Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Circulation ; 149(25): 1982-2001, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38390737

RESUMO

BACKGROUND: Reparative macrophages play a crucial role in limiting excessive fibrosis and promoting cardiac repair after myocardial infarction (MI), highlighting the significance of enhancing their reparative phenotype for wound healing. Metabolic adaptation orchestrates the phenotypic transition of macrophages; however, the precise mechanisms governing metabolic reprogramming of cardiac reparative macrophages remain poorly understood. In this study, we investigated the role of NPM1 (nucleophosmin 1) in the metabolic and phenotypic shift of cardiac macrophages in the context of MI and explored the therapeutic effect of targeting NPM1 for ischemic tissue repair. METHODS: Peripheral blood mononuclear cells were obtained from healthy individuals and patients with MI to explore NPM1 expression and its correlation with prognostic indicators. Through RNA sequencing, metabolite profiling, histology, and phenotype analyses, we investigated the role of NPM1 in postinfarct cardiac repair using macrophage-specific NPM1 knockout mice. Epigenetic experiments were conducted to study the mechanisms underlying metabolic reprogramming and phenotype transition of NPM1-deficient cardiac macrophages. The therapeutic efficacy of antisense oligonucleotide and inhibitor targeting NPM1 was then assessed in wild-type mice with MI. RESULTS: NPM1 expression was upregulated in the peripheral blood mononuclear cells from patients with MI that closely correlated with adverse prognostic indicators of MI. Macrophage-specific NPM1 deletion reduced infarct size, promoted angiogenesis, and suppressed tissue fibrosis, in turn improving cardiac function and protecting against adverse cardiac remodeling after MI. Furthermore, NPM1 deficiency boosted the reparative function of cardiac macrophages by shifting macrophage metabolism from the inflammatory glycolytic system to oxygen-driven mitochondrial energy production. The oligomeric NPM1 recruited histone demethylase KDM5b to the promoter of Tsc1 (TSC complex subunit 1), the mTOR (mechanistic target of rapamycin kinase) complex inhibitor, reduced histone H3K4me3 modification, and inhibited TSC1 expression, which then facilitated mTOR-related inflammatory glycolysis and antagonized the reparative function of cardiac macrophages. The in vivo administration of antisense oligonucleotide targeting NPM1 or oligomerization inhibitor NSC348884 substantially ameliorated tissue injury and enhanced cardiac recovery in mice after MI. CONCLUSIONS: Our findings uncover the key role of epigenetic factor NPM1 in impeding postinfarction cardiac repair by remodeling metabolism pattern and impairing the reparative function of cardiac macrophages. NPM1 may serve as a promising prognostic biomarker and a valuable therapeutic target for heart failure after MI.


Assuntos
Epigênese Genética , Macrófagos , Infarto do Miocárdio , Proteínas Nucleares , Nucleofosmina , Animais , Macrófagos/metabolismo , Humanos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/genética , Camundongos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Camundongos Knockout , Masculino , Reprogramação Celular , Feminino , Glicólise , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
2.
Nat Immunol ; 12(5): 416-24, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21441935

RESUMO

The molecular mechanisms involved in the full activation of innate immunity achieved through Toll-like receptors (TLRs) remain to be fully elucidated. In addition to their classical antigen-presenting function, major histocompatibility complex (MHC) class II molecules might mediate reverse signaling. Here we report that deficiency in MHC class II attenuated the TLR-triggered production of proinflammatory cytokines and type I interferon in macrophages and dendritic cells, which protected mice from endotoxin shock. Intracellular MHC class II molecules interacted with the tyrosine kinase Btk via the costimulatory molecule CD40 and maintained Btk activation, but cell surface MHC class II molecules did not. Then, Btk interacted with the adaptor molecules MyD88 and TRIF and thereby promoted TLR signaling. Therefore, intracellular MHC class II molecules can act as adaptors, promoting full activation of TLR-triggered innate immune responses.


Assuntos
Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Inata/imunologia , Proteínas Tirosina Quinases/metabolismo , Receptores Toll-Like/imunologia , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Tirosina Quinase da Agamaglobulinemia , Animais , Células Apresentadoras de Antígenos/enzimologia , Células Apresentadoras de Antígenos/imunologia , Antígenos CD40/imunologia , Linhagem Celular , Citocinas/sangue , Citocinas/imunologia , Ativação Enzimática , Immunoblotting , Interferon gama/sangue , Interferon gama/imunologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/imunologia , Proteínas Tirosina Quinases/imunologia , Sepse/imunologia , Organismos Livres de Patógenos Específicos
3.
J Med Virol ; 95(1): e28161, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36124363

RESUMO

Messenger RNA (mRNA) vaccines are promising alternatives to conventional vaccines in many aspects. We previously developed a lipopolyplex (LPP)-based mRNA vaccine (SW0123) that demonstrated robust immunogenicity and strong protective capacity against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection in mice and rhesus macaques. However, the immune profiles and mechanisms of pulmonary protection induced by SW0123 remain unclear. Through high-resolution single-cell analysis, we found that SW0123 vaccination effectively suppressed SARS-CoV-2-induced inflammatory responses by inhibiting the recruitment of proinflammatory macrophages and increasing the frequency of polymorphonuclear myeloid-derived suppressor cells. In addition, the apoptotic process in both lung epithelial and endothelial cells was significantly inhibited, which was proposed to be one major mechanism contributing to vaccine-induced lung protection. Cell-cell interaction in the lung compartment was also altered by vaccination. These data collectively unravel the mechanisms by which the SW0123 protects against lung damage caused by SARS-CoV-2 infection.


Assuntos
COVID-19 , Vacinas Virais , Humanos , Animais , Camundongos , Vacinas contra COVID-19 , COVID-19/prevenção & controle , SARS-CoV-2/genética , RNA Mensageiro/genética , Macaca mulatta/genética , Células Endoteliais , Transcriptoma , Vacinação , Anticorpos Neutralizantes , Anticorpos Antivirais , Glicoproteína da Espícula de Coronavírus/genética , Imunogenicidade da Vacina
4.
J Immunol ; 203(5): 1338-1347, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31331970

RESUMO

Although the inflammatory response triggered by damage-associated molecular patterns (DAMPs) in the infarcted cardiac tissues after acute myocardial infarction (MI) contributes to cardiac repair, the unrestrained inflammation induces excessive matrix degradation and myocardial fibrosis, leading to the development of adverse remodeling and cardiac dysfunction, although the molecular mechanisms that fine tune inflammation post-MI need to be fully elucidated. Protein phosphatase Mg2+/Mn2+-dependent 1L (PPM1L) is a member of the serine/threonine phosphatase family. It is originally identified as a negative regulator of stress-activated protein kinase signaling and involved in the regulation of ceramide trafficking from the endoplasmic reticulum to Golgi apparatus. However, the role of PPM1L in MI remains unknown. In this study, we found that PPM1L transgenic mice exhibited reduced infarct size, attenuated myocardial fibrosis, and improved cardiac function. PPM1L transgenic mice showed significantly lower levels of inflammatory cytokines, including IL-1ß, IL-6, TNF-α, and IL-12, in myocardial tissue. In response to DAMPs, such as HMGB1 or HSP60, released in myocardial tissue after MI, macrophages from PPM1L transgenic mice consistently produced fewer inflammatory cytokines. PPM1L-silenced macrophages showed higher levels of inflammatory cytokine production induced by DAMPs. Mechanically, PPM1L overexpression selectively inhibited the activation of NF-κB signaling in myocardial tissue post-MI and DAMP-triggered macrophages. PPM1L directly bound IKKß and then inhibited its phosphorylation and activation, leading to impaired NF-κB signaling activation and suppressed inflammatory cytokine production. Thus, our data demonstrate that PPM1L prevents excessive inflammation and cardiac dysfunction after MI, which sheds new light on the protective regulatory mechanism underlying MI.

5.
Org Biomol Chem ; 18(48): 9831-9835, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33245315

RESUMO

We have developed a copper-mediated one-pot synthesis of 2,3,5-trisubstituted pyrroles from 1,3-dicarbonyl compounds and acrylates using ammonium acetate as a nitrogen source. The reaction achieves C-C and C-N bond formation and provides an efficient approach to access highly functionalized pyrroles without further raw material preparation. This method is operationally simple, compatible with a wide range of functional groups, and provides the target products in moderate to good yields.

6.
Org Biomol Chem ; 17(20): 5148-5152, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31073576

RESUMO

We have designed a general, inexpensive, and versatile method for the synthesis of (1H-benzo[d]imidazol-2-yl)(phenyl)methanone and the formation of C-N bonds via an aromatic aldehyde and o-phenylenediamine. In the presence of N,N-dimethylformamide/sulfur, (1H-benzo[d]imidazol-2-yl)(phenyl)methanone was obtained; however, in the absence of sulfur, quinoxaline was obtained in 1,4-dioxane. A wide range of quinoxalines and (1H-benzo[d]imidazol-2-yl)(phenyl)methanones was obtained under mild conditions.

7.
Org Biomol Chem ; 17(2): 240-243, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30534708

RESUMO

Indole is a ubiquitous structural motif with important applications in many areas of chemistry. Given this, a simple and efficient Ru(ii)-catalyzed synthesis of indole via intermolecular annulation of N-aryl-2-aminopyridines and sulfoxonium ylides was proposed and accomplished. Excellent selectivity and good functional group tolerance of this transformation were observed. This protocol provides easy access to a wide variety of useful indoles in the presence of a commercially available [Ru(p-cymene)Cl2]2 catalyst. A possible mechanism for the reaction pathway was also proposed. More importantly, this reaction will offer a useful method for the construction of enantioenriched indole frameworks.

8.
Org Biomol Chem ; 16(24): 4439-4442, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29855642

RESUMO

A novel [3 + 2] cycloaddition reaction of azaoxyallyl cations and aromatic ethylenes has been developed to afford multi-substituted pyrrolidinones in moderate to good yields. This method not only further expands the synthetic utility of α-halo hydroxamates, but also provides an alternative method for the synthesis of bioactive molecules containing pyrrolidinones.

9.
Org Biomol Chem ; 15(35): 7365-7368, 2017 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-28829100

RESUMO

Copper catalyzed chemoselective cleavage of the C(CO)-C(alkyl) bond leading to C-N bond formation with chelation assistance of N-containing directing groups is described. Inexpensive Cu(ii)-acetate serves as a convenient catalyst for this transformation. This method highlights the emerging strategy to transform unactivated alkyl ketones into amides in organic synthesis and provides a new strategy for C-C bond cleavage.


Assuntos
Amidas/síntese química , Monóxido de Carbono/química , Carbono/química , Cobre/química , Cetonas/química , Amidas/química , Catálise , Estrutura Molecular
11.
J Immunol ; 195(8): 3849-57, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26363053

RESUMO

The effective recognition of viral infection and subsequent type I IFN production is essential for the host antiviral innate immune responses. The phosphorylation and activation of kinase TANK-binding kinase 1 (TBK1) plays crucial roles in the production of type I IFN mediated by TLR and retinoic acid-inducible gene I-like receptors. Type I IFN expression must be tightly regulated to prevent the development of immunopathological disorders. However, how the activated TBK1 is negatively regulated by phosphatases remains poorly understood. In this study, we identified a previously unknown role of protein phosphatase (PP)4 by acting as a TBK1 phosphatase. PP4 expression was upregulated in macrophages infected with RNA virus, vesicular stomatitis virus, and Sendai virus in vitro and in vivo. Knockdown of PP4C, the catalytic subunit of PP4, significantly increased type I IFN production in macrophages and dentritic cells triggered by TLR3/4 ligands, vesicular stomatitis virus, and Sendai virus, and thus inhibited virus replication. Similar results were also found in peritoneal macrophages with PP4C silencing in vivo and i.p. infection of RNA virus. Accordingly, ectopic expression of PP4C inhibited virus-induced type I IFN production and promoted virus replication. However, overexpression of a phosphatase-dead PP4C mutant abolished the inhibitory effects of wild-type PP4C on type I IFN production. Mechanistically, PP4 directly bound TBK1 upon virus infection, then dephosphorylated TBK1 at Ser(172) and inhibited TBK1 activation, and subsequently restrained IFN regulatory factor 3 activation, resulting in suppressed production of type I IFN and IFN-stimulated genes. Thus, serine/threonine phosphatase PP4 functions as a novel feedback negative regulator of RNA virus-triggered innate immunity.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Inata , Interferon Tipo I/imunologia , Fosfoproteínas Fosfatases/imunologia , Infecções por Respirovirus/imunologia , Infecções por Rhabdoviridae/imunologia , Vírus Sendai/fisiologia , Vesiculovirus/fisiologia , Replicação Viral/imunologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Fosforilação/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Infecções por Respirovirus/patologia , Infecções por Rhabdoviridae/patologia
12.
Proc Natl Acad Sci U S A ; 110(27): 11097-102, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23776228

RESUMO

Toll-like receptor (TLR) signaling is critical in innate response against invading pathogens. However, the molecular mechanisms for full activation of TLR-triggered innate immunity need to be fully elucidated. The broad complex tramtrack bric-a-brac/poxvirus and zinc finger (BTB/POZ) family is a class of transcription factors involved in many biological processes. However, few BTB/POZ proteins were reported to function in innate immune response. Zinc finger and BTB domain-containing 20 (ZBTB20), a member of BTB/POZ family, functions in neurogenesis and represses α-fetoprotein gene transcription in liver. However, the immunological functions of ZBTB20 remain unknown. Here, we found that myeloid cell-specific ZBTB20 KO mice were resistant to endotoxin shock and Escherichia coli-caused sepsis. ZBTB20 deficiency attenuated TLR-triggered production of proinflammatory cytokines and type I IFN in macrophages, which attributed to higher abundance of IκBα protein and impaired activity of NF-κB. Furthermore, ChIP and next generation high-throughput DNA sequencing assay showed that ZBTB20 specifically bound to IκBα gene promoter (+1 to +60 region) after TLR activation. ZBTB20 could inhibit IκBα gene transcription, govern IκBα protein expression, and then promote NF-κB activation. Therefore, transcriptional repressor ZBTB20 is needed to promote full activation of TLR signaling and TLR-triggered innate immune response by selectively suppressing the suppressor IκBα gene transcription.


Assuntos
Proteínas I-kappa B/antagonistas & inibidores , Proteínas I-kappa B/genética , Supressão Genética , Receptores Toll-Like/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/imunologia , Animais , Regulação para Baixo/genética , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/patologia , Feminino , Proteínas I-kappa B/metabolismo , Imunidade Inata/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/patologia , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica/genética , Choque Séptico/genética , Choque Séptico/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
13.
J Immunol ; 190(4): 1685-94, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23296707

RESUMO

TLRs are essential for sensing the invading pathogens and initiating protective immune responses. However, aberrant activation of TLR-triggered inflammatory innate responses leads to the inflammatory disorders and autoimmune diseases. The molecular mechanisms that fine-tune TLR responses remain to be fully elucidated. Protein tyrosine phosphatase with proline-glutamine-serine-threonine-rich motifs (PTP-PEST) has been shown to be important in cell adhesion, migration, and also T cell and B cell activation. However, the roles of PTP-PEST in TLR-triggered immune response remain unclear. In this study, we report that PTP-PEST expression was upregulated in macrophages by TLR ligands. PTP-PEST inhibited TNF-α, IL-6, and IFN-ß production in macrophages triggered by TLR3, TLR4, and TLR9. Overexpression of catalytically inactive mutants of PTP-PEST abolished the inhibitory effects, indicating that PTP-PEST inhibits TLR response in a phosphatase-dependent manner. Accordingly, PTP-PEST knockdown increased TLR3, -4, and -9-triggered proinflammatory cytokine and type I IFN production. PTP-PEST selectively inhibited TLR-induced NF-κB activation, whereas it had no substantial effect on MAPK and IFN regulatory factor 3 activation. Moreover, PTP-PEST directly interacted with IκB kinase ß (IKKß) then inhibited IKKß phosphorylation at Ser(177/181) and Tyr(188/199), and subsequently suppressed IKKß activation and kinase activity as well as downstream NF-κB activation, resulting in suppression of the TLR-triggered innate immune response. Thus, PTP-PEST functions as a feedback-negative regulator of TLR-triggered innate immune responses by selectively impairing IKKß/NF-κB activation.


Assuntos
Regulação para Baixo/imunologia , Quinase I-kappa B/antagonistas & inibidores , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 12/química , Proteína Tirosina Fosfatase não Receptora Tipo 12/fisiologia , Receptores Toll-Like/fisiologia , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/imunologia , Animais , Linhagem Celular , Células Cultivadas , Regulação para Baixo/genética , Glutamina/metabolismo , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Imunidade Inata/genética , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Prolina/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteína Tirosina Fosfatase não Receptora Tipo 12/biossíntese , Serina/metabolismo , Treonina/metabolismo , Distribuição Tecidual/genética , Distribuição Tecidual/imunologia , Receptores Toll-Like/antagonistas & inibidores , Receptores Toll-Like/genética
14.
NPJ Regen Med ; 8(1): 7, 2023 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-36774363

RESUMO

The irreversible loss of cardiomyocytes in the adult heart following cardiac injury leads to adverse cardiac remodeling and ventricular dysfunction. However, the role of B cells in cardiomyocyte proliferation and heart regeneration has not been clarified. Here, we found that the neonatal mice with B cell depletion showed markedly reduced cardiomyocyte proliferation, leading to cardiac dysfunction, fibrosis scar formation, and the complete failure of heart regeneration after apical resection. B cell depletion also significantly impaired heart regeneration and cardiac function in neonatal mice following myocardial infarction (MI). However, B cell depletion in adult mice suppressed tissue inflammation, inhibited myocardial fibrosis, and improved cardiac function after MI. Interestingly, B cell depletion partially restricted cardiomyocyte proliferation in adult mice post-MI. Single-cell RNA sequencing showed that cardiac B cells possessed a more powerful ability to inhibit inflammatory responses and enhance angiogenesis in the postnatal day 1 (P1) mice compared with P7 and adult mice. Besides, the proportion of cardioprotective B cell clusters with high expression levels of S100a6 (S100 calcium-binding protein A6) and S100a4 (S100 calcium-binding protein A4) was greatly decreased in adult heart tissues compared with neonatal mice after cardiac damage. Thus, our study discovers that cardiac B cells in neonatal mice are required for cardiomyocyte proliferation and heart regeneration, while adult B cells promote inflammation and impair cardiac function after myocardial injury.

15.
Cell Death Differ ; 30(5): 1279-1292, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36914768

RESUMO

Macrophages play a critical role in the immune homeostasis and host defense against invading pathogens. However, uncontrolled activation of inflammatory macrophages leads to tissue injury and even fuels autoimmunity. Hence the molecular mechanisms underlying macrophage activation need to be further elucidated. The effects of epigenetic modifications on the function of immune cells draw increasing attention. Here, we demonstrated that lysine-specific demethylase 5B (KDM5B), a classical transcriptional repressor in stem cell development and cancer, was required for the full activation of NF-κB signaling cascade and pro-inflammatory cytokine production in macrophages. KDM5B deficiency or inhibitor treatment protected mice from immunologic injury in both collagen-induced arthritis (CIA) model and endotoxin shock model. Genome-wide analysis of KDM5B-binding peaks identified that KDM5B was selectively recruited to the promoter of Nfkbia, the gene encoding IκBα, in activated macrophages. KDM5B mediated the H3K4me3 modification erasing and decreased chromatin accessibility of Nfkbia gene locus, coordinating the elaborate suppression of IκBα expression and the enhanced NF-κB-mediated macrophage activation. Our finding identifies the indispensable role of KDM5B in macrophage-mediated inflammatory responses and provides a candidate therapeutic target for autoimmune and inflammatory disorders.


Assuntos
Histona Desmetilases , NF-kappa B , Animais , Camundongos , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , NF-kappa B/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Regulação da Expressão Gênica , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo
16.
MedComm (2020) ; 4(3): e306, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37313329

RESUMO

Epigenetic regulation of inflammatory macrophages governs inflammation initiation and resolution in the pathogenesis of rheumatoid arthritis (RA). Nevertheless, the mechanisms underlying macrophage-mediated arthritis injuries remain largely obscure. Here, we found that increased expression of lysine acetyltransferase 2A (KAT2A) in synovial tissues was closely correlated with inflammatory joint immunopathology in both RA patients and experimental arthritis mice. Administration of MB-3, the KAT2A-specific chemical inhibitor, significantly ameliorated the synovitis and bone destruction in collagen-induced arthritis model. Both pharmacological inhibition and siRNA silencing of KAT2A, not only suppressed innate stimuli-triggered proinflammatory gene (such as Il1b and Nlrp3) transcription but also impaired NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in vivo and in vitro. Mechanistically, KAT2A facilitated macrophage glycolysis reprogramming through suppressing nuclear factor-erythroid 2-related factor 2 (NRF2) activity as well as downstream antioxidant molecules, which supported histone 3 lysine 9 acetylation (H3K9ac) and limited NRF2-mediated transcriptional repression of proinflammatory genes. Our study proves that acetyltransferase KAT2A licenses metabolic and epigenetic reprogramming for NLRP3 inflammasome activation in inflammatory macrophages, thereby targeting KAT2A represents a potential therapeutic approach for patients suffering from RA and relevant inflammatory diseases.

17.
ACS Biomater Sci Eng ; 9(1): 363-374, 2023 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-36564012

RESUMO

The large amount of reactive oxygen species (ROS) produced by high glucose metabolism in diabetic patients not only induces inflammation but also damages blood vessels, finally resulting in low limb temperature, and the high glucose environment in diabetic patients also makes them susceptible to bacterial infection. Therefore, diabetic foot ulcer (DFU) usually presents as a nonhealing wound. To efficaciously prevent and treat DFU, we proposed a near-infrared (NIR) responsive microneedle (MN) patch hierarchical microparticle (HMP)-ZnO-MN-vascular endothelial growth factor and basic fibroblast growth factor (H-Z-MN-VEGF&bFGF), which could deliver drugs to the limbs painlessly, accurately, and controllably under NIR irradiation. Therein, the hair-derived HMPs exhibited the capacity of scavenging ROS, thereby preventing damage to the blood vessels. Meanwhile, zinc oxide (ZnO) nanoparticles endowed the MN patch with excellent antibacterial activity which could be further enhanced with the photothermal effect of HMPs under NIR irradiation. Moreover, vascular endothelial growth factor and basic fibroblast growth factor could promote the angiogenesis. A series of experiments proved that the MN patch exhibited broad-spectrum antibacterial and anti-inflammatory capacities. In vivo, it obviously increased the temperature of fingertips in diabetic rats as well as promoted collagen deposition and angiogenesis during wound healing. In conclusion, this therapeutic platform provides a promising method for the prevention and treatment of DFU.


Assuntos
Diabetes Mellitus Experimental , Pé Diabético , Óxido de Zinco , Ratos , Animais , Pé Diabético/prevenção & controle , Pé Diabético/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Espécies Reativas de Oxigênio/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Óxido de Zinco/farmacologia , Óxido de Zinco/uso terapêutico , Cicatrização , Cabelo/metabolismo , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico
18.
Biochem Pharmacol ; 213: 115614, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37209857

RESUMO

Acute myocardial infarction (MI) and chemotherapeutic drug administration can induce myocardial damage and cardiomyocyte cell death, and trigger the release of damage-associated molecular patterns (DAMPs) that initiate the aseptic inflammatory response. The moderate inflammatory response is beneficial for repairing damaged myocardium, while an excessive inflammatory response exacerbates myocardial injury, promotes scar formation, and results in a poor prognosis of cardiac diseases. Immune responsive gene 1 (IRG1) is specifically highly expressed in activated macrophages and mediates the production of tricarboxylic acid (TCA) cycle metabolite itaconate. However, the role of IRG1 in the inflammation and myocardial injury of cardiac stress-related diseases remains unknown. Here, we found that IRG1 knockout mice exhibited increased cardiac tissue inflammation and infarct size, aggravated myocardial fibrosis, and impaired cardiac function after MI and in vivo doxorubicin (Dox) administration. Mechanically, IRG1 deficiency enhanced the production of IL-6 and IL-1ß by suppressing the nuclear factor red lineage 2-related factor 2 (NRF2) and activating transcription factor 3 (ATF3) pathway in cardiac macrophages. Importantly, 4-octyl itaconate (4-OI), a cell-permeable derivative of itaconate, reversed the inhibited expression of NRF2 and ATF3 caused by IRG1 deficiency. Moreover, in vivo 4-OI administration inhibited the cardiac inflammation and fibrosis, and prevented adverse ventricle remodeling in IRG1 knockout mice with MI or Dox-induced myocardial injury. Our study uncovers the critical protective role of IRG1 in suppressing inflammation and preventing cardiac dysfunction under ischemic or toxic injury conditions, providing a potential target for the treatment of myocardial injury.


Assuntos
Infarto do Miocárdio , Fator 2 Relacionado a NF-E2 , Animais , Camundongos , Doxorrubicina , Inflamação/metabolismo , Camundongos Knockout , Fator 2 Relacionado a NF-E2/metabolismo
19.
J Immunol ; 185(12): 7244-51, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21068402

RESUMO

MicroRNAs (miRNAs) are involved in the regulation of immunity, including the lymphocyte development and differentiation, and inflammatory cytokine production. Dendritic cells (DCs) play important roles in linking innate and adaptive immune responses. However, few miRNAs have been found to regulate the innate response and APC function of DCs to date. Calcium/calmodulin-dependent protein kinase II (CaMKII), a major downstream effector of calcium (Ca(2+)), has been shown to be an important regulator of the maturation and function of DCs. Our previous study showed that CaMKIIα could promote TLR-triggered production of proinflammatory cytokines and type I IFN. Inspired by the observations that dicer mutant Drosophila display defect in endogenous miRNA generation and higher CaMKII expression, we wondered whether miRNAs can regulate the innate response and APC function of DCs by targeting CaMKIIα. By predicting with software and confirming with functional experiments, we demonstrate that three members of the miRNA (miR)-148 family, miR-148a, miR-148b, and miR-152, are negative regulators of the innate response and Ag-presenting capacity of DCs. miR-148/152 expression was upregulated, whereas CaMKIIα expression was downregulated in DCs on maturation and activation induced by TLR3, TLR4, and TLR9 agonists. We showed that miR-148/152 in turn inhibited the production of cytokines including IL-12, IL-6, TNF-α, and IFN-ß upregulation of MHC class II expression and DC-initiated Ag-specific T cell proliferation by targeting CaMKIIα. Therefore, miRNA-148/152 can act as fine-tuner in regulating the innate response and Ag-presenting capacity of DCs, which may contribute to the immune homeostasis and immune regulation.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/fisiologia , MicroRNAs/imunologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/biossíntese , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Drosophila melanogaster , Camundongos , MicroRNAs/biossíntese , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
20.
Front Cardiovasc Med ; 9: 933060, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35872916

RESUMO

The mortality due to heart diseases remains highest in the world every year, with ischemic cardiomyopathy being the prime cause. The irreversible loss of cardiomyocytes following myocardial injury leads to compromised contractility of the remaining myocardium, adverse cardiac remodeling, and ultimately heart failure. The hearts of adult mammals can hardly regenerate after cardiac injury since adult cardiomyocytes exit the cell cycle. Nonetheless, the hearts of early neonatal mammals possess a stronger capacity for regeneration. To improve the prognosis of patients with heart failure and to find the effective therapeutic strategies for it, it is essential to promote endogenous regeneration of adult mammalian cardiomyocytes. Mitochondrial metabolism maintains normal physiological functions of the heart and compensates for heart failure. In recent decades, the focus is on the changes in myocardial energy metabolism, including glucose, fatty acid, and amino acid metabolism, in cardiac physiological and pathological states. In addition to being a source of energy, metabolites are becoming key regulators of gene expression and epigenetic patterns, which may affect heart regeneration. However, the myocardial energy metabolism during heart regeneration is majorly unknown. This review focuses on the role of energy metabolism in cardiac regeneration, intending to shed light on the strategies for manipulating heart regeneration and promoting heart repair after cardiac injury.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA