5 research outputs found

    Investigation of the molecular mechanisms of pathogenesis of psoriasis: participation of the glycolytic enzyme Pyruvate Kinase M2 (PKM2)

    No full text
    A psoríase é uma doença inflamatória crônica com uma elevada incidência, que afeta a pele. A patogênese da psoríase caracteriza-se pela participação de inúmeras células, incluindo os queratinócitos que são as principais células efetoras da citocina IL-17, críticas para a doença, que produzidas pelas células T. Evidências crescentes sugerem o importante papel da piruvato quinase M2 (PKM2) na regulação da resposta inflamatória, mas o mecanismo subjacente permanece obscuro. Nesse sentido, no presente estudo investigamos o papel da PKM2 no desenvolvimento da psoríase. Observamos o aumento de PKM2 em biópsia humana, em modelo de psoríase induzida por imiquimode e em modelos espontâneos K14-IL-17Aind e DC-IL-17Aind. Em adição, esse aumento observado na enzima foi predominante nos queratinócitos e isso foi associado a marcadores de ativação de queratinócitos. Utilizando o inibidor de PKM2, Shikonin (SKN), como abordagem farmacológica, observamos que o tratamento com esse composto foi capaz de reverter a psoríase experimental e a reduzir marcadores associados a doença como: K17, LCN2, TNF-?, KC, S100A8, S100A9, IL-6 e IL-17A. Associado a isso, observamos a redução na frequência de células T (?? e ??) produtoras de IL-17 e do número de neutrófilos na pele em modelo de imiquimode após inibição da PKM2. O SKN, também, reduziu o número de neutrófilos no modelo DC-IL-17Aind. Em nosso próximo passo, observamos que queratinócitos HACAT estimulados com IL-17A apresentou um aumento da expressão de PKM2 e que a sua inibição foi associada a redução da ativação de queratinócitos e de mediadores inflamatórios como a IL-8. Além disso, a deleção da PKM2, utilizando a tecnologia CRISPR/Cas9, reduziu a expressão do receptor de IL-17. Por fim, o desenvolvimento da psoríase por imiquimode foi atenuada em animais deficientes para PKM2 em queratinócitos (K14-PKM2fl/+), no qual foi observado a redução de neutrófilos na pele e, além disso, evidenciamos a redução da expressão de IL-17A nesses animais. O conjunto de resultados apresentados nesse trabalho demonstram que a PKM2 apresenta um papel crítico no desenvolvimento da psoríase e que a ativação do receptor de IL-17 promove um aumento da PKM2 em queratinócitos e esta contribui para ativação de mediadores que é responsável diretamente para o desenvolvimento da psoríase. Esses resultados, ainda, sugerem a PKM2 como um biomarcador para diagnóstico da psoríase e consequentemente, um potencial alvo terapêutico para tratamento dessa doença e outras doenças inflamatórias.Psoriasis is a chronic inflammatory skin disease with high incidence in the global population. The pathogenesis of psoriasis is characterized by involvement of many cells, including keratinocytes that are targets for IL-17-producing T cells. Evidences suggests a critical role of pyruvate kinase M2 (PKM2) in inflammatory response, but the underlying mechanism remains unclear. In this context, here we investigated the role of PKM2 in the development of psoriasis. We observed overexpression of PKM2 in psoriatic human skin, imiquimod-induced psoriasis and spontaneous K14-IL-17Aind and DC-IL-17Aind models. In addition, the overexpression of this enzyme was observed in keratinocytes associated with keratinocytes activation markers. Using the PKM2 inhibitor, Shikonin (SKN), as a pharmacological approach, we observed that the treatment with this compound was able to reduce experimental psoriasis and disease-associated markers such as K17, LCN2, TNF-?, KC, S100A8, S100A9, IL-6 and IL-17A. Moreover, we observed reduction of frequency of IL-17-producing T cells (?? and ??) and the number of neutrophils in the skin after imiquimod application plus inhibition of PKM2. SKN, also, reduced the number of neutrophils in the DC-IL-17Aind model. In our next step, we observed overexpression of PKM2 in human keratinocytes HACAT stimulated with IL-17A and that its inhibition was associated with less keratinocytes activation and inflammatory mediators such as IL-8. In addition, deletion of PKM2, using CRISPR/Cas9 technology, reduced IL-17 receptor expression. Finally, the development of imiquimod-induced psoriasis was attenuated in PKM2-deficient mice in keratinocytes (K14-PKM2f/+), with reduction in the number of neutrophils in the skin. In addition, we evidenced the reduction of IL-17A expression these animals. Taken together, these results demonstrate that PKM2 plays a critical role in the development of psoriasis and that IL-17 receptor activation promotes an increase of PKM2 in keratinocytes and this contributes to the release of mediators that is directly responsible for development of psoriasis. These results, suggest PKM2 as a biomarker for the diagnosis of psoriasis and consequently a potential therapeutic target for the treatment of this disease and other inflammatory diseases

    Investigation of the molecular mechanisms of pathogenesis of psoriasis: participation of the glycolytic enzyme Pyruvate Kinase M2 (PKM2)

    No full text
    A psoríase é uma doença inflamatória crônica com uma elevada incidência, que afeta a pele. A patogênese da psoríase caracteriza-se pela participação de inúmeras células, incluindo os queratinócitos que são as principais células efetoras da citocina IL-17, críticas para a doença, que produzidas pelas células T. Evidências crescentes sugerem o importante papel da piruvato quinase M2 (PKM2) na regulação da resposta inflamatória, mas o mecanismo subjacente permanece obscuro. Nesse sentido, no presente estudo investigamos o papel da PKM2 no desenvolvimento da psoríase. Observamos o aumento de PKM2 em biópsia humana, em modelo de psoríase induzida por imiquimode e em modelos espontâneos K14-IL-17Aind e DC-IL-17Aind. Em adição, esse aumento observado na enzima foi predominante nos queratinócitos e isso foi associado a marcadores de ativação de queratinócitos. Utilizando o inibidor de PKM2, Shikonin (SKN), como abordagem farmacológica, observamos que o tratamento com esse composto foi capaz de reverter a psoríase experimental e a reduzir marcadores associados a doença como: K17, LCN2, TNF-?, KC, S100A8, S100A9, IL-6 e IL-17A. Associado a isso, observamos a redução na frequência de células T (?? e ??) produtoras de IL-17 e do número de neutrófilos na pele em modelo de imiquimode após inibição da PKM2. O SKN, também, reduziu o número de neutrófilos no modelo DC-IL-17Aind. Em nosso próximo passo, observamos que queratinócitos HACAT estimulados com IL-17A apresentou um aumento da expressão de PKM2 e que a sua inibição foi associada a redução da ativação de queratinócitos e de mediadores inflamatórios como a IL-8. Além disso, a deleção da PKM2, utilizando a tecnologia CRISPR/Cas9, reduziu a expressão do receptor de IL-17. Por fim, o desenvolvimento da psoríase por imiquimode foi atenuada em animais deficientes para PKM2 em queratinócitos (K14-PKM2fl/+), no qual foi observado a redução de neutrófilos na pele e, além disso, evidenciamos a redução da expressão de IL-17A nesses animais. O conjunto de resultados apresentados nesse trabalho demonstram que a PKM2 apresenta um papel crítico no desenvolvimento da psoríase e que a ativação do receptor de IL-17 promove um aumento da PKM2 em queratinócitos e esta contribui para ativação de mediadores que é responsável diretamente para o desenvolvimento da psoríase. Esses resultados, ainda, sugerem a PKM2 como um biomarcador para diagnóstico da psoríase e consequentemente, um potencial alvo terapêutico para tratamento dessa doença e outras doenças inflamatórias.Psoriasis is a chronic inflammatory skin disease with high incidence in the global population. The pathogenesis of psoriasis is characterized by involvement of many cells, including keratinocytes that are targets for IL-17-producing T cells. Evidences suggests a critical role of pyruvate kinase M2 (PKM2) in inflammatory response, but the underlying mechanism remains unclear. In this context, here we investigated the role of PKM2 in the development of psoriasis. We observed overexpression of PKM2 in psoriatic human skin, imiquimod-induced psoriasis and spontaneous K14-IL-17Aind and DC-IL-17Aind models. In addition, the overexpression of this enzyme was observed in keratinocytes associated with keratinocytes activation markers. Using the PKM2 inhibitor, Shikonin (SKN), as a pharmacological approach, we observed that the treatment with this compound was able to reduce experimental psoriasis and disease-associated markers such as K17, LCN2, TNF-?, KC, S100A8, S100A9, IL-6 and IL-17A. Moreover, we observed reduction of frequency of IL-17-producing T cells (?? and ??) and the number of neutrophils in the skin after imiquimod application plus inhibition of PKM2. SKN, also, reduced the number of neutrophils in the DC-IL-17Aind model. In our next step, we observed overexpression of PKM2 in human keratinocytes HACAT stimulated with IL-17A and that its inhibition was associated with less keratinocytes activation and inflammatory mediators such as IL-8. In addition, deletion of PKM2, using CRISPR/Cas9 technology, reduced IL-17 receptor expression. Finally, the development of imiquimod-induced psoriasis was attenuated in PKM2-deficient mice in keratinocytes (K14-PKM2f/+), with reduction in the number of neutrophils in the skin. In addition, we evidenced the reduction of IL-17A expression these animals. Taken together, these results demonstrate that PKM2 plays a critical role in the development of psoriasis and that IL-17 receptor activation promotes an increase of PKM2 in keratinocytes and this contributes to the release of mediators that is directly responsible for development of psoriasis. These results, suggest PKM2 as a biomarker for the diagnosis of psoriasis and consequently a potential therapeutic target for the treatment of this disease and other inflammatory diseases

    S100A9 Drives the Chronification of Psoriasiform Inflammation by Inducing IL-23/Type 3 Immunity

    Full text link
    Psoriasis is a chronic inflammatory skin disorder driven by the IL-23/type 3 immune response. However, molecular mechanisms sustaining the chronicity of inflammation and psoriatic lesions remain elusive. Combining systematic analyses of several transcriptomic datasets, we delineated gene signatures across human psoriatic skin, identifying S100A9 as one of the most up-regulated genes, which was confirmed in lesioned skin from patients with psoriasis and preclinical psoriasiform skin inflammation models. Genetic ablation or pharmacologic inhibition of S100A9 alleviated Aldara-induced skin inflammation. By single-cell mapping of human psoriatic skin and bone marrow chimeric mice experiments, we identified keratinocytes as the major source of S100A9. Mechanistically, S100A9 induced IL-23 production by dendritic cells, driving the IL-23/type 3 immunity in psoriasiform skin inflammation. In addition, the cutaneous IL-23/IL-17 axis induced epidermal S100A9 expression in human and experimental psoriasis. Thus, we showed an autoregulatory circuit between keratinocyte-derived S100A9 and IL-23/type 3 immunity during psoriasiform inflammation, identifying a crucial function of S100A9 in the chronification of psoriasis

    The metabolic function of pyruvate kinase M2 regulates reactive oxygen species production and microbial killing by neutrophils

    No full text
    Abstract Neutrophils rely predominantly on glycolytic metabolism for their biological functions, including reactive oxygen species (ROS) production. Although pyruvate kinase M2 (PKM2) is a glycolytic enzyme known to be involved in metabolic reprogramming and gene transcription in many immune cell types, its role in neutrophils remains poorly understood. Here, we report that PKM2 regulates ROS production and microbial killing by neutrophils. Zymosan-activated neutrophils showed increased cytoplasmic expression of PKM2. Pharmacological inhibition or genetic deficiency of PKM2 in neutrophils reduced ROS production and Staphylococcus aureus killing in vitro. In addition, this also resulted in phosphoenolpyruvate (PEP) accumulation and decreased dihydroxyacetone phosphate (DHAP) production, which is required for de novo synthesis of diacylglycerol (DAG) from glycolysis. In vivo, PKM2 deficiency in myeloid cells impaired the control of infection with Staphylococcus aureus. Our results fill the gap in the current knowledge of the importance of lower glycolysis for ROS production in neutrophils, highlighting the role of PKM2 in regulating the DHAP and DAG synthesis to promote ROS production in neutrophils

    Sepsis expands a CD39+ plasmablast population that promotes immunosuppression via adenosine-mediated inhibition of macrophage antimicrobial activity

    No full text
    Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease
    corecore