Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Int Immunopharmacol ; 128: 111552, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38280335

RESUMO

Colonic mucosal defect constitutes the major reason of recurrence and deterioration of ulcerative colitis (UC), and mucosal healing has become the therapeutic endpoint of UC. Unfortunately, specific promoter of mucosal healing is still absent. Our previous researches demonstrated that arctigenin could alleviate colitis symptoms in mice, but whether it has a positive impact on colonic mucosal healing remains unclear. This study explores whether and how arctigenin promotes mucosal healing. Orally administered arctigenin was shown to alleviate colitis in mice primarily by enhancing mucosal healing. In vitro, arctigenin was shown to promote the wound healing by accelerating colonic epithelial cell migration but not proliferation. Acceleration of the focal adhesion turnover, especially assembly, is crucial for arctigenin promoting the cell migration. Arctigenin was able to activate focal adhesion kinase (FAK) in colonic epithelial cells through directly binding with Tyr251 site of FAK, as evidenced by surface plasmon resonance assay and site-directed mutagenesis experiment. In the colonic epithelial cells of UC patients and colitis mice, FAK activation was significantly down-regulated compared with the controls. Arctigenin promoted colonic epithelial cell migration and mucosal healing in dextran sulphate sodium (DSS)-induced colitis mice dependent on activating FAK, as confirmed by combined use with FAK inhibitor. In summary, arctigenin can directly promote mucosal healing in colitis mice through facilitating focal adhesion turnover, especially assembly, and consequent migration of epithelial cells via targeting FAK. Arctigenin may be developed as a mucosal healing promoter, and FAK is a potential therapeutic target for UC and other mucosal defect-related diseases.


Assuntos
Colite Ulcerativa , Colite , Furanos , Lignanas , Humanos , Camundongos , Animais , Colite Ulcerativa/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/uso terapêutico , Adesões Focais/metabolismo , Colite/induzido quimicamente , Movimento Celular , Cicatrização , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Sulfato de Dextrana , Camundongos Endogâmicos C57BL
2.
Front Cardiovasc Med ; 10: 1174081, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37731521

RESUMO

Background: Immunoglobulin E (IgE) is implicated in the pathogenesis of acute myocardial infarction (AMI), and smokers often exhibit elevated plasma IgE levels. However, it remains uncertain whether the role of smoking in the development and prognosis of AMI is influenced by IgE levels. This study aimed to investigate the potential contribution of IgE in mediating the association between smoking and AMI. Methods: We conducted a prospective study involving 348 consecutive patients with chest discomfort who underwent coronary angiography. Plasma cotinine, an alkaloid present in tobacco, and IgE levels were measured. The patients were followed up for mean 39-months to assess their long-term prognosis based on major adverse cardiac and cerebrovascular events (MACCE). Results: Our findings indicate that patients with AMI had higher plasma levels of cotinine and IgE. Univariate analyses demonstrated a positive association between plasma cotinine (OR = 1.7, 95% CI: 1.27-2.26, P < 0.001) and IgE (OR = 2.8, 95% CI: 1.75-4.39, P < 0.001) with AMI. Receiver operating characteristics analyses showed that the combined use of cotinine and IgE (AUC: 0.677) had a larger predictive performance compared to cotinine alone (AUC: 0.639) or IgE alone (AUC: 0.657), although the improvement did not reach statistical significance. Multivariable logistic regression revealed a positive association between plasma cotinine and AMI (OR = 1.70, 95% CI: 1.04-2.78, P = 0.036). Furthermore, the inclusion of plasma IgE in the regression model led to a decrease in the OR and 95% CI of plasma cotinine (OR = 1.66, 95% CI: 1.01-2.73, P = 0.048). Process mediation analyses showed a significant indirect effect of plasma cotinine on AMI mediated through increased plasma IgE. Kaplan-Meier analysis during a mean 39-months follow-up revealed that higher plasma levels of IgE were associated with an increased risk of MACCE following AMI (P = 0.047). However, in the context of the COX regression analysis, no significant correlation was observed between IgE, cotinine and AMI. Conclusion: Cotinine exhibits a positive association with AMI, wherein IgE plays a mediating role. Elevated plasma levels of IgE was positively associated with AMI and poor prognosis, which further confirms the adverse role of smoking on the incidence of AMI and prognosis. (Clinical trial registration: ChiCTR2100053000).

3.
J Colloid Interface Sci ; 652(Pt A): 577-589, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37611467

RESUMO

Li-rich Mn-based oxides (LRMOs) are considered as one of the most-promising cathode materials for next generation Li-ion batteries (LIBs) because of their high energy density. Nevertheless, the intrinsic shortcomings, such as the low first coulomb efficiency, severe capacity/voltage fade, and poor rate performance seriously limit its commercial application in the future. In this work, we construct successfully g-C3N4 coating layer to modify Li1.2Mn0.54Ni0.13Co0.13O2 (LMNC) via a facile solution. The g-C3N4 layer can alleviate the side-reaction between electrolyte and LMNC materials, and improve electronic conduction of LMNC. In addition, the g-C3N4 layer can suppress the collapse of structure and improve cyclic stability of LMNC materials. Consequently, g-C3N4 (4 wt%)-coated LMNC sample shows the highest initial coulomb efficiency (78.5%), the highest capacity retention ratio (78.8%) and the slightest voltage decay (0.48 V) after 300 loops. Besides, it also can provide high reversible capacity of about 300 and 93 mAh g-1 at 0.1 and 10C, respectively. This work proposes a novel approach to achieve next-generation high-energy density cathode materials, and g-C3N4 (4 wt%)-coated LMNC shows an enormous potential as the cathode materials for next generation LIBs with excellent performance.

4.
Phytomedicine ; 119: 154985, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37516090

RESUMO

BACKGROUND: Mume Fructus (MF) is the fruit of Prunus mume Sieb. et Zucc, a plant of Rosaceae family. Previous studies demonstrated that MF was capable of ameliorating ulcerative colitis (UC) in mice, its action mechanism needs to be clarified. PURPOSE: This study deciphered whether and how MF extract accelerates colonic mucosal healing, the therapeutic endpoint of UC. METHODS: Biochemical, histopathological and qRT-PCR analyses were utilized to define the therapeutic efficacy of MF on dextran sulfate sodium (DSS)-induced colitis in mice. UHPLC-QTOF-MS/MS-based metabolomics technique was adopted to explore the changes of endogenous metabolites associated with UC and responses to MF intervention. qRT-PCR analysis was performed to confirm the molecular pathway in vivo. The effects of MF and lysophosphatidylcholine (LPC) on cell viability, wound healing, proliferation, and migration were examined through a series of in vitro experiments. Moreover, the effects of different subtypes of phospholipase A2 (PLA2) inhibitors on MF-treated colonic epithelial cells were detected by wound healing test and transwell assay. RESULTS: Orally administered MF could alleviate colitis in mice mainly by accelerating the healing of colonic mucosa. Guided by an unbiased metabolomics screen, we identified LPC synthesis as a major modifying pathway in colitis mice after MF treatment. Notably, MF facilitated the synthesis of LPC by enhancing the expression of PLA2 in colitis mice. Mechanistically, MF and LPC accelerated wound closure by promoting cell migration. Moreover, the promotion of MF on wound healing and migration of colonic epithelial cells was blunted by a cytosolic phospholipase A2 (cPLA2) inhibitor. CONCLUSION: MF can facilitate colonic mucosal healing of mice with colitis through cPLA2-mediated intestinal LPC synthesis, which may become a novel therapeutic agent of UC.


Assuntos
Colite Ulcerativa , Colite , Prunus , Camundongos , Animais , Sulfato de Dextrana/efeitos adversos , Lisofosfatidilcolinas/metabolismo , Prunus/química , Frutas/química , Espectrometria de Massas em Tandem , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Colo/patologia , Colite Ulcerativa/tratamento farmacológico , Cicatrização , Mucosa Intestinal/metabolismo , Fosfolipases A2 Citosólicas/análise , Fosfolipases A2 Citosólicas/metabolismo , Fosfolipases A2 Citosólicas/farmacologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
5.
Environ Geochem Health ; 45(10): 7115-7130, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37453967

RESUMO

The upstream basin of Hanjiang River is an important water source for the middle route of China's South-to-North Water Diversion Project. The quality of water and soil in the Hanjiang River have enormous biological and environmental impacts, and resistant genetic contamination has emerged, but only few studies are concerned the correlation between heavy metals and metal resistance genes (MRGs). In this study, 8 antibiotics and 19 heavy metals were analyzed, the results showed that the highest antibiotic content was tetracycline, with mean concentrations of 43.201 µg/kg and 0.022 µg/L. Mn was the highest heavy metal in soil with a content of 1408.284 µg/kg, and in water was Zn with a content of 10.611 µg/L. We found that the most abundant antibiotic resistance genes (ARGs) and metal resistance genes (MRGs) in the study area were bacA and arsT genes, coding for resistance mechanisms to bacitracin and arsenic, respectively. The data showed that heavy metals had a greater impact on antibiotic resistance genes than antibiotics, and the correlation between resistance genes was significantly positive. This work expands our understanding of the correlations of antibiotics, heavy metals, and resistance genes in the Hanjiang River, indicating that more attention should be paid to the effects of resistance genes and the quality of water.


Assuntos
Antibacterianos , Metais Pesados , Antibacterianos/farmacologia , Antibacterianos/análise , Rios , Genes Bacterianos , Metais Pesados/análise , Solo , Água , China , Monitoramento Ambiental
6.
Acta Pharm Sin B ; 13(4): 1600-1615, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37139415

RESUMO

Both cholinergic dysfunction and protein citrullination are the hallmarks of rheumatoid arthritis (RA), but the relationship between the two phenomena remains unclear. We explored whether and how cholinergic dysfunction accelerates protein citrullination and consequently drives the development of RA. Cholinergic function and protein citrullination levels in patients with RA and collagen-induced arthritis (CIA) mice were collected. In both neuron-macrophage coculture system and CIA mice, the effect of cholinergic dysfunction on protein citrullination and expression of peptidylarginine deiminases (PADs) was assessed by immunofluorescence. The key transcription factors for PAD4 expression were predicted and validated. Cholinergic dysfunction in the patients with RA and CIA mice negatively correlated with the degree of protein citrullination in synovial tissues. The cholinergic or alpha7 nicotinic acetylcholine receptor (α7nAChR) deactivation and activation resulted in the promotion and reduction of protein citrullination in vitro and in vivo, respectively. Especially, the activation deficiency of α7nAChR induced the earlier onset and aggravation of CIA. Furthermore, deactivation of α7nAChR increased the expression of PAD4 and specificity protein-3 (SP3) in vitro and in vivo. Our results suggest that cholinergic dysfunction-induced deficient α7nAChR activation, which induces the expression of SP3 and its downstream molecule PAD4, accelerating protein citrullination and the development of RA.

7.
Mol Nutr Food Res ; 67(12): e2200784, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36938915

RESUMO

SCOPE: Colonic mucosal healing is the terminal goal for the treatment of ulcerative colitis (UC), but there is currently no specific drug available. This study investigates the beneficial effect of diallyl trisulfide (DATS) on the colonic mucosal healing. METHODS AND RESULTS: Dextran sulfate sodium (DSS) is used to induce colitis in female C57BL/6 mice, and DATS is orally administered during the recovery period. DATS hardly impacts the inflammation of the colonic tissues, but significantly promotes the mucosal repair. DATS promotes the migration but not proliferation of colonic epithelial cells in the colitis mice. In addition, DATS accelerates the wound healing, cell migration, focal adhesion assembly, and phosphorylation of focal adhesion kinase (FAK) of colonic epithelial cells in vitro, which are evidently reversed by combined use of FAK inhibitor PF-573228. Similar results are shown in colitis mice. Mechanically, DATS promotes the binding of Rab21 to integrin ß1 and accelerates the endocytosis of integrin ß1, which is significantly attenuated by the knockdown of Rab21. CONCLUSIONS: DATS promotes the binding of Rab21 to integrin ß1 and the endocytosis of integrin ß1, thereby increases FAK phosphorylation and focal adhesion assembly, finally accelerates the migration of colonic epithelial cells and mucosal healing.


Assuntos
Colite Ulcerativa , Colite , Alho , Feminino , Camundongos , Animais , Integrina beta1/metabolismo , Integrina beta1/farmacologia , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Adesões Focais , Camundongos Endogâmicos C57BL , Movimento Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Células Epiteliais/metabolismo
8.
Immunology ; 169(4): 412-430, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36930164

RESUMO

Several aryl hydrocarbon receptor (AhR) agonists have been reported to promote the generation of regulatory T cells (Treg cells), and the action mechanisms need to be identified. In this study, we addressed the underlying mechanism of AhR activation to induce the generation of Treg cells in the view of cellular metabolism. Naïve CD4+ T cells were purified with mouse CD4+ CD62L+ T Cells Isolation Kits. The proportions of Treg cells were detected by flow cytometry. The value of oxygen consumption rate (OCR) of CD4+ T cells was detected by the Seahorse XFe 96 analyzer. The activation of fatty acid oxidation (FAO)-related metabolic pathways was detected by Western blotting. Intracellular localization of Lkb1 was detected by immunofluorescence. The Strad-Mo25-Lkb1 complex formation and K63 chain ubiquitination modification of Lkb1 were detected by co-immunoprecipitation. The binding of AhR to the Skp2 promoter was detected by constructing luciferase reporter gene. AhR or carnitine palmitoyltransferases 1 was knockdown in dextran sulphate sodium (DSS)-induced colitis or collagen-induced arthritis (CIA) mice by infecting mice with adeno-associated virus via the tail vein injection. Compared to the control group, exogenous and endogenous AhR agonists 3,3'-diindolylmethane (DIM) and 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) were shown to preferentially upregulate the mRNA expression of FAO-related enzymes and the value of OCR. Consistently, pharmacological or genetic inhibition of FAO markedly diminished the induction of DIM and ITE on the differentiation of Treg cells. DIM and ITE functioned mainly through activating the liver kinase B1 (Lkb1)-AMPK pathway via promotion of Lkb1-Strad-Mo25 complex formation and Lkb1 K63 ubiquitination. DIM and ITE were also shown to upregulate the mRNA expression of Skp2, a ubiquitination-related enzyme, and facilitate the binding of AhR to the xenobiotic responsive element of Skp2 promoter region by luciferase reporter gene assay. Furthermore, the contribution of Skp2/K63 ubiquitination/Lkb1/FAO axis was verified in (DSS)-induced colitis or CIA mice. In summary, these findings indicate that AhR activation promotes Treg cell generation by enhancing Lkb1-mediated FAO via the Skp2/K63-ubiquitination pathway, and AhR agonists may be used as inducers of Treg cells to prevent and treat autoimmune diseases.


Assuntos
Colite , Linfócitos T Reguladores , Camundongos , Animais , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Colite/metabolismo , Ubiquitinação , Ácidos Graxos/metabolismo , RNA Mensageiro
9.
Life Sci ; 309: 120972, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36116532

RESUMO

Butyrate, given by oral administration or in drinking water, has been shown to improve experimental pulmonary fibrosis (PF) in mice despite of very low bioavailability. The pharmacokinetic-pharmacodynamics disconnection attracts us to explore its anti-PF mechanism in view of the intestinal expression of anti-PF factors. In bleomycin-induced PF in mice, rectal administration of butyrate (500 mg/kg) exhibited a significant anti-PF effect, with a maximum plasma concentration largely lower than the minimum effective concentration (1 mM) at which butyrate inhibited the expression of pro-inflammatory factors by lung epithelial cells and the production of extracellular matrix by lung fibroblasts. The rectal administration of butyrate significantly upregulated the mRNA expression of hepatocyte growth factor (HGF) in the colons of PF mice, but showed no significant effect on the mRNA expression of HGF in the small intestines, lungs and livers. In colon epithelial cells, the monocarboxylate transporter inhibitor α-cyano-4-hydroxycinnamic acid (CHC) abrogated butyrate-induced expression of HGF, indicating that butyrate functions through entering into cells. Butyrate showed no significant effect on the histone acetylation in the promoter region of HGF, suggesting that it promotes HGF expression not by directly affecting the histone deacetylation of HGF but by other pathways. GW9662, the inhibitor of PPARγ, significantly attenuated the effect of butyrate to promote the mRNA expression of HGF. Butyrate was able to enhance the acetylation of PPARγ, and a targeted mutation of lysine at the position 240 (K240) of PPARγ markedly diminished the induction of butyrate on HGF expression, suggesting that butyrate promoted HGF expression in colon epithelial cells by upregulating PPARγ K240 acetylation. In summary, rectal administration of butyrate promotes the expression of HGF in colonic epithelial cells through upregulating PPARγ acetylation via inhibition of HDAC activity. The findings of the present study provide a reasonable explanation for the anti-PF action mode of butyrate based on the 'lung-gut axis', and found that intestine-derived butyrate and HGF may be involved in the modulation of the occurrence and progression of PF.


Assuntos
Água Potável , Fibrose Pulmonar , Camundongos , Animais , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/farmacologia , PPAR gama/metabolismo , Butiratos/farmacologia , Butiratos/metabolismo , Butiratos/uso terapêutico , Administração Retal , Lisina/metabolismo , Água Potável/efeitos adversos , Água Potável/metabolismo , Histonas/metabolismo , Bleomicina/farmacologia , Colo/metabolismo , RNA Mensageiro/metabolismo
11.
Biochem Pharmacol ; 202: 115138, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35700756

RESUMO

INTRODUCTION: Madecassic acid (MA), a triterpene compound isolated from Centella Asiatica herbs, has previously been shown to attenuate colitis induced by DSS in mice. In the present study, we address whether and how MA ameliorates colitis-associated colorectal cancer (CAC), which accounts for a considerable proportion of colorectal cancer. METHODS: CAC was induced by AOM/DSS in mice, and MA was administered orally once a day. To identify the source cells of IL-17 and the target cells for MA reducing the expression of IL-17 in the colons of CAC mice, single-cell suspensions were prepared from the colons of CAC mice and analyzed by flow cytometry. An adoptive transfer experiment was performed to verify the importance of the decreasing γδT17 cell population in the anti-CAC effect of MA. RESULTS: Oral administration of MA reduced the burden and incidence of tumors in the CAC mice. MA decreased the number of MDSCs in the colon tissues of CAC mice and ameliorated anti-tumor immune responses. MA could prevent the migration of MDSCs by inhibiting the activation of γδT17 cells and the expression of chemokines. The population of activated-γδT17 cells in the tumor microenvironment of CAC mice positively correlated with the number of MDSCs and tumors as well as tumor load. Moreover, the anti-CAC effect of MA was significantly counteracted by the adoptive transfer of γδT17 cells. CONCLUSIONS: MA alleviates CAC by blocking the recruitment of MDSCs to increase the population of anti-tumor immune cells in tumor microenvironment via inhibition of the activation of γδT17 cells.


Assuntos
Neoplasias Associadas a Colite , Colite , Neoplasias Colorretais , Células Supressoras Mieloides , Triterpenos , Animais , Azoximetano , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Neoplasias Colorretais/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Interleucina-17/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Transdução de Sinais , Células Th17 , Triterpenos/farmacologia , Triterpenos/uso terapêutico , Microambiente Tumoral
12.
Int Immunopharmacol ; 108: 108843, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35598399

RESUMO

The neuropeptide cortistatin (CST) has been reported to attenuate Th17 cell response in multiple disease models, but the mechanism of action remains obscure. Here, we show that either overexpression or exogenous addition of CST obviously restricts Th17 cell differentiation. As metabolic reprogramming plays an important role in Th17 cell development, we explore whether CST inhibits Th17 cell differentiation by regulating glycolysis. The results show that CST substantially attenuates the glycolysis during Th17 differentiation and down-regulates the mRNA expression of myelocytomatosis oncogene (Myc) and hexokinase 2 (HK2), the glycolysis rate-limiting enzyme. Following the overexpression of Myc and HK2, the inhibitory effect of CST on Th17 differentiation nearly disappears, suggesting that Myc-HK2 pathway is deeply involved. Furthermore, growth hormone secretagogue receptor 1 (GHSR1) is identified as the key receptor subtype for CST attenuating glycolysis and Th17 cell differentiation by the combined uses of CST with various receptor subtype blockers. The knockdown of GHSR1 abrogates the inhibition of CST on Th17 differentiation and glycolysis. Finally, in the colitis mice induced by dextran sulfate sodium, an intraperitoneal injection of CST markedly inhibits Th17 cell response and down-regulates the expression of HK2 in the Th17 cells, which is reversed by the combined use of GHSR1 antagonist. These findings suggest that inhibition of glycolysis is the key pathway for CST attenuating Th17 cell response, and GHSR1, Myc and HK2 are potential therapeutic targets of Th17 cell-related diseases.


Assuntos
Neuropeptídeos , Células Th17 , Animais , Glicólise , Camundongos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Receptores de Grelina/metabolismo
13.
Immunology ; 166(4): 492-506, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35569103

RESUMO

Our previous studies have demonstrated that tetrandrine can induce the generation of regulatory T (Treg) cells in vitro and in vivo. But, the underlying mechanism of tetrandrine remains obscure. Naïve CD4+ T cells are isolated from the mesenteric lymph nodes of mice for the differentiation of Treg cells. Flow cytometry is used to detect the frequencies of Treg cells. Non-targeted metabolomics analysis based on UHPLC-QTOF/MS is performed to assess the intracellular metabolic profiles. ChIP-PCR analysis is conducted to detect the level of H3K27ac at Foxp3 promoter and CNS regions. Tetrandrine treatment alters the metabolic profile of Treg cells, and pathway enrichment of differential metabolites mainly involves fatty acid oxidation (FAO). Tetrandrine promotes the mRNA expression of carnitine palmitoyl transferase-1, and increases the level of acetyl coenzyme A (acetyl-CoA) and the intracellular oxygen consumption rate. Either CPT1 inhibitor (etomoxir) or siRNA markedly diminishes the promotion of tetrandrine on Treg cell differentiation. Furthermore, tetrandrine enhances the acetylation of H3K27 in the promoter and CNS1 regions of Foxp3 through the acetyl-CoA derived from FAO. In the mice with collagen-induced arthritis, tetrandrine also induces Treg cell generation through FAO pathway. In addition, tetrandrine enhances the immunosuppressive function of Treg cells both in vitro and in vivo. The findings indicate that tetrandrine promotes Treg cell differentiation by enhancing FAO-mediated Foxp3 acetylation, and the CPT1-mediated FAO can serve as the target for the discovery of novel inducers of Treg cell generation.


Assuntos
Alcaloides , Antineoplásicos , Acetilcoenzima A/metabolismo , Alcaloides/metabolismo , Animais , Benzilisoquinolinas , Ácidos Graxos/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Imunossupressores/farmacologia , Camundongos , Linfócitos T Reguladores/metabolismo
14.
Thorac Cancer ; 13(11): 1631-1641, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35437954

RESUMO

BACKGROUND: Immune checkpoint inhibitors (ICIs) have become standard-of-care in patients with pretreated advanced esophageal squamous cell carcinoma (ESCC). However, reliable biomarkers for clinical outcomes are lacking for ICIs. The exploration of effective biomarkers is therefore needed to optimize patient benefit in the treatment of ESCC. METHODS: Sixty-nine patients with advanced ESCC enrolled at one center from two prospective trials were consecutively analyzed. NLR was dynamically collected and high-resolution HLA-I genotyping were performed on genomic DNA. Overall response rate (ORR), median progression-free survival (mPFS) and median overall survival (mOS) were investigated. RESULTS: Thirty-three (47.8%) of 69 patients with baseline NLR ≥4 demonstrated significantly worse clinical outcomes (ORR 9.1% vs. 36.1%, p = 0.018; mPFS 1.8 vs. 3.2 months, hazard ratio [HR] 1.79, p = 0.026; mOS 7.4 vs. 11.0 months, HR 2.28, p = 0.008). An NLR decrease ≥20% at the first radiological evaluation was associated with longer OS (median, 14.0 vs. 7.9 months, p = 0.038). Eleven (15.9%) patients with HLA-I homozygosity presented poorer clinical outcomes (ORR 0 vs. 27.6%, p = 0.056; mPFS 1.8 vs. 2.4 months, HR 3.37, p = 0.010; mOS 5.6 vs. 10.5 months, HR 3.97, p = 0.004). Patients with baseline NLR ≥4 and HLA-I homozygosity had the worst outcome (ORR 0; mPFS 1.4 months; mOS 1.8 months) among all. The association between NLR, HLA-I genotyping and clinical outcomes was independent of programmed death receptor ligand-1 expression. CONCLUSIONS: NLR and HLA-I genotyping could have predictive and prognostic value in patients with advanced ESCC receiving camrelizumab, and the combination of biomarkers may help to identify more patient benefit from immunotherapy.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Biomarcadores , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/genética , Genótipo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Linfócitos/patologia , Neutrófilos/patologia , Prognóstico , Estudos Prospectivos
15.
FASEB J ; 36(4): e22230, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35233835

RESUMO

Norisoboldine (NOR), an alkaloid isolated from Radix Lindera, was previously reported to promote the differentiation of regulatory T cells (Treg cells), an important subtype of lymphocytes capable of controlling autoimmune diseases. The present study was performed to explore the mechanism of NOR in the view of cellular metabolism. A global metabolomic analysis indicated that NOR preferentially altered the fatty acid oxidation (FAO) pathway and elevated the content of related metabolites during Treg cell differentiation. The detection of oxygen consumption rate (OCR) and mRNA expression of FAO-related enzymes demonstrated that NOR promoted FAO in the early stage of Treg cell differentiation. Consistently, pharmacological or genetic inhibition of FAO markedly diminished the induction of NOR on Treg cell differentiation. Furthermore, NOR was shown to elevate the level of acetyl-CoA derived from FAO and acetylation of lysine 27 on histone 3 (H3K27) at the Foxp3 promoter and CNS2 regions. A knockdown of CPT1, the rate-limiting enzyme of FAO, weakened the promotion of NOR on the development, acetyl-CoA level, and acetylation of H3K27 of Treg cells in vitro and in the mice with collagen-induced arthritis, and attenuated the anti-arthritic effect of NOR. These findings demonstrate that NOR induces the development of Treg cells through promoting FAO, therefore, facilitating gene transcription of Foxp3 via acetyl-CoA-mediated H3K27 acetylation modification, and FAO might serve as a novel target to induce Treg cell development.


Assuntos
Alcaloides/farmacologia , Ácidos Graxos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Histonas/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Acetilação , Animais , Diferenciação Celular/efeitos dos fármacos , Feminino , Fatores de Transcrição Forkhead/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Oxirredução , Regiões Promotoras Genéticas , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/fisiologia
16.
Diabetes Res Clin Pract ; 185: 109776, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35149165

RESUMO

BACKGROUND: In diet-induced arterial atherosclerosis, increased KCa3.1 channel was associated with atherosclerotic plaque progression and instability. Macrophages are involved in the formation of atherosclerotic plaques, and the release of inflammatory cytokines and oxygen free radicals promotes plaque progression. However, whether the macrophage KCa3.1 channel facilitates diabetes-accelerated atherosclerosis is still unclear. This study investigated atherosclerotic plaque in ApoE-/- mice regulated by the KCa3.1 channel. METHODS AND RESULTS: In vivo, blocking KCa3.1channel inhibit the development of the atherosclerotic lesion in diabetic ApoE-/- mice fed with a high-fat diet. In vitro, upregulation of KCa3.1 channel level occurred in RAW264.7 cells treated with HG plus ox-LDL in a time-dependent manner. Blocking KCa3.1 significantly reduced the uptake of ox-LDL in mice peritoneal macrophages. Further studies indicated the KCa3.1 siRNA and TRAM-34 (KCa3.1 inhibitor) attenuated the scavenger receptor CD36 expression via inhibiting STAT3 phosphorylation. CONCLUSION: Blockade of macrophage KCa3.1 channel inhibit cellular oxidized low-density lipoprotein accumulation and decrease proinflammation factors expression via STAT3/CD36 axis. This study provided a novel therapeutic target to reduce the risk of atherosclerosis development in diabetic patients.


Assuntos
Aterosclerose , Diabetes Mellitus , Placa Aterosclerótica , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Antígenos CD36/genética , Antígenos CD36/metabolismo , Humanos , Lipoproteínas LDL , Camundongos , Camundongos Knockout , Placa Aterosclerótica/etiologia , Fator de Transcrição STAT3/metabolismo
17.
Chemosphere ; 291(Pt 2): 132948, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34800504

RESUMO

Antibiotic and heavy metal residues in shallow lakes caused by aquaculture and human activities such as sewage discharge have attracted much attention and public concern. However, mechanisms by which these environmental pollutants affect the microorganism-mediated biogeochemical cycle are unknown. This study focused on the effects of antibiotics, heavy metal, and antibiotic resistance genes (ARGs) on denitrification in shallow lakes. The results showed that antibiotics and metal elements had inhibitory effects on denitrification, whereas AGRs exhibited stimulating effects. Specifically, the enrofloxacin concentration showed a significant negative correlation with the copy number of denitrifying bacteria, whereas the copy number of the ARGs sulI, sulII, and tetG showed significant positive correlations. In addition, tetG was closely related to the community structure of nirS-type denitrifiers, and nirS-type denitrifiers were significantly correlated with the potential denitrification rate (PDR). Furthermore, the ARGs sulI, sulII, and tetG were positively correlated with PDR (P < 0.05). By contrast, the metal elements arsenic, manganese, cobalt, and antimony were negatively correlated with the copy number of denitrifying bacteria. Arsenic was significantly correlated with the community composition of nirK-type denitrifiers, but nirK-type denitrifiers did not show a significant correlation with the PDR. This work extends our understanding of the effects of antibiotics and heavy metals on denitrification, but further studies are needed to determine the interaction effects of pollutants.


Assuntos
Lagos , Metais Pesados , Antibacterianos , Bactérias/genética , Desnitrificação , Genes Bacterianos , Humanos , Metais Pesados/análise
18.
Int J Mol Med ; 49(1)2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34850962

RESUMO

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that the OGD + HYSA and OGD + HYSA + AKBA plots in Fig. 5B on p. 1507 appeared to share a similar patterning with respect to many of the data points. The authors have re-examined their original data and realize that they made inadvertent errors during the assembly of this figure. The FCS files were read and analyzed by FlowJo cell analysis software. The authors have carefully examined the raw data (fcs files), and have identified the errors that occurred when applying the setting to all files and saving the resulting fluorescence data to dot-plot graphs. The corrected version of Fig. 5, showing the correct flow cytometric analysis data in Fig. 5B and a re-evaluation of the quantification of the data in the associated bar chart, is shown on the next page. Note that the errors made during the assembly of this figure did not affect the major conclusions reported in the paper. All the authors have agreed to this Corrigendum, and thank the Editor of International Journal of Molecular Medicine for allowing them the opportunity to publish this. The authors regret these errors went unnoticed prior to the publication of the paper, and apologize to the readership for any confusion that this may have caused. [the original article was published in International Journal of Molecular Medicine 37: 1501-1510, 2016; DOI: 10.3892/ijmm.2016.2571].

19.
Sci Rep ; 11(1): 24279, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930980

RESUMO

With the rapid aging of the population, coronary artery disease (CAD) has become one of the most fatal chronic diseases. However, the genetic mechanism of CAD is still unclear. The purpose of this study is to construct the lncRNA-miRNA-mRNA regulatory network for CAD diseases and systematically identify differentially expressed genes in patients with coronary heart disease. In this study, two lncRNA datasets (GSE69587 and GSE113079) and a microRNA dataset (GSE105449) which contained 393 and 38 CAD samples were selected. In addition, two mRNA datasets which named GSE113079 (98 CAD samples) and GSE9820 (8 CAD samples) were selected to search the differentially expressed genes (DEGs). By comparing the expression data between CAD and control samples, a total of 1111 lncRNAs, 2595 mRNAs and 22 miRNAs were identified. Based on the DEGs, a lncRNA-miRNA-mRNA ceRNA network was constructed to explore the hub nodes in CAD. In the ceRNA network, the lncRNAs KCNQ1OT1 and H19 showed high connectivity with the nine miRNAs. GO and KEGG results showed that genes in ceRNA networks were mainly involved in nitrogen compound metabolic process, PI3K-Akt signaling pathway and retrograde endocannabinoid signaling. These findings will improve the understanding of the occurrence and development mechanism of CAD.


Assuntos
Doença da Artéria Coronariana/genética , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Transdução de Sinais , Biomarcadores/metabolismo , Sistema Cardiovascular , Biologia Computacional , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Macrófagos/metabolismo , MicroRNAs/metabolismo , Permeabilidade , Fosfatidilinositol 3-Quinases/metabolismo
20.
Int J Mol Med ; 48(6)2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34633051

RESUMO

Subsequently to the publication of the above article, an interested reader drew to the authors' attention that some of the data panels shown in Fig. 6A on p. 90 appeared to contain overlapping data, such that the data may have been derived from the same original source where different experimental conditions were portrayed in the figure. The data that appeared to be overlapping were featured in the H/R and H/R+DH+Z panels (both the merged and the unmerged data panels). The authors have re-examined their original data and realize that they made an inadvertent error during the assembly of this figure. The corrected version of Fig. 6A, showing the correct TUNEL staining data for the H/R+DH+Z experiment, is shown on the next page. Note that the errors made during the assembly of this figure did not affect the major conclusions reported in the paper. All the authors have agreed to this Corrigendum, and thank the Editor of International Journal of Molecular Medicine for allowing them the opportunity to publish this. The authors regret these errors went unnoticed during the compilation of the figure in question, and apologize to the readership for any confusion that this may have caused. [the original article was published in International Journal of Molecular Medicine 38: 83-94, 2016; DOI: 10.3892/ijmm.2016.2584].

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...