Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Total Environ ; 912: 168924, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38036146

RESUMO

The global prevalence of Neurological disorders has increased alarmingly in response to environmental and lifestyle changes. Atrazine (ATZ) is a difficult to degrade soil and water pollutant with well-known neurotoxicity. Melatonin (MT), an antioxidant with chemoprotective properties, has a potential therapeutic effect on cerebellar damage caused by ATZ exposure. The aim of this study was to explore the effects and underlying mechanisms of MT on the cerebellar inflammatory response and pyroptosis induced by ATZ exposure. In this study, C57BL/6J mice were treated with ATZ (170 mg/kg BW/day) and MT (5 mg/kg BW/day) for 28 days. Our results revealed that MT alleviated the histopathological changes, ultrastructural damage, oxidative stress and decrease of mitochondrial membrane potential (ΔΨm) in the cerebellum induced by ATZ exposure. ATZ exposure damaged the mitochondria leading to release of mitochondrial DNA (mtDNA) to the cytoplasm, MT activated the cyclic GMP-AMP synthetase interferon gene stimulator (cGAS-STING) axis to alleviate inflammation and pyroptosis caused by ATZ exposure. In general, our study provided new evidence that the cGAS-STING-NLRP3 axis plays an important role in the treatment of ATZ-induced cerebellar injury by MT.


Assuntos
Atrazina , Melatonina , Nucleotídeos Cíclicos , Animais , Camundongos , Atrazina/toxicidade , Atrazina/metabolismo , Melatonina/metabolismo , Piroptose , Interferons/metabolismo , Interferons/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Camundongos Endogâmicos C57BL , Mitocôndrias , DNA Mitocondrial , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia
2.
Antivir Ther ; 28(6): 13596535231219639, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38037795

RESUMO

BACKGROUND: Previously, we have demonstrated that Apolipoprotein A-I (ApoA-I) could inhibit the secretion of Hepatitis B virus (HBV), suggesting that stimulation of ApoA-I may block particle production. In the present study, we evaluated the anti-HBV effect of RVX-208, a small-molecule stimulator of ApoA-I gene expression. METHODS: RVX-208 was used to treat HepG2.2.15 cell, a HepG2 derived cell line stably producing HBV virus. Real-time PCR was performed to examine the HBV DNA levels. Magnetic particles, which were coated with anti-HBS or anti-HBE antibody, were used to examine the HBsAg and HBeAg levels in the supernatant of cultured HepG2.2.15 cells in combination with the enzyme conjugates that were prepared with horseradish peroxidase labelled anti-HBS or anti-HBE antibody in a double antibody sandwich manner. RNA-seq, immunoblots and real-time PCR were used to analyze the functional mechanism of RVX-208. RESULTS: RVX-208 could elevate the ApoA-I protein levels in HepG2.2.15 cells. In the meantime, RVX-208 significantly repressed HBV DNA, HBsAg and HBeAg levels in the supernatants of HepG2.2.15 cells. RNA-seq data revealed that RVX-208 treatment not only affected the cholesterol metabolism, which is closely related to ApoA-I, but also regulated signalling pathways that are associated with antiviral immune response. Moreover, mechanistic studies demonstrated that RVX-208 could activate cGAS-STING pathway and upregulate the transcription of a series of interferons, pro-inflammatory cytokines and chemokines with antiviral potential that are at the downstream of cGAS-STING pathway. CONCLUSION: Our study demonstrated that RVX-208, an inducer of ApoA-I, could suppress HBV particle production through activation of cGAS-STING pathway.


Assuntos
Apolipoproteína A-I , Vírus da Hepatite B , Humanos , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Antígenos de Superfície da Hepatite B , DNA Viral , Antígenos E da Hepatite B , Células Hep G2 , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia
3.
Leukemia ; 37(12): 2457-2467, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37816954

RESUMO

Somatic loss-of-function mutations of the dioxygenase Ten-eleven translocation-2 (TET2) occur frequently in individuals with clonal hematopoiesis (CH) and acute myeloid leukemia (AML). These common hematopoietic disorders can be recapitulated in mouse models. However, the underlying mechanisms by which the deficiency in TET2 promotes these disorders remain unclear. Here we show that the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway is activated to mediate the effect of TET2 deficiency in dysregulated hematopoiesis in mouse models. DNA damage arising in Tet2-deficient hematopoietic stem/progenitor cells (HSPCs) leads to activation of the cGAS-STING pathway which in turn promotes the enhanced self-renewal and development of CH. Notably, both pharmacological inhibition and genetic deletion of STING suppresses Tet2 mutation-induced aberrant hematopoiesis. In patient-derived xenograft (PDX) models, STING inhibition specifically attenuates the proliferation of leukemia cells from TET2-mutated individuals. These observations suggest that the development of CH associated with TET2 mutations is powered through chronic inflammation dependent on the activated cGAS-STING pathway and that STING may represent a potential target for intervention of relevant hematopoietic diseases.


Assuntos
Dioxigenases , Doenças Hematológicas , Camundongos , Animais , Humanos , Transformação Celular Neoplásica/genética , Translocação Genética , Hematopoese/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/farmacologia , Células-Tronco/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/genética
4.
Biochim Biophys Acta Rev Cancer ; 1878(6): 188983, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37717857

RESUMO

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway is a key pathway through which the host regulates immune responses by recognizing cytoplasmic double-stranded DNA of abnormal origin, and it plays an important role in tumor growth as well as metastasis, with relevant molecular details constantly being explored and updated. The significant immunomodulatory effects make STING an attractive target for cancer immunotherapy, and STING agonists have been receiving great attention for their development and clinical translation. Despite exciting results in preclinical work, the application of STING agonists to cancer therapy remains challenging due to their poor pharmacokinetic and physicochemical properties, as well as toxic side effects they produce. Here, we summarize the dichotomous role of cGAS-STING in cancer and discuss the limitations of cancer immunotherapy based on STING activation as well as feasible strategies to overcome them to achieve tumor regression.


Assuntos
Neoplasias , Humanos , Neoplasias/metabolismo , Transdução de Sinais , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , DNA
5.
Phytomedicine ; 118: 154961, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37453191

RESUMO

BACKGROUND: Liver diseases have a negative impact on global health and are a leading cause of death worldwide. Chlorogenic acids (CGAs), a family of esters formed between certain trans-cinnamic acids and quinic acid, are natural polyphenols abundant in coffee, tea, and a variety of traditional Chinese medicines (TCMs). They are reported to have good hepatoprotective effects against various liver diseases. PURPOSE: This review aims to analyze the available literature on the hepatoprotective effect of CGAs, with particular emphasis on their mechanisms. METHODS: Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. PubMed and Web of Science databases were adopted to retrieve all relevant literature on CGAs for liver disease from 2013 to March 2023. RESULTS: Research has indicated that CGAs play a crucial role in improving different types of liver diseases, including drug-induced liver injury (DILI), alcoholic liver disease (ALD), metabolic (dysfunction)-associated fatty liver disease (MAFLD), cholestatic liver disease (CLD), liver fibrosis, and liver cancer. CGAs display remarkable antioxidant and anti-inflammatory effects by activating erythroid 2-related factor 2 (Nrf2) and inhibiting toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling pathways. Some important molecules such as AMP-activated protein kinase (AMPK) and extracellular signal-regulated kinases 1 and 2 (ERK1/2), and other key physiological processes like intestinal barrier and gut microbiota have also been discovered to participate in CGAs-provided amelioration on various liver diseases. CONCLUSION: In this review, different studies indicate that CGAs have an excellent protective effect against various liver diseases associated with various signaling pathways.


Assuntos
Ácido Clorogênico , Hepatopatias Alcoólicas , Humanos , Ácido Clorogênico/farmacologia , Polifenóis/farmacologia , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Hepatopatias Alcoólicas/metabolismo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , Fígado
6.
Nano Lett ; 23(11): 5083-5091, 2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37220198

RESUMO

Pyroptosis is a proinflammatory form of programmed cell death that results in the release of cellular contents and activation of immune responses. However, GSDME (a pyroptosis-executed protein) is suppressed in many cancers. Herein, we constructed a nanoliposome (GM@LR) for codelivering the GSDME-expressing plasmid and manganese carbonyl (MnCO) into TNBC cells. MnCO generated Mn2+ and carbon monoxide (CO) in the presence of H2O2. The CO-activated caspase-3, which cleaved the expressed GSDME, converting apoptosis to pyroptosis in 4T1 cells. In addition, Mn2+ promoted maturation of dendritic cells (DCs) by the activation of STING signaling pathway. The increased proportion of intratumoral mature DCs brought about massive infiltration of cytotoxic lymphocytes, leading to a robust immune response. Besides, Mn2+ could be applied for magnetic resonance imaging (MRI)-guided metastasis detection. Taken together, our study showed that GM@LR nanodrug could effectively inhibit tumor growth via pyroptosis and STING activation combined immunotherapy.


Assuntos
Nanopartículas , Neoplasias de Mama Triplo Negativas , Linhagem Celular Tumoral , Peróxido de Hidrogênio/farmacologia , Nanopartículas/uso terapêutico , Nucleotidiltransferases/farmacologia , Piroptose , Feminino , Animais , Camundongos
7.
J Control Release ; 358: 190-203, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37116543

RESUMO

At present, the most widely used aluminum adjuvants have poor ability to induce effective Th1 type immune responses. Existing evidence suggests that manganese is a potential metal adjuvant by activating cyclic guanosine phospho-adenosine synthase (cGAS)-interferon gene stimulator protein (STING) signaling pathway to enhance humoral and cellular immune response. Hence, the effective modulation of metal components is expected to be a new strategy to improve the efficiency of vaccine immunization. Here, we constructed a manganese and aluminum dual-adjuvant antigen co-delivery system (MnO2-Al-OVA) to enhance the immune responses of subunit vaccines. Namely, the aluminum hydroxide was first fused on the surface of the pre-prepared MnO2 nanoparticles, which were synthesized by a simple redox reaction with potassium permanganate (KMnO4) and oleic acid (OA). The engineered MnO2-Al-OVA could remarkably promote cellular internalization and maturation of dendritic cells. After subcutaneous vaccination, MnO2-Al-OVA rapidly migrated into the lymph nodes (LNs) and efficiently activate the cGAS-STING pathway, greatly induced humoral and cellular immune responses. Of note, our findings underscore the importance of coordination manganese adjuvants in vaccine design by promoting the activation of the cGAS-STING-IFN-I pathway. With a good safety profile and facile preparation process, this dual-adjuvant antigen co-delivery nanovaccine has great potential for clinical translation prospects.


Assuntos
Alumínio , Nanopartículas , Alumínio/farmacologia , Manganês , Compostos de Manganês/farmacologia , Óxidos , Adjuvantes Imunológicos , Imunidade Celular , Antígenos , Vacinas de Subunidades Antigênicas , Nucleotidiltransferases/farmacologia , Células Dendríticas , Imunidade Humoral
8.
BMC Pharmacol Toxicol ; 24(1): 19, 2023 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-36964634

RESUMO

BACKGROUND: The severe unfavorable effects of doxorubicin on the heart restrict its clinical usage. Numerous investigations document that cyclic GMP-AMP synthase (cGAS) activator of interferon genes (STING) cascade influences inflammation along with the immune response in a variety of diseases. The pathophysiological function of the cGAS-STING cascade in Doxorubicin-induced cardiomyopathy (DIC) is, nevertheless, unknown. METHODS: In vivo, cardiotoxicity was triggered by a single dose of intra-peritoneal inoculation of doxorubicin (15 mg/kg) in wild-type C57BL/6J mice and STING knockdown animals. Adeno-associated virus 9 (AAV9) was utilized to silence STING. qPCR along with Western blotting were adopted to assess alterations in the cGAS/STING cascade. To assess cardiac function, we employed echocardiography coupled with histology, as well as molecular phenotyping. In vitro, HL-1 cardiomyocytes were introduced as test models. RESULTS: In wild type mice, doxorubicin stimulation significantly activated the cGAS/STING pathway. STING silencing increased rate of survival along with heart function in mice, as well as diminished myocardial inflammatory cytokines along with apoptosis. These observations were also confirmed by utilizing siRNA of STING in vitro studies. CONCLUSION: This research premise established that STING inhibition could alleviate Dox-triggered cardiotoxicity in mice. As a result, preventing DIC by repressing STING in cardiomyocytes might be a possible treatment approach.


Assuntos
Cardiotoxicidade , Doxorrubicina , Camundongos , Animais , Cardiotoxicidade/tratamento farmacológico , Camundongos Endogâmicos C57BL , Doxorrubicina/toxicidade , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , Miócitos Cardíacos
9.
Toxicol Lett ; 373: 94-104, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36435412

RESUMO

Microcystin-LR (MC-LR) has been recognized as a typical hepatotoxic cyclic peptides produced by cyanobacteria. Nowadays, due to the frequent occurrence of cyanobacterial blooms, the underlying hepatotoxic mechanism of MC-LR has become the focus of attention. In our present work, the mutagenic effect of MC-LR on human normal hepatic (HL-7702) cells regulated by cGAS was mainly studied. Here, we showed that exposure to MC-LR for 1-4 days could activate the cGAS-STING signaling pathway and then trigger immune response in HL-7702 cells. Notably, relative to the treatment with 1 µM MC-LR for 1-3 days, it was observed that when HL-7702 cells were exposed to 1 µM MC-LR for 4 days, the mutation frequency at the Hprt locus was remarkably increased. In addition, cGAS in HL-7702 cells was also found to complete the nuclear translocation after 4-day exposure. Moreover, co-immunoprecipitation and homologous recombination (HR)-directed DSB repair assay were applied to show that homologous recombination repair was inhibited after 4-day exposure. However, the intervention of the nuclear translocation of cGAS by transfecting BLK overexpression plasmid restored homologous recombination repair and reduced the mutation frequency at the Hprt locus in HL-7702 cells exposed to MC-LR. Our study unveiled the distinct roles of cGAS in the cytoplasm and nucleus of human hepatocytes as well as potential mutagenic mechanism under the early and late stage of exposure to MC-LR, and provided a novel insight into the prevention and control measures about the hazards of cGAS-targeted MC-LR.


Assuntos
Cianobactérias , Reparo de DNA por Recombinação , Humanos , Hipoxantina Fosforribosiltransferase/farmacologia , Microcistinas/toxicidade , Hepatócitos , Nucleotidiltransferases/farmacologia , Mutagênese
10.
Am J Chin Med ; 51(1): 91-105, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36437551

RESUMO

Carbon tetrachloride (CCl4)-induced lipid peroxidation associated with hepatic oxidative stress and cell death is an important mechanism of acute liver injury (ALI). Ginsenoside Rd is considered an active ingredient of ginseng. Evidence suggests that ginsenoside Rd may improve ischaemic stroke, nerve damage, cancer and other diseases involving apoptosis, inflammation, oxidative stress, mitochondrial injury and autophagy. However, the effects of ginsenoside Rd on CCl4-induced ALI and its underlying mechanisms are still unclear. In this study, 0.25% CCl4 was injected intraperitoneally in mice to establish a CCl4-induced ALI model. In the Rd treatment group, Rd (10, 20[Formula: see text]mg/kg) doses were injected intraperitoneally 1[Formula: see text]h before and 23[Formula: see text]h after CCl4 administration. Ferroptosis inducer imidazole ketone erastin (IKE) was injected intraperitoneally 4[Formula: see text]h before CCl4 administration to explore the mechanism. The blood and liver were collected 24[Formula: see text]h after CCl4 administration to investigate the effect and mechanism of ginsenoside Rd on CCl4-induced ALI. Our results showed that ginsenoside Rd inhibited CCl4-induced ALI in mice. Ginsenoside Rd also downregulated CCl4-induced serum and liver iron, 4-hydroxynonenal, and 8-hydroxy-2 deoxyguanosine levels. Furthermore, it upregulated glutathione and glutathione peroxidase 4 levels. In addition, ginsenoside Rd downregulated the expression of cGAS and STING. Subsequently, the ferroptosis inducer imidazole ketone erastin significantly reversed the hepatoprotective effect and influence of ginsenoside Rd with regard to the indicators mentioned above. Our study confirmed that ginsenoside Rd ameliorated CCl4-induced ALI in mice, which was related to the reduction of ferroptosis. Simultaneously, the ginsenoside Rd-mediated inhibition of the cGAS/STING pathway contributed to its antiferroptosis effect. In conclusion, our results suggested that ginsenoside Rd inhibited ferroptosis via the cGAS/STING pathway, thereby protecting mice from CCl4-induced ALI. These results suggested ginsenoside Rd may be used as a potential intervention treatment against CCl4-induced ALI.


Assuntos
Isquemia Encefálica , Doença Hepática Induzida por Substâncias e Drogas , Ferroptose , Acidente Vascular Cerebral , Camundongos , Animais , Isquemia Encefálica/metabolismo , Fígado/metabolismo , Estresse Oxidativo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , Tetracloreto de Carbono/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
11.
J Adv Res ; 51: 161-179, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36334887

RESUMO

INTRODUCTION: Meteorin-like hormone (Metrnl) is ubiquitously expressed in skeletal muscle, heart, and adipose with beneficial roles in obesity, insulin resistance, and inflammation. Metrnl is found to protect against cardiac hypertrophy and doxorubicin-induced cardiotoxicity. However, its role in diabetic cardiomyopathy (DCM) is undefined. OBJECTIVES: We aimed to elucidate the potential roles of Metrnl in DCM. METHODS: Gain- andloss-of-function experimentswere utilized to determine the roles of Metrnl in the pathological processes of DCM. RESULTS: We found that plasma Metrnl levels, myocardial Metrnl protein and mRNA expressions were significantly downregulated in both streptozotocin (STZ)-induced (T1D) mice and leptin receptor deficiency (db/db) (T2D) mice. Cardiac-specific overexpression (OE) of Metrnl markedly ameliorated cardiac injury and dysfunction in both T1D and T2D mice. In sharp contrast, specific deletion of Metrnl in the heart had the opposite phenotypes. In parallel, Metrnl OE ameliorated, whereas Metrnl downregulation exacerbated high glucose (HG)-elicited hypertrophy, apoptosis and oxidative damage in primary neonatal rat cardiomyocytes. Antibody-induced blockade of Metrnl eliminated the effects of benefits of Metrnl in vitro and in vivo. Mechanistically, Metrnl activated the autophagy pathway and inhibited the cGAS/STING signaling in a LKB1/AMPK/ULK1-dependent mechanism in cardiomyocytes. Besides, Metrnl-induced ULK1 phosphorylation facilitated the dephosphorylation and mitochondrial translocation of STING where it interacted with tumor necrosis factor receptor-associated factor 2 (TRAF2), a scaffold protein and E3 ubiquitin ligase that was responsible for ubiquitination and degradation of STING, rendering cardiomyocytes sensitive to autophagy activation. CONCLUSION: Thus, Metrnl may be an attractive therapeutic target or regimen for treating DCM.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Cardiomiopatias Diabéticas , Animais , Camundongos , Ratos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/farmacologia , Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Miócitos Cardíacos , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia
12.
Mol Pharm ; 20(1): 136-146, 2023 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-36326450

RESUMO

Acute kidney injury (AKI) is the most common side effect of the anti-cancer drug cisplatin, and currently, no effective preventive measures are available in clinical practice. Oxidative stress and DNA damage mechanisms may be involved in cisplatin-induced AKI. In this study, we prepared Kolliphor HS15-based myricetin-loaded (HS15-Myr) nanomicelles and explored the mechanism of protection against cisplatin-induced AKI. In vitro results showed that the HS15-Myr nanomicelles enhanced the antioxidant activity of myricetin (Myr) and inhibited cisplatin-induced proliferation inhibition of HK-2 cells. Moreover, the HS15-Myr nanomicelles inhibited cisplatin-induced reactive oxygen species accumulation, mitochondrial membrane potential reduction, and DNA damage, which might be related to the inhibition of the cyclic GMP-AMP synthase (cGAS)─stimulating interferon gene (STING) signaling pathway. In vivo results in mice showed that the significant reductions in body weight and renal indices and the increased blood urea nitrogen and serum creatinine levels induced by cisplatin could be significantly reversed by pretreating with the HS15-Myr nanomicelles. Furthermore, nanomicelle pretreatment significantly altered the activities of antioxidant enzymes (e.g., GSH, MDA, and SOD) induced by cisplatin. In addition, cisplatin-induced inflammatory responses in mouse kidney tissue were found to be inhibited by pretreatment with HS15-Myr nanomicelles, such as IL-1ß and TNF-α expression. The nanomicelles also significantly inhibited cisplatin-induced activation of the DNA damage-cGAS-STING pathway in kidney tissues. Together, our findings suggest that Myr-loaded nanomicelles are potential nephroprotective drugs.


Assuntos
Injúria Renal Aguda , Cisplatino , Animais , Camundongos , Cisplatino/farmacologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/prevenção & controle , Transdução de Sinais , Antioxidantes/uso terapêutico , Dano ao DNA , Nucleotidiltransferases/farmacologia , Nucleotidiltransferases/uso terapêutico , Rim
13.
Int J Pharm ; 626: 122161, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36058409

RESUMO

Silk fibroin (SF) is a natural polymeric biomaterial widely used in the preparation of drug delivery systems. Herein, silk fibroin peptide (SFP) was self-assembled into nanofibers, encapsulated a poorly water-soluble drug baicalein (SFP/BA NFs), and then used to protect against cisplatin-induced acute kidney injury (AKI). Specifically, the SFP/BA NFs significantly enhanced the aqueous dispersity, storage stability, and in vitro antioxidant activity of BA. SFP/BA NFs increased the drug uptake and localization to mitochondria. In vitro results demonstrated that SFP/BA NFs can relieve the cisplatin-induced HK-2 cell damage, and inhibit the cisplatin-induced accumulation of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) disruption. Mechanism studies demonstrated that SFP/BA NFs may exert nephroprotective effects by inhibiting both the cisplatin-induced DNA damage and the cGAS/STING pathway activation. In vivo results showed that cisplatin treatment resulted in decreased body weight, increased serum creatinine (SCr), and increased blood urea nitrogen (BUN) levels, while SFP/BA NFs reversed the above symptoms. Furthermore, SFP/BA NFs reversed the cisplatin-induced abnormal changes of antioxidant enzymes (e.g., SOD and GSH), and inhibited the cisplatin-induced DNA damage as well as the activation of cGAS/TING. Above all, our results revealed the potential of SFP/BA NFs to protect against cisplatin-induced AKI.


Assuntos
Injúria Renal Aguda , Fibroínas , Nanofibras , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/prevenção & controle , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Apoptose , Materiais Biocompatíveis/uso terapêutico , Cisplatino/farmacologia , Creatinina , Fibroínas/química , Flavanonas , Humanos , Rim/metabolismo , Nanofibras/química , Nucleotidiltransferases/farmacologia , Nucleotidiltransferases/uso terapêutico , Peptídeos/química , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase , Tolnaftato/efeitos adversos , Água/farmacologia
14.
Glia ; 70(12): 2409-2425, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35959803

RESUMO

Inflammasome involvement in Parkinson's disease (PD) has been intensively investigated. Absent in melanoma 2 (AIM2) is an essential inflammasome protein known to contribute to the development of several neurological diseases. However, a specific role for AIM2 in PD has not been reported. In this study, we investigated the effect of AIM2 in the N-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced PD model by use of various knockout and bone marrow chimeric mice. The mechanism of action for AIM2 in PD was assessed by RNA-sequencing and in vitro primary microglial transfection. Results were validated in the A30P transgenic mouse model of PD. In the MPTP mouse model, AIM2 activation was found to negatively regulate neuro-inflammation independent of the inflammasome. Microglial AIM2 deficiency exacerbated behavioral and pathological features of both MPTP-induced and transgenic PD mouse models. Mechanistically, AIM2 reduced cyclic GMP-AMP synthase (cGAS)-mediated antiviral-related inflammation by inhibition of AKT-interferon regulatory factor 3 (IRF3) phosphorylation. These results demonstrate microglial AIM2 to inhibit the antiviral-related neuro-inflammation associated with PD and provide for a foundation upon which to identify new therapeutic targets for treatment of the disease.


Assuntos
Melanoma , Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Antivirais/farmacologia , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/farmacologia , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , RNA/metabolismo
15.
Free Radic Biol Med ; 188: 26-34, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35697291

RESUMO

Triptolide (TPL) is a bioactive component extracted from the traditional Chinese herb Tripterygium wilfordii Hook F., and has multiple pharmacological activities, such as anti-tumor activity. However, severe adverse effects and toxicity, especially nephrotoxicity, limit its clinical application. It has been demonstrated that mitochondrial defect is a major toxic effects of TPL. In this study, we show that triptolide activated the cGAS-STING signaling pathway in kidney tubular cells in vivo and in vitro. Renal injury models were established in BALB/c mice and human tubular epithelial cells using TPL. We found that TPL enhanced the phosphorylation levels of STING, TBK1 and IRF3, and upregulated the expression of IFNß, which is the production of cGAS-STING signaling pathway. STING inhibitor C176 had protective effects in TPL-induced nephrocyte damage. STING siRNA down regulated the expression level of IFNß. In addition, triptolide induced an increase in protein levels of the transcription factor BACH1, while transcriptional expression of the antioxidant enzyme HMOX1 was reduced due to the increased expression of BACH1. Furthermore, oxidative stress-induced mtDNA damage and DNA leakage caused activation of the cGAS-STING signaling pathway. Altogether, cGAS-STING signaling pathway involved in TPL induced nephrotoxicity. Inhibiting cGAS-STING over-activation may be a new strategy for alleviating renal injury of triptolide.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Fenantrenos , Animais , Diterpenos , Compostos de Epóxi , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , Estresse Oxidativo , Fenantrenos/toxicidade , Transdução de Sinais
16.
Redox Biol ; 52: 102305, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35367811

RESUMO

Hepatocellular cell death and macrophage proinflammatory activation contribute to the pathology of various liver diseases, during which XBP1 plays an important role. However, the function and mechanism of XBP1 in thioacetamide (TAA)-induced acute liver injury (ALI) remains unknown. Here, we investigated the effects of XBP1 inhibition on promoting hepatocellular pyroptosis to activate macrophage STING signaling during ALI. While both TAA- and LPS-induced ALI triggered XBP1 activation in hepatocytes, hepatocyte-specific XBP1 knockout mice exhibited exacerbated ALI with increased hepatocellular pyroptosis and enhanced macrophage STING activation. Mechanistically, mtDNA released from TAA-stressed hepatocytes could be engulfed by macrophages, further inducing macrophage STING activation in a cGAS- and dose-dependent manner. XBP1 deficiency increased ROS production to promote hepatocellular pyroptosis by activating NLRP3/caspase-1/GSDMD signaling, which facilitated the extracellular release of mtDNA. Moreover, impaired mitophagy was found in XBP1 deficient hepatocytes, which was reversed by PINK1 overexpression. Mitophagy restoration also inhibited macrophage STING activation and ALI in XBP1 deficient mice. Activation of XBP1-mediated hepatocellular mitophagy and pyroptosis and macrophage STING signaling pathway were observed in human livers with ALI. Collectively, these findings demonstrate that XBP1 deficiency promotes hepatocyte pyroptosis by impairing mitophagy to activate mtDNA/cGAS/STING signaling of macrophages, providing potential therapeutic targets for ALI.


Assuntos
Mitofagia , Piroptose , Proteína 1 de Ligação a X-Box/metabolismo , Animais , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia , Transdução de Sinais
17.
Int J Biol Sci ; 18(4): 1737-1754, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35280696

RESUMO

Atrial Natriuretic Peptide (ANP) has known anti-inflammatory effects. However, the role of ANP in Ulcerative colitis (UC) remains unclear. This study aimed to explore the expression and function of ANP in UC, and its potential regulatory role in the stimulator of interferon genes (STING) pathway. Human colon biopsy and serum samples were collected between September 2018 and December 2019 at Wuhan Union Hospital. Levels of ANP and its receptors and STING pathway components were detected in people with UC and mice with dextran sulfate sodium (DSS)-induced colitis. These mice and HT-29 cells were treated with ANP and an agonist of the STING pathway. The level of inflammation, STING pathway, gut barrier, and endoplasmic reticulum (ER) stress-induced autophagy were measured. We found that the levels of ANP and its receptor decreased and the STING pathway activated statistically in people with UC and the mouse model of colitis. ANP treatment attenuated DSS-induced colitis and inhibited STING pathway phosphorylation in colonic tissue and epithelial cells. An interaction between cGAS and NPR-A was verified. ANP repaired the gut barrier and inhibited ER stress-induced autophagy via the STING pathway. ANP may thus alter colonic barrier function and regulate ER stress-induced autophagy as a promising therapy for UC.


Assuntos
Colite Ulcerativa , Colite , Animais , Fator Natriurético Atrial , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Colite Ulcerativa/metabolismo , Colo/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Humanos , Camundongos , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/farmacologia
18.
Biochemistry ; 29(10): 2501-6, 1990 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-2159322

RESUMO

We have previously reported for the first time the purification to homogeneity of the enzyme NMN adenylyltransferase (EC 2.7.7.1) from yeast and its major molecular and catalytic properties. The homogeneous enzyme was found to be a glycoprotein containing 2% carbohydrate and 1 mol of adenine residue and 2 mol of phosphate covalently bound per mole of protein. Such a stoichiometry, apparently consistent with that of ADP-ribose, prompted us to further investigate the possibility that NMN adenylyltransferase could be subjected to poly(ADP-ribosylation) in vitro in a reconstituted system. Poly(ADP-ribose) polymerase was purified to homogeneity from bull testis by means of a rapid procedure involving two batchwise steps on DNA-agarose and Reactive Blue 2 cross-linked agarose and a column affinity chromatography step on 3-aminobenzamide-Sepharose; the optimal conditions for the poly(ADP-ribosylation) of exogenous substrates were determined. When pure NMN adenylyltransferase was incubated in the presence of the homogeneous poly(ADP-ribose) polymerase, a marked inhibition of the polymerase was observed, both in the presence and in the absence of histones, while the activity of NMN adenylyltransferase was not affected. The inhibition could not be prevented by increasing the concentrations of either DNA or NAD. Mg2+ did not affect the activity or the inhibition. The significance of such a phenomenon is at present unknown, but it may be of biological relevance in view of the close topological and metabolic relationship between the two enzymes.


Assuntos
Nicotinamida-Nucleotídeo Adenililtransferase/farmacologia , Nucleotidiltransferases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Leveduras/enzimologia , Animais , Bovinos , DNA/metabolismo , Eletroforese em Gel de Poliacrilamida , Histonas/genética , Histonas/metabolismo , Cinética , Magnésio/farmacologia , Masculino , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Testículo/enzimologia , Leveduras/efeitos dos fármacos
19.
J Biol Chem ; 261(34): 15995-9, 1986 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-2877983

RESUMO

Gentamicin nucleotidyltransferase-catalyzed reaction of (Sp)-[alpha-17O]dATP with tobramycin produced 2"-(2'-deoxyadenosine 5'-[17O]phosphoryl)tobramycin. The configuration at phosphorus in this product was shown to be Rp by chemical degradation to chiral [17O, 18O]dAMP using a stereochemically defined procedure, and determination of the configuration at phosphorus in this product. Periodate-base treatment of 2"-(2'-deoxyadenosine 5'-[17O]phosphoryl)tobramycin followed by NaBH4 reduction produced (2-glyceryl)-[17O]dAMP, which upon snake venom phosphodiesterase-catalyzed hydrolysis in H(2)18O produced [17O,18O] dAMP. The configuration at phosphorus in this product was shown to be S by enzymatic phosphorylation to [17O,18O]dATP, adenylylcyclase (Bordetella pertussis)-catalyzed cyclization to 3',5'-cyclic [17O,18O]dAMP, and 31P NMR analysis of the ethyl esters. Since snake venom phosphodiesterase-catalyzed hydrolyses proceed with retention of configuration at phosphorus, (Sp)-[17O,18O]dAMP must have been produced from (Rp)-(2-glyceryl)-[17O]dAMP; and since the chemical degradation to the latter compound did not involve cleavage of any bonds to phosphorus, the initial enzymatic product must have been (Rp)-2"-(2'-deoxyadenosine 5'-[17O]phosphoryl)tobramycin. Therefore, nucleotidyl transfer catalyzed by gentamicin nucleotidyl-transferase proceeds with inversion of configuration at phosphorus, and the reaction mechanism involves an uneven number of phosphotransfer steps. Inasmuch as this is an uncomplicated two-substrate group transfer reaction, the mechanism probably involves direct nucleotidyl transfer from the nucleoside triphosphate to the aminoglycoside. The B. pertussis adenylylcyclase reaction was shown to proceed with inversion at phosphorus, as has been established for other adenylylcyclases.


Assuntos
Nucleotídeos de Desoxiadenina/metabolismo , Nucleotidiltransferases/farmacologia , Tobramicina/metabolismo , Adenilil Ciclases/farmacologia , Bordetella pertussis/enzimologia , Conformação Molecular , Isótopos de Oxigênio , Fósforo
20.
Infect Immun ; 34(1): 147-53, 1981 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-6271678

RESUMO

Exoenzyme S differs from toxin A and diphtheria toxin in that it does not adenosine diphosphate (ADP)-ribosylate elongation factor-2, but rather catalyzes the transfer of the ADP-ribose moiety of nicotinamide adenine dinucleotide to a number of different proteins in extracts of eucaryotic cells. Polyoma-transformed BHK-21 cells were isolated which were resistant to diphtheria toxin and toxin A. Extracts from these cells are ADP-ribosylated by exoenzyme S but not toxin A or diphtheria toxin, providing an assay which distinguishes between S and A activities. A total of 124 clinical isolates of P. aeruginosa were analyzed for production of toxin A and exoenzyme S. Exoenzyme S production was detected in 38% of the strains, whereas 80% of the strains produced toxin A.


Assuntos
ADP Ribose Transferases , Toxinas Bacterianas , Nucleotidiltransferases/biossíntese , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/enzimologia , Fatores de Virulência , Adenosina Difosfato Ribose/metabolismo , Animais , Bioensaio , Linhagem Celular , Transformação Celular Viral , Cricetinae , Exotoxinas/biossíntese , Nucleotidiltransferases/farmacologia , Poli(ADP-Ribose) Polimerases , Polyomavirus , Proteínas/metabolismo , Exotoxina A de Pseudomonas aeruginosa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA