RESUMO

The impacts of glucocorticoids (GCs) are mainly mediated by a nuclear receptor (GR) existing in almost every tissue. The GR regulates a wide range of physiological functions, including inflammation, cell metabolism, and differentiation playing a major role in cellular responses to GCs and stress. Therefore, the dysregulation or disruption of GR can cause deficiencies in the adaptation to stress and the preservation of homeostasis. The number of GR polymorphisms associated with different diseases has been mounting per year. Tackling these clinical complications obliges a comprehensive understanding of the molecular network action of GCs at the level of the GR structure and its signaling pathways. Beyond genetic variation in the GR gene, epigenetic changes can enhance our understanding of causal factors involved in the development of diseases and identifying biomarkers. In this review, we highlight the relationships of GC receptor gene polymorphisms and epigenetics with different diseases.


Assuntos
Doenças Autoimunes/genética , Doenças Ósseas/genética , Doenças Cardiovasculares/genética , Epigênese Genética , Transtornos Mentais/genética , Doenças Metabólicas/genética , Receptores de Glucocorticoides/genética , Adaptação Fisiológica/genética , Adaptação Fisiológica/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Doenças Ósseas/imunologia , Doenças Ósseas/patologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/patologia , Metilação de DNA , Glucocorticoides/imunologia , Glucocorticoides/metabolismo , Homeostase/genética , Homeostase/imunologia , Humanos , Inflamação , Transtornos Mentais/imunologia , Transtornos Mentais/patologia , Doenças Metabólicas/imunologia , Doenças Metabólicas/patologia , Polimorfismo Genético , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/imunologia , Transdução de Sinais , Estresse Fisiológico/genética , Estresse Fisiológico/imunologia
6.
J Biol Chem ; 296: 100687, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33891947

RESUMO

Glucocorticoids are potent anti-inflammatory drugs that are used to treat an extraordinary range of human disease, including COVID-19, underscoring the ongoing importance of understanding their molecular mechanisms. Early studies of GR signaling led to broad acceptance of models in which glucocorticoid receptor (GR) monomers tether repressively to inflammatory transcription factors, thus abrogating inflammatory gene expression. However, newer data challenge this core concept and present an exciting opportunity to reframe our understanding of GR signaling. Here, we present an alternate, two-part model for transcriptional repression by glucocorticoids. First, widespread GR-mediated induction of transcription results in rapid, primary repression of inflammatory gene transcription and associated enhancers through competition-based mechanisms. Second, a subset of GR-induced genes, including targets that are regulated in coordination with inflammatory transcription factors such as NF-κB, exerts secondary repressive effects on inflammatory gene expression. Within this framework, emerging data indicate that the gene set regulated through the cooperative convergence of GR and NF-κB signaling is central to the broad clinical effectiveness of glucocorticoids in terminating inflammation and promoting tissue repair.


Assuntos
Anti-Inflamatórios/uso terapêutico , Tratamento Farmacológico da COVID-19 , Dexametasona/uso terapêutico , Glucocorticoides/uso terapêutico , NF-kappa B/genética , Receptores de Glucocorticoides/genética , Animais , COVID-19/imunologia , COVID-19/patologia , COVID-19/virologia , Regulação da Expressão Gênica , Genômica/métodos , Humanos , Inflamação/prevenção & controle , Modelos Genéticos , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/imunologia , SARS-CoV-2/crescimento & desenvolvimento , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Transdução de Sinais , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/imunologia
7.
Cell Rep ; 34(6): 108742, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33567280

RESUMO

Glucocorticoids (GCs) are effective anti-inflammatory drugs; yet, their mechanisms of action are poorly understood. GCs bind to the glucocorticoid receptor (GR), a ligand-gated transcription factor controlling gene expression in numerous cell types. Here, we characterize GR's protein interactome and find the SETD1A (SET domain containing 1A)/COMPASS (complex of proteins associated with Set1) histone H3 lysine 4 (H3K4) methyltransferase complex highly enriched in activated mouse macrophages. We show that SETD1A/COMPASS is recruited by GR to specific cis-regulatory elements, coinciding with H3K4 methylation dynamics at subsets of sites, upon treatment with lipopolysaccharide (LPS) and GCs. By chromatin immunoprecipitation sequencing (ChIP-seq) and RNA-seq, we identify subsets of GR target loci that display SETD1A occupancy, H3K4 mono-, di-, or tri-methylation patterns, and transcriptional changes. However, our data on methylation status and COMPASS recruitment suggest that SETD1A has additional transcriptional functions. Setd1a loss-of-function studies reveal that SETD1A/COMPASS is required for GR-controlled transcription of subsets of macrophage target genes. We demonstrate that the SETD1A/COMPASS complex cooperates with GR to mediate anti-inflammatory effects.


Assuntos
Elementos Facilitadores Genéticos/imunologia , Macrófagos/imunologia , Complexos Multiproteicos , RNA-Seq , Receptores de Glucocorticoides , Transcrição Gênica/imunologia , Animais , Inflamação/genética , Inflamação/imunologia , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/imunologia
8.
Am J Reprod Immunol ; 85(5): e13366, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33099840

RESUMO

PROBLEM: The immune system represents a leading pathway of interest in the pathophysiology of preterm birth. The majority of human clinical studies interrogating this pathway have utilized circulating immune biomarkers; however, these concentrations typically reflect only basal production but not key functional properties of the immune system, particularly variation in the pro-inflammatory response to antigen challenge and the regulation of this response. Thus, in this study, we utilized an ex vivo stimulation protocol that quantifies these processes, and we examined their prospective association with the gestation length and risk of preterm birth. METHOD OF STUDY: Immune responsiveness and regulation were assessed in 128 pregnant women in mid-gestation using an ex vivo stimulation protocol. Maternal pro-inflammatory responsivity of leukocytes was quantified by assessing the release of the pro-inflammatory cytokines IL-6, TNF-α, and IL-1ß in response to antigen stimulation, and regulation of the pro-inflammatory response was quantified by assessing the suppression of stimulated cytokine response upon co-incubation with increasing dexamethasone concentrations (ie, glucocorticoid receptor resistance; GRR). RESULTS: Higher maternal GRR, indicating impaired regulation of the pro-inflammatory response, was significantly and independently associated with shorter gestational length (ß = -0.42, p = .0091) and a 3.0-fold increase in risk for preterm birth (OR = 3.01, 95% CI = 1.17-7.70, p = .0218). Basal circulating IL-6 and TNF-α were not associated with either outcome. CONCLUSION: The association of maternal GRR with length of gestation and preterm birth risk suggests that the processes represented by this measure-maternal pro-inflammatory propensity and immune regulation-may provide further mechanistic insight into the pathophysiology of preterm birth.


Assuntos
Citocinas/imunologia , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Leucócitos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Nascimento Prematuro/imunologia , Receptores de Glucocorticoides/imunologia , Adulto , Células Cultivadas , Citocinas/sangue , Feminino , Humanos , Leucócitos/imunologia , Gravidez
9.
J Virol ; 95(4)2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33208447

RESUMO

Following acute infection, herpes simplex virus 1 (HSV-1) lytic cycle viral gene expression is silenced; consequently, lifelong latency in neurons is established. Certain external stimuli that trigger reactivation from latency also activate the glucocorticoid receptor (GR). The synthetic corticosteroid dexamethasone, but not a GR-specific antagonist, increases the frequency of explant-induced reactivation from latency and stimulates productive infection. Furthermore, dexamethasone increases expression of cellular transcription factors in trigeminal ganglionic neurons: for example, SLUG and three Krüppel-like transcription factor (KLF) family members, KLF4, KLF15, and promyelocytic leukemia zinc finger protein (PLZF). Consequently, we hypothesized that stress-induced transcription factors stimulate expression of ICP4, a viral transcriptional regulator required for productive infection. New studies demonstrated that GR and KLF4, PLZF, or SLUG cooperatively transactivate the ICP4 enhancer upstream of a minimal promoter in monkey kidney cells (Vero) and mouse neuroblastoma cells (Neuro-2A). Strikingly, mutagenesis of two KLF4/Sp1 binding sites reduced GR- plus KLF4-, PLZF-, or SLUG-mediated transactivation to basal levels. A consensus enhancer (E)-Box adjacent to a KLF4/Sp1 binding site was also required for GR- and SLUG-, but not KLF family member-, mediated transactivation of the ICP4 promoter. Chromatin immunoprecipitation studies (ChIP) revealed GR and stress-induced transcription factors occupy ICP4 enhancer sequences. Conversely, specific binding was generally reduced in the KLF4/Sp1 mutant. Furthermore, GR and SLUG occupancy of ICP4 enhancer sequences was reduced in the E-Box mutant. Based on these studies, we suggest stressful stimuli can trigger productive infection because GR and specific stress-induced transcription factors activate ICP4 expression.IMPORTANCE Certain stressful stimuli activate the glucocorticoid receptor (GR) and increase the incidence of herpes simplex virus 1 (HSV-1) reactivation from latency. For example, a corticosteroid antagonist impairs productive infection and virus shedding following explant of trigeminal ganglia from latently infected mice. Infected cell protein 4 (ICP4) is the only immediate early viral transcriptional regulator required for productive infection, suggesting stressful stimuli stimulate ICP4 expression. New studies revealed GR and stress-induced transcription factors identified during reactivation from latency, SLUG and three Krüppel-like transcription factor family members (KLF4, KLF15, and promyelocytic leukemia zinc finger protein), cooperatively transactivate the ICP4 enhancer. Two KLF4 consensus binding sites were crucial for cooperative transactivation of the ICP4 enhancer. A consensus enhancer-box also mediated cooperative transactivation of the ICP4 enhancer by GR and SLUG. The ability of GR and stress-induced transcription factors to transactivate ICP4 enhancer activity is predicted to trigger productive infection following stressful stimuli.


Assuntos
Herpes Simples , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/imunologia , Receptores de Glucocorticoides/imunologia , Ativação Viral , Latência Viral , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação Viral da Expressão Gênica , Herpes Simples/imunologia , Herpes Simples/virologia , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/imunologia , Camundongos , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia , Fatores de Transcrição da Família Snail/imunologia , Ativação Transcricional , Células Vero
10.
Nat Rev Immunol ; 21(4): 233-243, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33149283

RESUMO

Glucocorticoids (GCs) are small lipid hormones produced by the adrenals that maintain organismal homeostasis. Circadian and stress-induced changes in systemic GC levels regulate metabolism, cardiovascular and neural function, reproduction and immune activity. Our understanding of GC effects on immunity comes largely from administration of exogenous GCs to treat immune or inflammatory disorders. However, it is increasingly clear that endogenous GCs both promote and suppress T cell immunity. Examples include selecting an appropriate repertoire of T cell receptor (TCR) self-affinities in the thymus, regulating T cell trafficking between anatomical compartments, suppressing type 1 T helper (TH1) cell responses while permitting TH2 cell and, especially, IL-17-producing T helper cell responses, and promoting memory T cell differentiation and maintenance. Furthermore, in addition to functioning at a distance, extra-adrenal (local) production allows GCs to act as paracrine signals, specifically targeting activated T cells in various contexts in the thymus, mucosa and tumours. These pleiotropic effects on different T cell populations during development and immune responses provide a nuanced understanding of how GCs shape immunity.


Assuntos
Glucocorticoides/imunologia , Linfopoese/imunologia , Receptores de Glucocorticoides/imunologia , Linfócitos T/imunologia , Diferenciação Celular/imunologia , Linhagem da Célula , Rearranjo Gênico do Linfócito T/genética , Rearranjo Gênico do Linfócito T/imunologia , Humanos , Tolerância Imunológica/imunologia , Inflamação/imunologia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T Auxiliares-Indutores/imunologia , Timo
11.
Pharmacol Res ; 166: 105334, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33276107

RESUMO

Glucocorticoids (GCs) plays an irreplaceable role in inflammation and immune responses, fat metabolism and sugar metabolism, it is often used for the treatment of asthma, rheumatoid arthritis and allergic rhinitis clinically, but long-term or high-dose use will produce adverse drug reactions (ADRs). Its biological action is mediated by glucocorticoid receptors (GRs), of which the oligomerization state is closely related to the target gene of which the GRs act. A leading hypothesis is that the beneficial anti-inflammatory effects of GCs occur through the transrepression mechanism mediated by GR monomers, while ADRs may be dependent on the transactivation mechanism mediated by GR dimers. However, in recent years, multiple studies have shown that the transactivation and transrepression functions of the GR dimer also confer anti-inflammatory effects. Furthermore, some studies have shown that some selective glucocorticoid receptor agonists and modulators (SEGRAMs) have good separation characteristics (i.e., preferentially mediate the transrepression of proinflammatory genes or preferentially activate anti-inflammatory target genes). This article reviewed the formation of GR dimers, the role of GR dimers in the inflammation and immune responses, and the progress of SEGRAMs to provide novel ideas for further understanding the anti-inflammatory mechanism of GR and the development of SEGRAMs.


Assuntos
Inflamação/imunologia , Receptores de Glucocorticoides/imunologia , Animais , Glucocorticoides/imunologia , Humanos , Imunidade , Multimerização Proteica , Receptores de Glucocorticoides/química
12.
Cell Rep ; 33(7): 108404, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33207196

RESUMO

Stressful experiences early in life can increase the risk of cardiovascular diseases. However, it remains largely unknown how stress influences susceptibility to the disease onset. Here, we show that exposure to brain-processed stress disrupts myocardial growth by reducing cardiomyocyte mitotic activity. Activation of the glucocorticoid receptor (GR), the primary stress response pathway, reduces cardiomyocyte numbers, disrupts trabecular formation, and leads to contractile dysfunction of the developing myocardium. However, a physiological level of GR signaling is required to prevent cardiomyocyte hyperproliferation. Mechanistically, we identify an antagonistic interaction between the GR and the cytokine interleukin-4 (IL-4) as a key player in cardiac development. IL-4 signals transcription of key regulators of cell-cycle progression in cardiomyocytes via signal transducer and activator of transcription 3 (Stat3). GR, on the contrary, inhibits this signaling system. Thus, our findings uncover an interplay between stress and immune signaling pathways critical to orchestrating physiological growth of the heart.


Assuntos
Interleucina-4 , Miócitos Cardíacos , Receptores de Glucocorticoides , Estresse Fisiológico , Animais , Feminino , Masculino , Camundongos , Glucocorticoides/metabolismo , Interleucina-4/imunologia , Interleucina-4/metabolismo , Interleucina-4/fisiologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitose , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Receptores de Glucocorticoides/imunologia , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição STAT3/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
13.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796067

RESUMO

Neurotropic Alphaherpesvirinae subfamily members such as bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) establish and maintain lifelong latent infections in neurons. Following infection of ocular, oral, or nasal cavities, sensory neurons within trigeminal ganglia (TG) are an important site for latency. Certain external stressors can trigger reactivation from latency, in part because activation of the glucocorticoid receptor (GR) stimulates productive infection and promoters that drive expression of key viral transcriptional regulators. The Akt serine/threonine protein kinase family is linked to maintaining latency. For example, Akt3 is detected in more TG neurons during BoHV-1 latency than in reactivation and uninfected calves. Furthermore, Akt signaling correlates with maintaining HSV-1 latency in certain neuronal models of latency. Finally, an active Akt protein kinase is crucial for the ability of the HSV-1 latency-associated transcript (LAT) to inhibit apoptosis in neuronal cell lines. Consequently, we hypothesized that viral and/or cellular factors impair stress-induced transcription and reduce the incidence of reactivation triggered by low levels of stress. New studies demonstrate that Akt1 and Akt2, but not Akt3, significantly reduced GR-mediated transactivation of the BoHV-1 immediate early transcription unit 1 (IEtu1) promoter, the HSV-1 infected cell protein 0 (ICP0) promoter, and the mouse mammary tumor virus long terminal repeat (MMTV-LTR). Akt3, but not Akt1 or Akt2, significantly enhanced neurite formation in mouse neuroblastoma cells, which correlates with repairing damaged neurons. These studies suggest that unique biological properties of the three Akt family members promote the maintenance of latency in differentiated neurons.IMPORTANCE External stressful stimuli are known to increase the incidence of reactivation of Alphaherpesvirinae subfamily members. Activation of the glucocorticoid receptor (GR) by the synthetic corticosteroid dexamethasone (DEX) stimulates bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) reactivation. Furthermore, GR and dexamethasone stimulate productive infection and promoters that drive expression of viral transcriptional regulators. These observations lead us to predict that stress-induced transcription is impaired by factors abundantly expressed during latency. Interestingly, activation of the Akt family of serine/threonine protein kinases is linked to maintenance of latency. New studies reveal that Akt1 and Ak2, but not Akt3, impaired GR- and dexamethasone-mediated transactivation of the BoHV-1 immediate early transcription unit 1 and HSV-1 ICP0 promoters. Strikingly, Akt3, but not Akt1 or Akt2, stimulated neurite formation in mouse neuroblastoma cells, a requirement for neurogenesis. These studies provide insight into how Akt family members may promote the maintenance of lifelong latency.


Assuntos
Herpes Simples/imunologia , Infecções por Herpesviridae/imunologia , Interações Hospedeiro-Patógeno/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Células Receptoras Sensoriais/virologia , Animais , Bovinos , Diferenciação Celular , Linhagem Celular Tumoral , Herpes Simples/genética , Herpes Simples/patologia , Herpes Simples/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Camundongos , Neuritos/imunologia , Neuritos/ultraestrutura , Neuritos/virologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/imunologia , Células Receptoras Sensoriais/imunologia , Células Receptoras Sensoriais/patologia , Transdução de Sinais , Ativação Transcricional/imunologia , Gânglio Trigeminal/imunologia , Gânglio Trigeminal/patologia , Gânglio Trigeminal/virologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
15.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32629782

RESUMO

Breast cancer (BC) patients experience increased stress with elevated cortisol levels, increasing risk of cancer recurrence. Cortisol binds to a cytoplasmic receptor, glucocorticoid receptor (GR) encoded by GR gene (NR3C1). We hypothesized that not only cancer cells, but even immune cells in the tumor microenvironment (TME) may contribute to GR expression in bulk tumor and influence prognosis. To test this, mRNA expression data was accessed from METABRIC and TCGA. "High" and "low" expression was based on highest and lowest quartiles of NR3C1 gene expression, respectively. Single-cell sequencing data were obtained from GSE75688 and GSE114725 cohorts. Computer algorithms CIBERSORT, Gene Set Enrichment Analysis and TIMER were used. GR-high BC has better median disease-free and disease-specific survival. Single cell sequencing data showed higher GR expression on immune cells compared to cancer and stromal cells. Positive correlation between GR-high BC and CD8+ T-cells was noted. In GR-high tumors, higher cytolytic activity (CYT) with decreased T-regulatory and T-follicular helper cells was observed. High GR expression was associated with lower proliferation index Ki67, enriched in IL-2_STAT5, apoptosis, KRAS, TGF-ß signaling, and epithelial-to-mesenchymal transition. Immune cells significantly contribute to GR expression of bulk BC. GR-high BC has a favorable TME with higher CYT with favorable outcomes.


Assuntos
Neoplasias da Mama/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Receptores de Glucocorticoides/genética , Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/fisiologia , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Glucocorticoides/genética , Glucocorticoides/metabolismo , Humanos , Recidiva Local de Neoplasia/genética , Prognóstico , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/imunologia , Receptores de Glucocorticoides/metabolismo , Microambiente Tumoral/imunologia
16.
J Immunol ; 205(3): 619-629, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32571841

RESUMO

Glucocorticoids promote CXCR4 expression by T cells, monocytes, macrophages, and eosinophils, but it is not known if glucocorticoids regulate CXCR4 in B cells. Considering the important contributions of CXCR4 to B cell development and function, we investigated the glucocorticoid/CXCR4 axis in mice. We demonstrate that glucocorticoids upregulate CXCR4 mRNA and protein in mouse B cells. Using a novel strain of mice lacking glucocorticoid receptors (GRs) specifically in B cells, we show that reduced CXCR4 expression associated with GR deficiency results in impaired homing of mature B cells to bone marrow, whereas migration to other lymphoid tissues is independent of B cell GRs. The exchange of mature B cells between blood and bone marrow is sensitive to small, physiologic changes in glucocorticoid activity, as evidenced by the lack of circadian rhythmicity in GR-deficient B cell counts normally associated with diurnal patterns of glucocorticoid secretion. B cellGRKO mice mounted normal humoral responses to immunizations with T-dependent and T-independent (Type 1) Ags, but Ab responses to a multivalent T-independent (Type 2) Ag were impaired, a surprise finding considering the immunosuppressive properties commonly attributed to glucocorticoids. We propose that endogenous glucocorticoids regulate a dynamic mode of B cell migration specialized for rapid exchange between bone marrow and blood, perhaps as a means to optimize humoral immunity during diurnal periods of activity.


Assuntos
Linfócitos B/imunologia , Medula Óssea/imunologia , Movimento Celular/imunologia , Receptores de Glucocorticoides/imunologia , Animais , Movimento Celular/genética , Masculino , Camundongos , Camundongos Knockout , Receptores CXCR4/genética , Receptores CXCR4/imunologia , Receptores de Glucocorticoides/genética
17.
Am J Physiol Endocrinol Metab ; 319(1): E48-E66, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32315214

RESUMO

Although glucocorticoid resistance contributes to increased inflammation, individuals with posttraumatic stress disorder (PTSD) exhibit increased glucocorticoid receptor (GR) sensitivity along with increased inflammation. It is not clear how inflammation coexists with a hyperresponsive hypothalamic-pituitary-adrenal (HPA) axis. To understand this better, we developed and analyzed an integrated mathematical model for the HPA axis and the immune system. We performed mathematical simulations for a dexamethasone (DEX) suppression test and IC50-dexamethasone for cytokine suppression by varying model parameters. The model analysis suggests that increasing the steepness of the dose-response curve for GR activity may reduce anti-inflammatory effects of GRs at the ambient glucocorticoid levels, thereby increasing proinflammatory response. The adaptive response of proinflammatory cytokine-mediated stimulatory effects on the HPA axis is reduced due to dominance of the GR-mediated negative feedback on the HPA axis. To verify these hypotheses, we analyzed the clinical data on neuroendocrine variables and cytokines obtained from war-zone veterans with and without PTSD. We observed significant group differences for cortisol and ACTH suppression tests, proinflammatory cytokines TNFα and IL6, high-sensitivity C-reactive protein, promoter methylation of GR gene, and IC50-DEX for lysozyme suppression. Causal inference modeling revealed significant associations between cortisol suppression and post-DEX cortisol decline, promoter methylation of human GR gene exon 1F (NR3C1-1F), IC50-DEX, and proinflammatory cytokines. We noted significant mediation effects of NR3C1-1F promoter methylation on inflammatory cytokines through changes in GR sensitivity. Our findings suggest that increased GR sensitivity may contribute to increased inflammation; therefore, interventions to restore GR sensitivity may normalize inflammation in PTSD.


Assuntos
Citocinas/imunologia , Glucocorticoides/imunologia , Receptores de Glucocorticoides/imunologia , Transtornos de Estresse Pós-Traumáticos/imunologia , Hormônio Adrenocorticotrópico/imunologia , Hormônio Adrenocorticotrópico/metabolismo , Adulto , Campanha Afegã de 2001- , Proteína C-Reativa/imunologia , Estudos de Casos e Controles , Ritmo Circadiano , Metilação de DNA , Dexametasona , Glucocorticoides/metabolismo , Humanos , Hidrocortisona/imunologia , Hidrocortisona/metabolismo , Sistema Hipotálamo-Hipofisário/imunologia , Sistema Hipotálamo-Hipofisário/metabolismo , Inflamação , Concentração Inibidora 50 , Interleucina-6/imunologia , Guerra do Iraque 2003-2011 , Masculino , Modelos Teóricos , Testes de Função Adreno-Hipofisária , Sistema Hipófise-Suprarrenal/imunologia , Sistema Hipófise-Suprarrenal/metabolismo , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Veteranos
18.
Clin Ther ; 42(6): 993-1006, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32224031

RESUMO

PURPOSE: Chronic and persistent exposure to negative stress can lead to adverse consequences on health. Particularly, psychosocial factors were found to increase the risk and outcome of respiratory diseases like asthma. Glucocorticoids (GCs) are the most efficient anti-inflammatory therapy for asthma. However, a significant proportion of patients don't respond adequately to GC administration. GC sensitivity is modulated by genetic and acquired disease-related factors. Additionally, it was proposed that endogenous corticosteroids may limit certain actions of synthetic GCs, contributing to insensitivity. Psychological and physiological stresses activate the hypothalamic-pituitary-adrenal axis, increasing cortisol levels. Here, we review the mechanism involved in altered GC sensitivity in asthmatic patients under stressful situations. Strategies for modulation GC sensitivity and improving GC therapy are discussed. METHODS: PubMed was searched for publications on psychological chronic stress and asthma, GC resistance in asthma, biological mechanisms for GC resistance, and drugs for steroid-resistant asthma, including highly potent GCs. FINDINGS: GC resistance in patients with severe disease remains a major clinical problem. In asthma, experimental and clinical evidence suggests that chronic stress induces inflammatory changes, contributing to a worse GC response. GC resistant patients can be treated with other broad-spectrum anti-inflammatory drugs, but these generally have major side effects. Different mechanisms of GC resistance have been described and might be useful for developing new therapeutic strategies against it. Novel drugs, such as highly potent GCs, phosphoinositide 3-kinase-delta inhibitors that reestablish histone deacetylase-2 function, decrease of GC receptor phosphorylation by p38 mitogen-activated protein kinase inhibitors, or phosphatase activators, are currently in clinical development and might be combined with GC therapy in the future. Furthermore, microRNAs (small noncoding RNA molecules) operate as posttranscriptional regulators, providing another level of control of GC receptor levels. Empirical results allow postulating that the detection and study of microRNAs might be a promising approach to better characterize and treat asthmatic patients. IMPLICATIONS: Many molecular and cellular pathobiological mechanisms are responsible of GC resistance. Therefore detecting specific biomarkers to help identify patients who would benefit from new therapies is crucial. Stress consitutes a negative aspect of current lifestyles that increase asthma morbidity and mortality. Adequate stress management could be an important and positive intervention.


Assuntos
Asma/tratamento farmacológico , Resistência a Medicamentos , Glucocorticoides/uso terapêutico , Receptores de Glucocorticoides/imunologia , Estresse Psicológico/complicações , Animais , Asma/imunologia , Doença Crônica , Humanos , Estresse Psicológico/imunologia
19.
Biol Pharm Bull ; 43(1): 31-35, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31902928

RESUMO

Asthma patients are classified by phenotype and endotype. Although symptoms in most asthma patients are well controlled by glucocorticoid treatment, certain populations of severe eosinophilic asthma patients in T-helper 2 (Th2)/type 2 asthma and neutrophilic asthma patients in non-Th2/type 2 asthma show insensitivity to inhaled or oral glucocorticoid therapy. In some cases of severe eosinophilic asthma, eosinophils remain in the lungs despite glucocorticoid therapy. It was reported that interleukin (IL)-33-induced activation of type 2 innate lymphoid cells (ILC2) was resistant to glucocorticoid treatment in certain allergic conditions. Regarding neutrophilic airway inflammation in steroid-resistant asthma, IL-17 derived from Th17 cells and IL-8 and tumor necrosis factor-α derived mainly from macrophages were reported to be involved in the pathogenesis. Recently, "NETosis," a specific cell death of neutrophils, has been reported to be involved in asthmatic airway inflammation. When NETosis is induced in asthma, aggravation of inflammation and delay of tissue repair could occur, suggesting that NETosis may be associated with the development of steroid-resistant asthma. This article reviews the pathogenesis of steroid-resistant asthma by focusing mainly on neutrophils.


Assuntos
Asma/imunologia , Resistência a Medicamentos/imunologia , Neutrófilos/imunologia , Animais , Asma/tratamento farmacológico , Glucocorticoides/uso terapêutico , Humanos , Inflamação/imunologia , Receptores de Glucocorticoides/imunologia , Células Th2/imunologia
20.
Front Immunol ; 11: 628287, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33679723

RESUMO

Following severe trauma, fracture healing is impaired because of overwhelming systemic and local inflammation. Glucocorticoids (GCs), acting via the glucocorticoid receptor (GR), influence fracture healing by modulating the trauma-induced immune response. GR dimerization-dependent gene regulation is essential for the anti-inflammatory effects of GCs. Therefore, we investigated in a murine trauma model of combined femur fracture and thoracic trauma, whether effective GR dimerization influences the pathomechanisms of trauma-induced compromised fracture healing. To this end, we used mice with decreased GR dimerization ability (GRdim). The healing process was analyzed by cytokine/chemokine multiplex analysis, flow cytometry, gene-expression analysis, histomorphometry, micro-computed tomography, and biomechanical testing. GRdim mice did not display a systemic or local hyper-inflammation upon combined fracture and thorax trauma. Strikingly, we discovered that GRdim mice were protected from fracture healing impairment induced by the additional thorax trauma. Collectively and in contrast to previous studies describing the beneficial effects of intact GR dimerization in inflammatory models, we report here an adverse role of intact GR dimerization in trauma-induced compromised fracture healing.


Assuntos
Consolidação da Fratura/imunologia , Multimerização Proteica/imunologia , Receptores de Glucocorticoides/imunologia , Traumatismos Torácicos/imunologia , Animais , Consolidação da Fratura/genética , Masculino , Camundongos , Camundongos Transgênicos , Multimerização Proteica/genética , Receptores de Glucocorticoides/genética , Traumatismos Torácicos/genética , Traumatismos Torácicos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...