Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nat Immunol ; 23(3): 431-445, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35228694

RESUMO

Chronic inflammation triggers compensatory immunosuppression to stop inflammation and minimize tissue damage. Studies have demonstrated that endoplasmic reticulum (ER) stress augments the suppressive phenotypes of immune cells; however, the molecular mechanisms underpinning this process and how it links to the metabolic reprogramming of immunosuppressive macrophages remain elusive. In the present study, we report that the helper T cell 2 cytokine interleukin-4 and the tumor microenvironment increase the activity of a protein kinase RNA-like ER kinase (PERK)-signaling cascade in macrophages and promote immunosuppressive M2 activation and proliferation. Loss of PERK signaling impeded mitochondrial respiration and lipid oxidation critical for M2 macrophages. PERK activation mediated the upregulation of phosphoserine aminotransferase 1 (PSAT1) and serine biosynthesis via the downstream transcription factor ATF-4. Increased serine biosynthesis resulted in enhanced mitochondrial function and α-ketoglutarate production required for JMJD3-dependent epigenetic modification. Inhibition of PERK suppressed macrophage immunosuppressive activity and could enhance the efficacy of immune checkpoint programmed cell death protein 1 inhibition in melanoma. Our findings delineate a previously undescribed connection between PERK signaling and PSAT1-mediated serine metabolism critical for promoting immunosuppressive function in M2 macrophages.


Assuntos
Estresse do Retículo Endoplasmático , eIF-2 Quinase , Estresse do Retículo Endoplasmático/genética , Macrófagos/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
2.
Nat Immunol ; 22(11): 1403-1415, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34686867

RESUMO

Tumor-associated macrophages (TAMs) display pro-tumorigenic phenotypes for supporting tumor progression in response to microenvironmental cues imposed by tumor and stromal cells. However, the underlying mechanisms by which tumor cells instruct TAM behavior remain elusive. Here, we uncover that tumor-cell-derived glucosylceramide stimulated unconventional endoplasmic reticulum (ER) stress responses by inducing reshuffling of lipid composition and saturation on the ER membrane in macrophages, which induced IRE1-mediated spliced XBP1 production and STAT3 activation. The cooperation of spliced XBP1 and STAT3 reinforced the pro-tumorigenic phenotype and expression of immunosuppressive genes. Ablation of XBP1 expression with genetic manipulation or ameliorating ER stress responses by facilitating LPCAT3-mediated incorporation of unsaturated lipids to the phosphatidylcholine hampered pro-tumorigenic phenotype and survival in TAMs. Together, we uncover the unexpected roles of tumor-cell-produced lipids that simultaneously orchestrate macrophage polarization and survival in tumors via induction of ER stress responses and reveal therapeutic targets for sustaining host antitumor immunity.


Assuntos
Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Ativação de Macrófagos , Melanoma/metabolismo , Lipídeos de Membrana/metabolismo , Neoplasias Cutâneas/metabolismo , Macrófagos Associados a Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Retículo Endoplasmático/ultraestrutura , Glucosilceramidase/metabolismo , Membranas Intracelulares/ultraestrutura , Melanoma/genética , Melanoma/ultraestrutura , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/ultraestrutura , Evasão Tumoral , Microambiente Tumoral , Macrófagos Associados a Tumor/ultraestrutura , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
3.
Cell ; 170(4): 649-663.e13, 2017 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-28802038

RESUMO

Elevated risk of developing Alzheimer's disease (AD) is associated with hypomorphic variants of TREM2, a surface receptor required for microglial responses to neurodegeneration, including proliferation, survival, clustering, and phagocytosis. How TREM2 promotes such diverse responses is unknown. Here, we find that microglia in AD patients carrying TREM2 risk variants and TREM2-deficient mice with AD-like pathology have abundant autophagic vesicles, as do TREM2-deficient macrophages under growth-factor limitation or endoplasmic reticulum (ER) stress. Combined metabolomics and RNA sequencing (RNA-seq) linked this anomalous autophagy to defective mammalian target of rapamycin (mTOR) signaling, which affects ATP levels and biosynthetic pathways. Metabolic derailment and autophagy were offset in vitro through Dectin-1, a receptor that elicits TREM2-like intracellular signals, and cyclocreatine, a creatine analog that can supply ATP. Dietary cyclocreatine tempered autophagy, restored microglial clustering around plaques, and decreased plaque-adjacent neuronal dystrophy in TREM2-deficient mice with amyloid-ß pathology. Thus, TREM2 enables microglial responses during AD by sustaining cellular energetic and biosynthetic metabolism.


Assuntos
Doença de Alzheimer/patologia , Metabolismo Energético , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Doença de Alzheimer/metabolismo , Animais , Autofagia , Creatinina/análogos & derivados , Creatinina/metabolismo , Modelos Animais de Doenças , Humanos , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Microglia/patologia , Neuritos/metabolismo , Placa Amiloide/metabolismo , Receptores Imunológicos/genética , Serina-Treonina Quinases TOR/metabolismo
4.
Nat Immunol ; 20(6): 687-700, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31061528

RESUMO

Most tissue-resident macrophage populations develop during embryogenesis, self-renew in the steady state and expand during type 2 immunity. Whether shared mechanisms regulate the proliferation of macrophages in homeostasis and disease is unclear. Here we found that the transcription factor Bhlhe40 was required in a cell-intrinsic manner for the self-renewal and maintenance of large peritoneal macrophages (LPMs), but not that of other tissue-resident macrophages. Bhlhe40 was necessary for the proliferation, but not the polarization, of LPMs in response to the cytokine IL-4. During infection with the helminth Heligmosomoides polygyrus bakeri, Bhlhe40 was required for cell cycling of LPMs. Bhlhe40 repressed the expression of genes encoding the transcription factors c-Maf and Mafb and directly promoted expression of transcripts encoding cell cycle-related proteins to enable the proliferation of LPMs. In LPMs, Bhlhe40 bound to genomic sites co-bound by the macrophage lineage-determining factor PU.1 and to unique sites, including Maf and loci encoding cell-cycle-related proteins. Our findings demonstrate a tissue-specific control mechanism that regulates the proliferation of resident macrophages in homeostasis and type 2 immunity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Homeodomínio/genética , Homeostase/genética , Homeostase/imunologia , Imunidade/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores , Ciclo Celular/genética , Ciclo Celular/imunologia , Proliferação de Células , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Infecções por Helicobacter/genética , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Proteínas de Homeodomínio/metabolismo , Imunofenotipagem , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/metabolismo , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Transcriptoma
5.
Cell ; 166(1): 63-76, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27293185

RESUMO

Activated effector T (TE) cells augment anabolic pathways of metabolism, such as aerobic glycolysis, while memory T (TM) cells engage catabolic pathways, like fatty acid oxidation (FAO). However, signals that drive these differences remain unclear. Mitochondria are metabolic organelles that actively transform their ultrastructure. Therefore, we questioned whether mitochondrial dynamics controls T cell metabolism. We show that TE cells have punctate mitochondria, while TM cells maintain fused networks. The fusion protein Opa1 is required for TM, but not TE cells after infection, and enforcing fusion in TE cells imposes TM cell characteristics and enhances antitumor function. Our data suggest that, by altering cristae morphology, fusion in TM cells configures electron transport chain (ETC) complex associations favoring oxidative phosphorylation (OXPHOS) and FAO, while fission in TE cells leads to cristae expansion, reducing ETC efficiency and promoting aerobic glycolysis. Thus, mitochondrial remodeling is a signaling mechanism that instructs T cell metabolic programming.


Assuntos
Dinâmica Mitocondrial , Linfócitos T/citologia , Linfócitos T/metabolismo , Animais , Diferenciação Celular , Transporte de Elétrons , Ácidos Graxos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Glicólise , Humanos , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Transdução de Sinais , Linfócitos T/imunologia
6.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23746840

RESUMO

A "switch" from oxidative phosphorylation (OXPHOS) to aerobic glycolysis is a hallmark of T cell activation and is thought to be required to meet the metabolic demands of proliferation. However, why proliferating cells adopt this less efficient metabolism, especially in an oxygen-replete environment, remains incompletely understood. We show here that aerobic glycolysis is specifically required for effector function in T cells but that this pathway is not necessary for proliferation or survival. When activated T cells are provided with costimulation and growth factors but are blocked from engaging glycolysis, their ability to produce IFN-γ is markedly compromised. This defect is translational and is regulated by the binding of the glycolysis enzyme GAPDH to AU-rich elements within the 3' UTR of IFN-γ mRNA. GAPDH, by engaging/disengaging glycolysis and through fluctuations in its expression, controls effector cytokine production. Thus, aerobic glycolysis is a metabolically regulated signaling mechanism needed to control cellular function.


Assuntos
Glicólise , Ativação Linfocitária , Fosforilação Oxidativa , Linfócitos T/citologia , Linfócitos T/metabolismo , Regiões 3' não Traduzidas , Animais , Proliferação de Células , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Interferon gama/genética , Listeria monocytogenes , Listeriose/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Biossíntese de Proteínas , Linfócitos T/imunologia
7.
Immunity ; 49(6): 1021-1033.e6, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30566880

RESUMO

Metabolic engagement is intrinsic to immune cell function. Prostaglandin E2 (PGE2) has been shown to modulate macrophage activation, yet how PGE2 might affect metabolism is unclear. Here, we show that PGE2 caused mitochondrial membrane potential (Δψm) to dissipate in interleukin-4-activated (M(IL-4)) macrophages. Effects on Δψm were a consequence of PGE2-initiated transcriptional regulation of genes, particularly Got1, in the malate-aspartate shuttle (MAS). Reduced Δψm caused alterations in the expression of 126 voltage-regulated genes (VRGs), including those encoding resistin-like molecule α (RELMα), a key marker of M(IL-4) cells, and genes that regulate the cell cycle. The transcription factor ETS variant 1 (ETV1) played a role in the regulation of 38% of the VRGs. These results reveal ETV1 as a Δψm-sensitive transcription factor and Δψm as a mediator of mitochondrial-directed nuclear gene expression.


Assuntos
Núcleo Celular/efeitos dos fármacos , Dinoprostona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Animais , Núcleo Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Interleucina-4/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Nat Immunol ; 15(4): 323-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24562310

RESUMO

The ligation of Toll-like receptors (TLRs) leads to rapid activation of dendritic cells (DCs). However, the metabolic requirements that support this process remain poorly defined. We found that DC glycolytic flux increased within minutes of exposure to TLR agonists and that this served an essential role in supporting the de novo synthesis of fatty acids for the expansion of the endoplasmic reticulum and Golgi required for the production and secretion of proteins that are integral to DC activation. Signaling via the kinases TBK1, IKKɛ and Akt was essential for the TLR-induced increase in glycolysis by promoting the association of the glycolytic enzyme HK-II with mitochondria. In summary, we identified the rapid induction of glycolysis as an integral component of TLR signaling that is essential for the anabolic demands of the activation and function of DCs.


Assuntos
Células Dendríticas/imunologia , Glicólise , Quinase I-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T/imunologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Ácidos Graxos/biossíntese , Glicólise/efeitos dos fármacos , Glicólise/genética , Glicólise/imunologia , Hexoquinase/metabolismo , Quinase I-kappa B/genética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Receptores Toll-Like/agonistas
9.
Nat Immunol ; 15(9): 846-55, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25086775

RESUMO

Alternative (M2) activation of macrophages driven via the α-chain of the receptor for interleukin 4 (IL-4Rα) is important for immunity to parasites, wound healing, the prevention of atherosclerosis and metabolic homeostasis. M2 polarization is dependent on fatty acid oxidation (FAO), but the source of the fatty acids that support this metabolic program has not been clear. We found that the uptake of triacylglycerol substrates via the scavenger receptor CD36 and their subsequent lipolysis by lysosomal acid lipase (LAL) was important for the engagement of elevated oxidative phosphorylation, enhanced spare respiratory capacity (SRC), prolonged survival and expression of genes that together define M2 activation. Inhibition of lipolysis suppressed M2 activation during infection with a parasitic helminth and blocked protective responses to this pathogen. Our findings delineate a critical role for cell-intrinsic lysosomal lipolysis in M2 activation.


Assuntos
Antígenos CD36/imunologia , Ácidos Graxos/metabolismo , Interleucina-4/imunologia , Lipólise/imunologia , Lisossomos/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Fosforilação Oxidativa , Transdução de Sinais/imunologia , Esterol Esterase/imunologia , Animais , Respiração Celular , Helmintíase Animal/imunologia , Humanos , Camundongos , Consumo de Oxigênio , Receptores de Interleucina-4/imunologia , Transcriptoma
10.
Immunity ; 45(4): 817-830, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760338

RESUMO

Macrophage activation status is intrinsically linked to metabolic remodeling. Macrophages stimulated by interleukin 4 (IL-4) to become alternatively (or, M2) activated increase fatty acid oxidation and oxidative phosphorylation; these metabolic changes are critical for M2 activation. Enhanced glucose utilization is also characteristic of the M2 metabolic signature. Here, we found that increased glucose utilization is essential for M2 activation. Increased glucose metabolism in IL-4-stimulated macrophages required the activation of the mTORC2 pathway, and loss of mTORC2 in macrophages suppressed tumor growth and decreased immunity to a parasitic nematode. Macrophage colony stimulating factor (M-CSF) was implicated as a contributing upstream activator of mTORC2 in a pathway that involved PI3K and AKT. mTORC2 operated in parallel with the IL-4Rα-Stat6 pathway to facilitate increased glycolysis during M2 activation via the induction of the transcription factor IRF4. IRF4 expression required both mTORC2 and Stat6 pathways, providing an underlying mechanism to explain how glucose utilization is increased to support M2 activation.


Assuntos
Fatores Reguladores de Interferon/metabolismo , Ativação de Macrófagos/fisiologia , Macrófagos/fisiologia , Complexos Multiproteicos/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Interleucina-4/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT6/metabolismo
11.
Proc Natl Acad Sci U S A ; 119(23): e2204557119, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35653568

RESUMO

C-type lectin domain family 4, member a4 (Clec4a4) is a C-type lectin inhibitory receptor specific for glycans thought to be exclusively expressed on murine CD8α− conventional dendritic cells. Using newly generated Clec4a4-mCherry knock-in mice, we identify a subset of Clec4a4-expressing eosinophils uniquely localized in the small intestine lamina propria. Clec4a4+ eosinophils evinced an immunomodulatory signature, whereas Clec4a4− eosinophils manifested a proinflammatory profile. Clec4a4+ eosinophils expressed high levels of aryl hydrocarbon receptor (Ahr), which drove the expression of Clec4a4 as well as other immunomodulatory features, such as PD-L1. The abundance of Clec4a4+ eosinophils was dependent on dietary AHR ligands, increased with aging, and declined in inflammatory conditions. Mice lacking AHR in eosinophils expanded innate lymphoid cells of type 2 and cleared Nippostrongylus brasiliensis infection more effectively than did wild-type mice. These results highlight the heterogeneity of eosinophils in response to tissue cues and identify a unique AHR-dependent subset of eosinophils in the small intestine with an immunomodulatory profile.


Assuntos
Eosinófilos , Receptores de Hidrocarboneto Arílico , Receptores de Superfície Celular , Eosinofilia/terapia , Hipersensibilidade Alimentar/terapia , Imunomodulação , Intestino Delgado , Contagem de Leucócitos , Ligantes , Receptores de Hidrocarboneto Arílico/genética
12.
Immunity ; 42(3): 419-30, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25786174

RESUMO

Macrophage polarization involves a coordinated metabolic and transcriptional rewiring that is only partially understood. By using an integrated high-throughput transcriptional-metabolic profiling and analysis pipeline, we characterized systemic changes during murine macrophage M1 and M2 polarization. M2 polarization was found to activate glutamine catabolism and UDP-GlcNAc-associated modules. Correspondingly, glutamine deprivation or inhibition of N-glycosylation decreased M2 polarization and production of chemokine CCL22. In M1 macrophages, we identified a metabolic break at Idh, the enzyme that converts isocitrate to alpha-ketoglutarate, providing mechanistic explanation for TCA cycle fragmentation. (13)C-tracer studies suggested the presence of an active variant of the aspartate-arginosuccinate shunt that compensated for this break. Consistently, inhibition of aspartate-aminotransferase, a key enzyme of the shunt, inhibited nitric oxide and interleukin-6 production in M1 macrophages, while promoting mitochondrial respiration. This systems approach provides a highly integrated picture of the physiological modules supporting macrophage polarization, identifying potential pharmacologic control points for both macrophage phenotypes.


Assuntos
Redes Reguladoras de Genes/imunologia , Imunidade Inata , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Transcrição Gênica/imunologia , Animais , Ácido Argininossuccínico/imunologia , Ácido Argininossuccínico/metabolismo , Aspartato Aminotransferase Mitocondrial/genética , Aspartato Aminotransferase Mitocondrial/imunologia , Ácido Aspártico/imunologia , Ácido Aspártico/metabolismo , Quimiocina CCL22/genética , Quimiocina CCL22/imunologia , Ciclo do Ácido Cítrico , Regulação da Expressão Gênica , Glutamina/deficiência , Glicosilação , Interleucina-6/genética , Interleucina-6/imunologia , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/imunologia , Macrófagos/classificação , Macrófagos/citologia , Macrófagos/imunologia , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/imunologia , Camundongos , Mitocôndrias/genética , Mitocôndrias/imunologia , Óxido Nítrico/imunologia , Óxido Nítrico/metabolismo , Transdução de Sinais , Uridina Difosfato N-Acetilglicosamina/imunologia , Uridina Difosfato N-Acetilglicosamina/metabolismo
13.
Immunity ; 41(3): 351-353, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25238092

RESUMO

Proinflammatory macrophage activation is coupled to a metabolic switch toward glycolysis. In Cell Metabolism, Jin et al. (2014) show that this process is negatively regulated by mitochondrial electron transport chain complex I through both cell intrinsic and extrinsic pathways.


Assuntos
Reabsorção Óssea/complicações , Reabsorção Óssea/imunologia , Complexo I de Transporte de Elétrons/deficiência , Macrófagos/patologia , Doenças Mitocondriais/complicações , Doenças Mitocondriais/imunologia , Osteoclastos/patologia , Animais , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/imunologia , Feminino , Masculino
14.
Immunity ; 41(1): 75-88, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25001241

RESUMO

Generation of CD8(+) memory T cells requires metabolic reprogramming that is characterized by enhanced mitochondrial fatty-acid oxidation (FAO). However, where the fatty acids (FA) that fuel this process come from remains unclear. While CD8(+) memory T cells engage FAO to a greater extent, we found that they acquired substantially fewer long-chain FA from their external environment than CD8(+) effector T (Teff) cells. Rather than using extracellular FA directly, memory T cells used extracellular glucose to support FAO and oxidative phosphorylation (OXPHOS), suggesting that lipids must be synthesized to generate the substrates needed for FAO. We have demonstrated that memory T cells rely on cell intrinsic expression of the lysosomal hydrolase LAL (lysosomal acid lipase) to mobilize FA for FAO and memory T cell development. Our observations link LAL to metabolic reprogramming in lymphocytes and show that cell intrinsic lipolysis is deterministic for memory T cell fate.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Ácidos Graxos/metabolismo , Memória Imunológica/imunologia , Lipólise/imunologia , Esterol Esterase/metabolismo , 4-Butirolactona/análogos & derivados , 4-Butirolactona/farmacologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/genética , Ácidos Graxos/biossíntese , Glucose/metabolismo , Interleucina-15/imunologia , Interleucina-2/imunologia , Lipólise/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Oxirredução , Fosforilação Oxidativa , Oxigênio/metabolismo , Proteínas Quinases/genética , Interferência de RNA , RNA Interferente Pequeno , Esterol Esterase/biossíntese
16.
J Immunol ; 204(4): 923-932, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31900338

RESUMO

The transcription factor BHLHE40 is an emerging regulator of the immune system. Recent studies suggest that BHLHE40 regulates type 2 immunity, but this has not been demonstrated in vivo. We found that BHLHE40 is required in T cells for a protective TH2 cell response in mice infected with the helminth Heligmosomoides polygyrus bakeri H. polygyrus elicited changes in gene and cytokine expression by lamina propria CD4+ T cells, many of which were BHLHE40 dependent, including production of the common ß (CSF2RB) chain family cytokines GM-CSF and IL-5. In contrast to deficiency in GM-CSF or IL-5 alone, loss of both GM-CSF and IL-5 signaling impaired protection against H. polygyrus Overall, we show that BHLHE40 regulates the TH2 cell transcriptional program during helminth infection to support normal expression of Csf2, Il5, and other genes required for protection and reveal unexpected redundancy of common ß chain-dependent cytokines previously thought to possess substantially divergent functions.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Homeodomínio/metabolismo , Interleucina-5/metabolismo , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Células Th2/imunologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Subunidade beta Comum dos Receptores de Citocinas/genética , Subunidade beta Comum dos Receptores de Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Proteínas de Homeodomínio/genética , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/genética , Interleucina-5/antagonistas & inibidores , Interleucina-5/genética , Interleucina-5/imunologia , Camundongos , Camundongos Knockout , Mucosa/citologia , Mucosa/imunologia , Mucosa/metabolismo , Infecções por Strongylida/parasitologia , Células Th2/efeitos dos fármacos , Transcrição Gênica/imunologia
17.
Immunology ; 161(3): 165-174, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32418209

RESUMO

Metabolites are the essential substrates for epigenetic modification enzymes to write or erase the epigenetic blueprint in cells. Hence, the availability of nutrients and activity of metabolic pathways strongly influence the enzymatic function. Recent studies have shed light on the choreography between metabolome and epigenome in the control of immune cell differentiation and function, with a major focus on histone modifications. Yet, despite its importance in gene regulation, DNA methylation and its relationship with metabolism is relatively unclear. In this review, we will describe how the metabolic flux can influence epigenetic networks in innate and adaptive immune cells, with a focus on the DNA methylation cycle and the metabolites S-adenosylmethionine and α-ketoglutarate. Future directions will be discussed for this rapidly emerging field.


Assuntos
Ciclo do Ácido Cítrico/imunologia , Epigenoma/imunologia , Metaboloma/imunologia , Imunidade Adaptativa , Animais , Metilação de DNA , Epigênese Genética , Humanos , Imunidade Inata , Ácidos Cetoglutáricos/metabolismo , S-Adenosilmetionina/metabolismo
18.
PLoS Pathog ; 11(3): e1004676, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25816012

RESUMO

Over 25% of the world's population are infected with helminth parasites, the majority of which colonise the gastrointestinal tract. However, no vaccine is yet available for human use, and mechanisms of protective immunity remain unclear. In the mouse model of Heligmosomoides polygyrus infection, vaccination with excretory-secretory (HES) antigens from adult parasites elicits sterilising immunity. Notably, three purified HES antigens (VAL-1, -2 and -3) are sufficient for effective vaccination. Protection is fully dependent upon specific IgG1 antibodies, but passive transfer confers only partial immunity to infection, indicating that cellular components are also required. Moreover, immune mice show greater cellular infiltration associated with trapping of larvae in the gut wall prior to their maturation. Intra-vital imaging of infected intestinal tissue revealed a four-fold increase in extravasation by LysM+GFP+ myeloid cells in vaccinated mice, and the massing of these cells around immature larvae. Mice deficient in FcRγ chain or C3 complement component remain fully immune, suggesting that in the presence of antibodies that directly neutralise parasite molecules, the myeloid compartment may attack larvae more quickly and effectively. Immunity to challenge infection was compromised in IL-4Rα- and IL-25-deficient mice, despite levels of specific antibody comparable to immune wild-type controls, while deficiencies in basophils, eosinophils or mast cells or CCR2-dependent inflammatory monocytes did not diminish immunity. Finally, we identify a suite of previously uncharacterised heat-labile vaccine antigens with homologs in human and veterinary parasites that together promote full immunity. Taken together, these data indicate that vaccine-induced immunity to intestinal helminths involves IgG1 antibodies directed against secreted proteins acting in concert with IL-25-dependent Type 2 myeloid effector populations.


Assuntos
Anticorpos Anti-Helmínticos/imunologia , Antígenos de Helmintos/imunologia , Imunoglobulina G/imunologia , Interleucina-4/imunologia , Interleucinas/imunologia , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Vacinação , Animais , Anticorpos Anti-Helmínticos/genética , Humanos , Imunoglobulina G/genética , Interleucina-4/genética , Interleucinas/genética , Larva/imunologia , Camundongos , Camundongos Knockout , Infecções por Strongylida/genética , Infecções por Strongylida/prevenção & controle
19.
Cell Microbiol ; 17(6): 796-801, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25850569

RESUMO

Schistosomiasis is a neglected tropical disease caused by infection with trematode parasites of the genus Schistosoma. Despite ongoing treatment programmes, the prevalence of schistosomiasis has failed to decline and the disease remains a cause of severe morbidity in millions of people. Understanding the biology of egg production by schistosomes is critical since eggs allow transmission of the infection, and when trapped in host tissues induce the immune responses that are responsible for the pathologic changes that underlie disease development. Unusually among trematodes, adult schistosomes exhibit sexual dimorphism and display a fascinating codependency in that the female is dependent on the male to grow and sexually mature. Thus, virgin females are developmentally stunted compared with females from mixed-sex infections and are unable to lay eggs. Moreover, fecund female schistosomes rapidly lose the ability to produce eggs when placed in tissue culture. Here we discuss the metabolic regulation of egg production in schistosomes, and in particular the critical role played by fatty acid oxidation in this process.


Assuntos
Schistosoma/fisiologia , Zigoto/fisiologia , Animais , Ácidos Graxos/metabolismo , Feminino , Fertilidade , Humanos , Masculino , Oxirredução
20.
Proc Natl Acad Sci U S A ; 110(35): 14336-41, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23940348

RESUMO

A characteristic of memory T (TM) cells is their ability to mount faster and stronger responses to reinfection than naïve T (TN) cells do in response to an initial infection. However, the mechanisms that allow this rapid recall are not completely understood. We found that CD8 TM cells have more mitochondrial mass than CD8 TN cells and, that upon activation, the resulting secondary effector T (TE) cells proliferate more quickly, produce more cytokines, and maintain greater ATP levels than primary effector T cells. We also found that after activation, TM cells increase oxidative phosphorylation and aerobic glycolysis and sustain this increase to a greater extent than TN cells, suggesting that greater mitochondrial mass in TM cells not only promotes oxidative capacity, but also glycolytic capacity. We show that mitochondrial ATP is essential for the rapid induction of glycolysis in response to activation and the initiation of proliferation of both TN and TM cells. We also found that fatty acid oxidation is needed for TM cells to rapidly respond upon restimulation. Finally, we show that dissociation of the glycolysis enzyme hexokinase from mitochondria impairs proliferation and blocks the rapid induction of glycolysis upon T-cell receptor stimulation in TM cells. Our results demonstrate that greater mitochondrial mass endows TM cells with a bioenergetic advantage that underlies their ability to rapidly recall in response to reinfection.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Metabolismo Energético , Ativação Linfocitária , Camundongos , Tamanho Mitocondrial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA