Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 9(95): eade3814, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38787963

RESUMO

Patients with heart failure (HF) often experience repeated acute decompensation and develop comorbidities such as chronic kidney disease and frailty syndrome. Although this suggests pathological interaction among comorbidities, the mechanisms linking them are poorly understood. Here, we identified alterations in hematopoietic stem cells (HSCs) as a critical driver of recurrent HF and associated comorbidities. Bone marrow transplantation from HF-experienced mice resulted in spontaneous cardiac dysfunction and fibrosis in recipient mice, as well as increased vulnerability to kidney and skeletal muscle insults. HF enhanced the capacity of HSCs to generate proinflammatory macrophages. In HF mice, global chromatin accessibility analysis and single-cell RNA-seq showed that transforming growth factor-ß (TGF-ß) signaling was suppressed in HSCs, which corresponded with repressed sympathetic nervous activity in bone marrow. Transplantation of bone marrow from mice in which TGF-ß signaling was inhibited similarly exacerbated cardiac dysfunction. Collectively, these results suggest that cardiac stress modulates the epigenome of HSCs, which in turn alters their capacity to generate cardiac macrophage subpopulations. This change in HSCs may be a common driver of repeated HF events and comorbidity by serving as a key carrier of "stress memory."


Assuntos
Insuficiência Cardíaca , Imunidade Inata , Memória Imunológica , Camundongos Endogâmicos C57BL , Animais , Insuficiência Cardíaca/imunologia , Camundongos , Masculino , Multimorbidade , Fator de Crescimento Transformador beta/metabolismo , Células-Tronco Hematopoéticas/imunologia , Transdução de Sinais/imunologia , Macrófagos/imunologia , Imunidade Treinada
2.
JCI Insight ; 7(22)2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36509286

RESUMO

Recent studies have shown that cellular metabolism is tightly linked to the regulation of immune cells. Here, we show that activation of cholesterol metabolism, involving cholesterol uptake, synthesis, and autophagy/lipophagy, is integral to innate immune responses in macrophages. In particular, cholesterol accumulation within endosomes and lysosomes is a hallmark of the cellular cholesterol dynamics elicited by Toll-like receptor 4 activation and is required for amplification of myeloid differentiation primary response 88 (Myd88) signaling. Mechanistically, Myd88 binds cholesterol via its CLR recognition/interaction amino acid consensus domain, which promotes the protein's self-oligomerization. Moreover, a novel supramolecular compound, polyrotaxane (PRX), inhibited Myd88­dependent inflammatory macrophage activation by decreasing endolysosomal cholesterol via promotion of cholesterol trafficking and efflux. PRX activated liver X receptor, which led to upregulation of ATP binding cassette transporter A1, thereby promoting cholesterol efflux. PRX also inhibited atherogenesis in Ldlr-/- mice. In humans, cholesterol levels in circulating monocytes correlated positively with the severity of atherosclerosis. These findings demonstrate that dynamic changes in cholesterol metabolism are mechanistically linked to Myd88­dependent inflammatory programs in macrophages and support the notion that cellular cholesterol metabolism is integral to innate activation of macrophages and is a potential therapeutic and diagnostic target for inflammatory diseases.


Assuntos
Aterosclerose , Macrófagos , Camundongos , Humanos , Animais , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Macrófagos/metabolismo , Aterosclerose/metabolismo , Colesterol/metabolismo , Receptores X do Fígado/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo
3.
Nat Commun ; 13(1): 7058, 2022 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-36411280

RESUMO

Muscle regeneration requires the coordination of muscle stem cells, mesenchymal fibro-adipogenic progenitors (FAPs), and macrophages. How macrophages regulate the paracrine secretion of FAPs during the recovery process remains elusive. Herein, we systemically investigated the communication between CD206+ M2-like macrophages and FAPs during the recovery process using a transgenic mouse model. Depletion of CD206+ M2-like macrophages or deletion of CD206+ M2-like macrophages-specific TGF-ß1 gene induces myogenesis and muscle regeneration. We show that depletion of CD206+ M2-like macrophages activates FAPs and activated FAPs secrete follistatin, a promyogenic factor, thereby boosting the recovery process. Conversely, deletion of the FAP-specific follistatin gene results in impaired muscle stem cell function, enhanced fibrosis, and delayed muscle regeneration. Mechanistically, CD206+ M2-like macrophages inhibit the secretion of FAP-derived follistatin via TGF-ß signaling. Here we show that CD206+ M2-like macrophages constitute a microenvironment for FAPs and may regulate the myogenic potential of muscle stem/satellite cells.


Assuntos
Adipogenia , Folistatina , Animais , Camundongos , Macrófagos , Camundongos Transgênicos , Músculos , Receptor de Manose/imunologia
4.
Nat Commun ; 13(1): 5408, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36109558

RESUMO

The liver stores glycogen and releases glucose into the blood upon increased energy demand. Group 2 innate lymphoid cells (ILC2) in adipose and pancreatic tissues are known for their involvement in glucose homeostasis, but the metabolic contribution of liver ILC2s has not been studied in detail. Here we show that liver ILC2s are directly involved in the regulation of blood glucose levels. Mechanistically, interleukin (IL)-33 treatment induces IL-13 production in liver ILC2s, while directly suppressing gluconeogenesis in a specific Hnf4a/G6pc-high primary hepatocyte cluster via Stat3. These hepatocytes significantly interact with liver ILC2s via IL-13/IL-13 receptor signaling. The results of transcriptional complex analysis and GATA3-ChIP-seq, ATAC-seq, and scRNA-seq trajectory analyses establish a positive regulatory role for the transcription factor GATA3 in IL-13 production by liver ILC2s, while AP-1 family members are shown to suppress IL-13 release. Thus, we identify a regulatory role and molecular mechanism by which liver ILC2s contribute to glucose homeostasis.


Assuntos
Gluconeogênese , Interleucina-13 , Glicemia/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Glicogênio/metabolismo , Imunidade Inata , Interleucina-13/metabolismo , Fígado/metabolismo , Linfócitos/metabolismo , Receptores de Interleucina-13/metabolismo , Fator de Transcrição AP-1/metabolismo
5.
Nat Aging ; 2(2): 105-114, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-37117756

RESUMO

Canonically, hormones are produced in the endocrine organs and delivered to target tissues. However, for steroids, the concept of tissue intracrinology, whereby hormones are produced in the tissues where they exert their effect without release into circulation, has been proposed, but its role in physiology/disease remains unclear. The meibomian glands in the eyelids produce oil to prevent tear evaporation, which reduces with aging. Here, we demonstrate that (re)activation of local intracrine activity through nicotinamide adenine dinucleotide (NAD+)-dependent circadian 3ß-hydroxyl-steroid dehydrogenase (3ß-HSD) activity ameliorates age-associated meibomian gland dysfunction and accompanying evaporative dry eye disease. Genetic ablation of 3ß-HSD nullified local steroidogenesis and led to atrophy of the meibomian gland. Conversely, reactivation of 3ß-HSD activity by boosting its coenzyme NAD+ availability improved glandular cell proliferation and alleviated the dry eye disease phenotype. Both women and men express 3ß-HSD in the meibomian gland. Enhancing local steroidogenesis may help combat age-associated meibomian gland dysfunction.


Assuntos
Síndromes do Olho Seco , Disfunção da Glândula Tarsal , Feminino , Humanos , NAD , Glândulas Tarsais , Lágrimas/fisiologia , Esteroides , Hormônios
6.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34426497

RESUMO

Skeletal muscle atrophy is caused by various conditions, including aging, disuse related to a sedentary lifestyle and lack of physical activity, and cachexia. Our insufficient understanding of the molecular mechanism underlying muscle atrophy limits the targets for the development of effective pharmacologic treatments and preventions. Here, we identified Krüppel-like factor 5 (KLF5), a zinc-finger transcription factor, as a key mediator of the early muscle atrophy program. KLF5 was up-regulated in atrophying myotubes as an early response to dexamethasone or simulated microgravity in vitro. Skeletal muscle-selective deletion of Klf5 significantly attenuated muscle atrophy induced by mechanical unloading in mice. Transcriptome- and genome-wide chromatin accessibility analyses revealed that KLF5 regulates atrophy-related programs, including metabolic changes and E3-ubiquitin ligase-mediated proteolysis, in coordination with Foxo1. The synthetic retinoic acid receptor agonist Am80, a KLF5 inhibitor, suppressed both dexamethasone- and microgravity-induced muscle atrophy in vitro and oral Am80 ameliorated disuse- and dexamethasone-induced atrophy in mice. Moreover, in three independent sets of transcriptomic data from human skeletal muscle, KLF5 expression significantly increased with age and the presence of sarcopenia and correlated positively with the expression of the atrophy-related ubiquitin ligase genes FBXO32 and TRIM63 These findings demonstrate that KLF5 is a key transcriptional regulator mediating muscle atrophy and that pharmacological intervention with Am80 is a potentially preventive treatment.


Assuntos
Benzoatos/farmacologia , Desenvolvimento de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/fisiologia , Músculo Esquelético/efeitos dos fármacos , Atrofia Muscular/tratamento farmacológico , Tetra-Hidronaftalenos/farmacologia , Animais , Dexametasona/toxicidade , Glucocorticoides/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
7.
Nat Commun ; 12(1): 1910, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33771995

RESUMO

Cardiac arrhythmias are a primary contributor to sudden cardiac death, a major unmet medical need. Because right ventricular (RV) dysfunction increases the risk for sudden cardiac death, we examined responses to RV stress in mice. Among immune cells accumulated in the RV after pressure overload-induced by pulmonary artery banding, interfering with macrophages caused sudden death from severe arrhythmias. We show that cardiac macrophages crucially maintain cardiac impulse conduction by facilitating myocardial intercellular communication through gap junctions. Amphiregulin (AREG) produced by cardiac macrophages is a key mediator that controls connexin 43 phosphorylation and translocation in cardiomyocytes. Deletion of Areg from macrophages led to disorganization of gap junctions and, in turn, lethal arrhythmias during acute stresses, including RV pressure overload and ß-adrenergic receptor stimulation. These results suggest that AREG from cardiac resident macrophages is a critical regulator of cardiac impulse conduction and may be a useful therapeutic target for the prevention of sudden death.


Assuntos
Anfirregulina/fisiologia , Arritmias Cardíacas/complicações , Morte Súbita Cardíaca/prevenção & controle , Macrófagos/fisiologia , Miocárdio/metabolismo , Anfirregulina/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Morte Súbita Cardíaca/etiologia , Feminino , Junções Comunicantes/fisiologia , Células HeLa , Humanos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(25): 14365-14375, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32513690

RESUMO

Proper resolution of inflammation is vital for repair and restoration of homeostasis after tissue damage, and its dysregulation underlies various noncommunicable diseases, such as cardiovascular and metabolic diseases. Macrophages play diverse roles throughout initial inflammation, its resolution, and tissue repair. Differential metabolic reprogramming is reportedly required for induction and support of the various macrophage activation states. Here we show that a long noncoding RNA (lncRNA), lncFAO, contributes to inflammation resolution and tissue repair in mice by promoting fatty acid oxidation (FAO) in macrophages. lncFAO is induced late after lipopolysaccharide (LPS) stimulation of cultured macrophages and in Ly6Chi monocyte-derived macrophages in damaged tissue during the resolution and reparative phases. We found that lncFAO directly interacts with the HADHB subunit of mitochondrial trifunctional protein and activates FAO. lncFAO deletion impairs resolution of inflammation related to endotoxic shock and delays resolution of inflammation and tissue repair in a skin wound. These results demonstrate that by tuning mitochondrial metabolism, lncFAO acts as a node of immunometabolic control in macrophages during the resolution and repair phases of inflammation.


Assuntos
Ácidos Graxos/metabolismo , Inflamação/imunologia , Macrófagos/imunologia , Subunidade beta da Proteína Mitocondrial Trifuncional/genética , RNA Longo não Codificante/metabolismo , Animais , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Lipopolissacarídeos/imunologia , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Masculino , Camundongos , Subunidade beta da Proteína Mitocondrial Trifuncional/metabolismo , Oxirredução , Cultura Primária de Células , RNA Longo não Codificante/genética , Pele/imunologia , Pele/lesões , Cicatrização/imunologia
9.
Cell Immunol ; 323: 33-40, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29108648

RESUMO

Cyclosporin A (CsA) is a well-known immunosuppressant that is used against steroid-resistant asthma. Group 2 innate lymphoid cells (ILC2s) and type 2 helper T (Th2) cells produce Th2 cytokines including IL-5 and play important roles in asthma pathogenesis. Here, we studied the effects of CsA in allergen-induced lung inflammation in mice and found that CsA decreased the number of lung ILC2s and attenuated papain-induced activation of ILC2s accompanied with IL-5 expression. The ILC2 suppression mediated by CsA was not observed in culture or in lymphocyte-deficient Rag2-/- mice. Thus, we propose a new suppressive effect of CsA, i.e., administration of CsA indirectly suppresses maintenance and activation of lung ILC2s in addition to direct suppression of T-cell activation and cytokine production.


Assuntos
Ciclosporina/farmacologia , Linfócitos/efeitos dos fármacos , Pneumonia/tratamento farmacológico , Alérgenos , Animais , Asma/imunologia , Asma/metabolismo , Ciclosporina/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Imunidade Inata/imunologia , Pulmão/imunologia , Ativação Linfocitária , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Papaína/farmacologia , Pneumonia/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/imunologia
10.
JCI Insight ; 2(7): e90721, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28405615

RESUMO

IL-33 is one of the critical cytokines that activates group 2 innate lymphoid cells (ILC2s) and mediates allergic reactions. Accumulating evidence suggests that IL-33 is also involved in the pathogenesis of several chronic inflammatory diseases. Previously, we generated an IL-5 reporter mouse and revealed that lung IL-5-producing ILC2s played essential roles in regulating eosinophil biology. In this study, we evaluated the consequences of IL-33 administration over a long period, and we observed significant expansion of ILC2s and eosinophils surrounding pulmonary arteries. Unexpectedly, pulmonary arteries showed severe occlusive hypertrophy that was ameliorated in IL-5- or eosinophil-deficient mice, but not in Rag2-deficient mice. This indicates that IL-5-producing ILC2s and eosinophils play pivotal roles in pulmonary arterial hypertrophy. Administration of a clinically used vasodilator was effective in reducing IL-33-induced hypertrophy and repressed the expansion of ILC2s and eosinophils. Taken together, these observations demonstrate a previously unrecognized mechanism in the development of pulmonary arterial hypertrophy and the causative roles of ILC2 in the process.


Assuntos
Proteínas de Ligação a DNA/genética , Eosinófilos/imunologia , Interleucina-33/farmacologia , Interleucina-5/imunologia , Artéria Pulmonar/patologia , Células Th2/imunologia , Animais , Hipertrofia , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Immunology ; 147(1): 21-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26425820

RESUMO

Group 2 innate lymphoid cells (ILC2s) produce a significant amount of interleukin-5 (IL-5), which supports eosinophil responses in various tissues; they also produce IL-13, which induces mucus production and contributes to tissue repair or fibrosis. The ILC2s are activated by alarmins, such as IL-33 released from epithelia, macrophages and natural killer T (NKT) cells in response to infection and allergen exposure, leading to epithelial injury. We examined gene expression in lung ILC2s and found that ILC2s expressed Ifngr1, the receptor for interferon-γ (IFN-γ). Interferon-γ severely inhibited IL-5 and IL-13 production by lung and kidney ILC2s. To evaluate the effects in vivo, we used α-galactosylceramide (α-GalCer) to induce NKT cells to produce IL-33 and IFN-γ. Intraperitoneal injection of α-GalCer in mice induced NKT cell activation resulting in IL-5 and IL-13 production by ILC2s. Administration of anti-IFN-γ together with α-GalCer significantly enhanced the production of IL-5 and IL-13 by ILC2s in lung and kidney. Conversely, cytokine production from ILC2s was markedly suppressed after injection of exogenous IL-33 in Il33(-/-) mice pre-treated with α-GalCer. Hence, IFN-γ induced or already present in tissues can impact downstream pleiotropic functions mediated by ILC2s, such as inflammation and tissue repair.


Assuntos
Imunidade Inata/efeitos dos fármacos , Interferon gama/metabolismo , Rim/metabolismo , Pulmão/metabolismo , Linfócitos/metabolismo , Animais , Células Cultivadas , Galactosilceramidas/farmacologia , Interferon gama/imunologia , Interferon gama/farmacologia , Interleucina-13/imunologia , Interleucina-13/metabolismo , Interleucina-33/deficiência , Interleucina-33/genética , Interleucina-5/genética , Interleucina-5/imunologia , Interleucina-5/metabolismo , Rim/citologia , Rim/efeitos dos fármacos , Rim/imunologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Ativação Linfocitária , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Fenótipo , Receptores de Interferon/agonistas , Receptores de Interferon/genética , Receptores de Interferon/imunologia , Receptor de Interferon gama
12.
Immunol Lett ; 144(1-2): 24-32, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22445356

RESUMO

Extracellular nucleotides such as ATP, ADP, UTP, UDP and UDPG can trigger intracellular signal transduction via purinergic (P2Y) receptors, and their interaction induces a wide range of biological effects in various cells. In this study, we investigated P2Y expression and the effects of nucleotides on chemotaxis and phagocytosis in human neutrophils. RT-PCR detected broad expression of P2Y subfamilies in neutrophils, as well as monocytes. Moreover, intracellular Ca(2+) increased in response to ATP, ADP, UTP and UDP in these cells, suggesting that P2Y receptors were functionally expressed. In neutrophils, chemotactic activity was increased significantly in response to ATP and ADP, and moderately in response to UTP and UDP; actin polymerization by ATP, ADP, UTP and UDP was also evident in the cells. Interestingly, we found that ATP and ADP, which enhanced chemotaxis activity significantly, had inhibitory effects on phagocytosis by neutrophils. These findings provide new evidence for the regulation of neutrophil phagocytosis by nucleotides. Furthermore, this inhibitory effect was completely lost upon co-culture with fMLP or LPS, known constituents of bacteria, resulting in recovery of normal phagocytic activity. Taken together, these findings suggest that ATP and ADP constantly stimulate the chemotactic activity of neutrophils in peripheral blood, but may inhibit their phagocytic activity until they encounter pathogens, in order to prevent them acting against self-tissues or cells, as fMLP and LPS commonly present in pathogens would again trigger normal phagocytic activity.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Regulação para Baixo , Lipopolissacarídeos/imunologia , Neutrófilos/imunologia , Nucleotídeos/farmacologia , Fagocitose/efeitos dos fármacos , Receptores Purinérgicos P2Y/metabolismo , Sinalização do Cálcio , Humanos , Imunidade Inata , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Nucleotídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Purinérgicos P2Y/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...