Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 990874, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36081513

RESUMO

Akt is a PI3K-activated serine-threonine kinase that exists in three distinct isoforms. Akt's expression in most immune cells, either at baseline or upon activation, reflects its importance in the immune system. While Akt is most highly expressed in innate immune cells, it plays crucial roles in both innate and adaptive immune cell development and/or effector functions. In this review, we explore what's known about the role of Akt in innate and adaptive immune cells. Wherever possible, we discuss the overlapping and distinct role of the three Akt isoforms, namely Akt1, Akt2, and Akt3, in immune cells.


Assuntos
Sistema Imunitário , Proteínas Proto-Oncogênicas c-akt , Diferenciação Celular , Sistema Imunitário/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
Bioconjug Chem ; 32(8): 1629-1640, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34165285

RESUMO

Macrophage-mediated inflammation drives autoimmune and chronic inflammatory diseases. Treatment with anti-inflammatory agents can be an effective strategy to reduce this inflammation; however, high concentrations of these agents can have immune-dampening and other serious side effects. Synergistic combination of anti-inflammatory agents can mitigate dosing by requiring less drug. Multiple anti-inflammatory agents were evaluated in combination for synergistic inhibition of macrophage inflammation. The most potent synergy was observed between dexamethasone (DXM) and fumaric acid esters (e.g., monomethyl fumarate (MMF)). Furthermore, this combination was found to synergistically inhibit inflammatory nuclear factor κB (NF-κB) transcription factor activity. The optimal ratio for synergy was determined to be 1:1, and DXM and MMF were conjugated by esterification at this molar ratio. The DXM-MMF conjugate displayed improved inhibition of inflammation over the unconjugated combination in both murine and human macrophages. In the treatment of human donor monocyte-derived macrophages, the combination of DXM and MMF significantly inhibited inflammatory gene expression downstream of NF-κB and overall performed better than either agent alone. Further, the DXM-MMF conjugate significantly inhibited expression of NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome-associated genes. The potent anti-inflammatory activity of the DXM-MMF conjugate in human macrophages indicates that it may have benefits in the treatment of autoimmune and inflammatory diseases.


Assuntos
Anti-Inflamatórios/uso terapêutico , Dexametasona/uso terapêutico , Fumaratos/uso terapêutico , Inflamação/tratamento farmacológico , Macrófagos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/química , Citocinas/genética , Citocinas/metabolismo , Dexametasona/química , Sinergismo Farmacológico , Fumaratos/química , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos/patologia , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Células RAW 264.7
3.
Am J Pathol ; 191(9): 1537-1549, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34139193

RESUMO

Epithelial barrier impairment is a hallmark of several pathologic processes in the gut, including inflammatory bowel diseases. Several intracellular signals prevent apoptosis in intestinal epithelial cells. Herein, we show that in colonocytes, rictor/mammalian target of rapamycin complex 2 (mTORC2) signaling is a prosurvival stimulus. Mechanistically, mTORC2 activates Akt, which, in turn, inhibits apoptosis by phosphorylating B-cell lymphoma 2 (BCL2) associated agonist of cell death (Bad) and preventing caspase-3 activation. Nevertheless, during inflammation, rictor/mTORC2 signaling declines and Akt activity is reduced. Consequently, active caspase-3 increases in surface colonocytes undergoing apoptosis/anoikis and causes epithelial barrier breakdown. Likewise, Rictor ablation in intestinal epithelial cells interrupts mTORC2/Akt signaling and increases apoptosis/anoikis of surface colonocytes without affecting the crypt architecture. The increase in epithelial permeability induced by Rictor ablation produces a mild inflammatory response in the colonic mucosa, but minimally affects the development/establishment of colitis. The data identify a previously unknown mechanism by which rictor/mTORC2 signaling regulates apoptosis/anoikis in intestinal epithelial cells during colitis and clarify its role in the maintenance of the intestinal epithelial barrier.


Assuntos
Apoptose/fisiologia , Colite/patologia , Células Epiteliais/metabolismo , Mucosa Intestinal/patologia , Proteína Companheira de mTOR Insensível à Rapamicina/metabolismo , Animais , Colite/metabolismo , Células Epiteliais/patologia , Mucosa Intestinal/metabolismo , Camundongos , Transdução de Sinais/fisiologia
4.
Front Immunol ; 11: 597959, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329591

RESUMO

CD38 is a molecule that can act as an enzyme, with NAD-depleting and intracellular signaling activity, or as a receptor with adhesive functions. CD38 can be found expressed either on the cell surface, where it may face the extracellular milieu or the cytosol, or in intracellular compartments, such as endoplasmic reticulum, nuclear membrane, and mitochondria. The main expression of CD38 is observed in hematopoietic cells, with some cell-type specific differences between mouse and human. The role of CD38 in immune cells ranges from modulating cell differentiation to effector functions during inflammation, where CD38 may regulate cell recruitment, cytokine release, and NAD availability. In line with a role in inflammation, CD38 appears to also play a critical role in inflammatory processes during autoimmunity, although whether CD38 has pathogenic or regulatory effects varies depending on the disease, immune cell, or animal model analyzed. Given the complexity of the physiology of CD38 it has been difficult to completely understand the biology of this molecule during autoimmune inflammation. In this review, we analyze current knowledge and controversies regarding the role of CD38 during inflammation and autoimmunity and novel molecular tools that may clarify current gaps in the field.


Assuntos
ADP-Ribosil Ciclase 1/metabolismo , Autoimunidade , Imunomodulação , Inflamação/etiologia , Inflamação/metabolismo , Glicoproteínas de Membrana/metabolismo , ADP-Ribosil Ciclase 1/química , ADP-Ribosil Ciclase 1/genética , Animais , Apresentação de Antígeno/imunologia , Biomarcadores , Movimento Celular , Citocinas/metabolismo , Suscetibilidade a Doenças , Regulação da Expressão Gênica , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Fagocitose , Transporte Proteico
5.
Nat Metab ; 2(7): 635-647, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32694789

RESUMO

T cells undergo metabolic rewiring to meet their bioenergetic, biosynthetic and redox demands following antigen stimulation. To fulfil these needs, effector T cells must adapt to fluctuations in environmental nutrient levels at sites of infection and inflammation. Here, we show that effector T cells can utilize inosine, as an alternative substrate, to support cell growth and function in the absence of glucose in vitro. T cells metabolize inosine into hypoxanthine and phosphorylated ribose by purine nucleoside phosphorylase. We demonstrate that the ribose subunit of inosine can enter into central metabolic pathways to provide ATP and biosynthetic precursors, and that cancer cells display diverse capacities to utilize inosine as a carbon source. Moreover, the supplementation with inosine enhances the anti-tumour efficacy of immune checkpoint blockade and adoptive T-cell transfer in solid tumours that are defective in metabolizing inosine, reflecting the capability of inosine to relieve tumour-imposed metabolic restrictions on T cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Carbono/metabolismo , Glucose/deficiência , Inosina/metabolismo , Transferência Adotiva , Animais , Linhagem Celular Tumoral , Células HeLa , Humanos , Hipoxantina/metabolismo , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nutrientes , Purina-Núcleosídeo Fosforilase/metabolismo , Ribose/metabolismo
6.
J Clin Invest ; 130(4): 1683-1698, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32091410

RESUMO

Protein arginine methyltransferase 5 (PRMT5) catalyzes symmetric dimethylation (SDM) of arginine, a posttranslational modification involved in oncogenesis and embryonic development. However, the role and mechanisms by which PRMT5 modulates Th cell polarization and autoimmune disease have not yet been elucidated. Here, we found that PRMT5 promoted SREBP1 SDM and the induction of cholesterol biosynthetic pathway enzymes that produce retinoid-related orphan receptor (ROR) agonists that activate RORγt. Specific loss of PRMT5 in the CD4+ Th cell compartment suppressed Th17 differentiation and protected mice from developing experimental autoimmune encephalomyelitis (EAE). We also found that PRMT5 controlled thymic and peripheral homeostasis in the CD4+ Th cell life cycle and invariant NK (iNK) T cell development and CD8+ T cell maintenance. This work demonstrates that PRMT5 expression in recently activated T cells is necessary for the cholesterol biosynthesis metabolic gene expression program that generates RORγt agonistic activity and promotes Th17 differentiation and EAE. These results point to Th PRMT5 and its downstream cholesterol biosynthesis pathway as promising therapeutic targets in Th17-mediated diseases.


Assuntos
Autoimunidade , Diferenciação Celular/imunologia , Colesterol/imunologia , Encefalomielite Autoimune Experimental/imunologia , Proteína-Arginina N-Metiltransferases/imunologia , Células Th17/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular/genética , Colesterol/genética , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Camundongos , Camundongos Transgênicos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/patologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Proteína-Arginina N-Metiltransferases/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/imunologia , Células Th17/patologia
7.
J Leukoc Biol ; 105(5): 973-981, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30821871

RESUMO

Connections established between cytoskeleton and plasma membrane are essential in cellular processes such as cell migration, vesicular trafficking, and cytokinesis. Class I myosins are motor proteins linking the actin-cytoskeleton with membrane phospholipids. Previous studies have implicated these molecules in cell functions including endocytosis, exocytosis, release of extracellular vesicles and the regulation of cell shape and membrane elasticity. In immune cells, those proteins also are involved in the formation and maintenance of immunological synapse-related signaling. Thus, these proteins are master regulators of actin cytoskeleton dynamics in different scenarios. Although the localization of class I myosins has been described in vertebrates, their functions, regulation, and mechanical properties are not very well understood. In this review, we focused on and summarized the current understanding of class I myosins in vertebrates with particular emphasis in leukocytes.


Assuntos
Citoesqueleto de Actina/imunologia , Sistema Imunitário/citologia , Sinapses Imunológicas/metabolismo , Leucócitos/imunologia , Miosina Tipo I/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/ultraestrutura , Animais , Membrana Celular/imunologia , Membrana Celular/metabolismo , Movimento Celular , Citocinese/genética , Citocinese/imunologia , Endocitose , Exocitose , Vesículas Extracelulares/química , Vesículas Extracelulares/imunologia , Regulação da Expressão Gênica , Humanos , Leucócitos/metabolismo , Leucócitos/ultraestrutura , Mecanotransdução Celular/imunologia , Miosina Tipo I/classificação , Miosina Tipo I/imunologia , Fosfolipídeos/imunologia , Fosfolipídeos/metabolismo , Isoformas de Proteínas/classificação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia
8.
Front Immunol ; 9: 3118, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687322

RESUMO

Intestinal macrophages are highly mobile cells with extraordinary plasticity and actively contribute to cytokine-mediated epithelial cell damage. The mechanisms triggering macrophage polarization into a proinflammatory phenotype are unknown. Here, we report that during inflammation macrophages enhance its intercellular adhesion properties in order to acquire a M1-phenotype. Using in vitro and in vivo models we demonstrate that intercellular adhesion is mediated by integrin-αVß3 and relies in the presence of the unconventional class I myosin 1F (Myo1F). Intercellular adhesion mediated by αVß3 stimulates M1-like phenotype in macrophages through hyperactivation of STAT1 and STAT3 downstream of ILK/Akt/mTOR signaling. Inhibition of integrin-αVß3, Akt/mTOR, or lack of Myo1F attenuated the commitment of macrophages into a pro-inflammatory phenotype. In a model of colitis, Myo1F deficiency strongly reduces the secretion of proinflammatory cytokines, decreases epithelial damage, ameliorates disease activity, and enhances tissue repair. Together our findings uncover an unknown role for Myo1F as part of the machinery that regulates intercellular adhesion and polarization in macrophages.


Assuntos
Colite Ulcerativa/imunologia , Integrina alfaVbeta3/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Miosina Tipo I/metabolismo , Animais , Linhagem Celular Tumoral , Colite Ulcerativa/induzido quimicamente , Citoesqueleto/imunologia , Citoesqueleto/metabolismo , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Humanos , Integrina alfaVbeta3/imunologia , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina Tipo I/genética , Miosina Tipo I/imunologia , Cultura Primária de Células , Células RAW 264.7
9.
Vector Borne Zoonotic Dis ; 15(7): 449-52, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26186518

RESUMO

Previously, we identified five Leishmania mexicana antigens reacting with antibodies from cutaneous leishmaniasis patients, designated on the basis of their molecular weights as p26 (pI 7.8), p27 (pI 8.1), p28 (pI 8.6), p29 (pI 8.5), and p31 (pI 9.0). Among these antigens, p29 was most strongly recognized by the antibodies. Thereafter, p29 was identified as elongation factor-1α (EF-1α) of Leishmania mexicana through mass spectrometry analysis and western blot using a commercial antibody that reacted with EF-1α from different species. Our results showed that the p29 antigen of Leishmania mexicana is EF-1α.


Assuntos
Antígenos de Protozoários/imunologia , Leishmania mexicana/imunologia , Leishmaniose Cutânea/parasitologia , Fator 1 de Elongação de Peptídeos/imunologia , Animais , Proteômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...