Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
J Ethnopharmacol ; 335: 118606, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39038504

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Palmatine is a main bioactive alkaloid of Cortex Phellodendri, which has been commonly prescribed for the treatment of hyperuricemia (HUA) in China. The metabolites of palmatine were crucial to its prominent biological activity. 9-Hydroxy-8-oxypalmatine (9-OPAL) is a novel liver-mediated secondary oxymetabolite of palmatine. AIM OF THE STUDY: The current study was to assess the efficacy of 9-OPAL, a novel liver-mediated secondary oxymetabolite of palmatine derived from Cortex Phellodendri, in experimental HUA mouse model and further explore its underlying mechanism. MATERIALS AND METHODS: An in vitro metabolic experiment with oxypalmatine was carried out using liver samples. We separated and identified a novel liver metabolite, and investigated its anti-HUA effect in mice. HUA mice were induced by potassium oxonate and hypoxanthine daily for one week. After 1 h of modeling, mice were orally administered with different doses of 9-OPAL (5, 10 and 20 mg/kg). The pathological changes of the kidneys were evaluated using hematoxylin-eosin staining (H&E). The acute toxicity of 9-OPAL was assessed. The effects of 9-OPAL on serum levels of uric acid (UA), adenosine deaminase (ADA), xanthine oxidase (XOD), creatinine (CRE), blood urea nitrogen (BUN) and inflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA) or biochemical method. Furthermore, Western blot, quantitative real-time PCR (qRT-PCR) and molecular docking were used to investigate the effect of 9-OPAL on the expression of renal urate transporters and NLRP3 signaling pathway in HUA mice. RESULTS: 9-OPAL had been discovered to be a novel liver-mediated oxymetabolite of palmatine for the first time. Treatment with 9-OPAL significantly reduced the UA, CRE as well as BUN levels, and also effectively attenuated abnormal renal histopathological deterioration with favorable safety profile. Besides, 9-OPAL significantly decreased the serum and hepatic activities of XOD and ADA, dramatically inhibited the up-regulation of UA transporter protein 1 (URAT1) and glucose transporter protein 9 (GLUT9), and reversed the down-regulation of organic anion transporter protein 1 (OAT1). Additionally, 9-OPAL effectively mitigated the renal inflammatory markers (TNF-α, IL-1ß, IL-6 and IL-18), and downregulated the transcriptional and translational expressions of renal Nod-like receptor family pyrin domain containing 3 (NLRP3), caspase-1, apoptosis-associated speck-like (ASC) and IL-1ß in HUA mice. Molecular docking results revealed 9-OPAL bound firmly with XOD, OAT1, GLUT9, URAT1, NLRP3, caspase-1, ASC and IL-1ß. CONCLUSIONS: 9-OPAL was found to be a novel liver-mediated secondary metabolite of palmatine with favorable safety profile. 9-OPAL had eminent anti-hyperuricemic and renal-protective effects, and the mechanisms might be intimately associated with repressing XOD activities, modulating renal urate transporter expression and suppressing the NLRP3 inflammasome activation. Our investigation might also provide further experimental evidence for the traditional application of Cortex Phellodendri in the treatment of HUA.

2.
Int Immunopharmacol ; 138: 112634, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38971107

ABSTRACT

Metabolic dysfunction-associated steatohepatitis (MASH) is a severe metabolic dysfunction-associated steatotic liver disease (MASLD) characterized by abnormal hepatic steatosis and inflammation. Previous studies have shown that Patchouli alcohol (PA), the primary component of Pogostemonis Herba, can alleviate digestive system diseases. However, its protection against MASH remains unclear. This study explored the protective effects and underlying mechanism of PA against high-fat diet-induced MASH in rats. Results showed that PA considerably reduced body weight, epididymal fat, and liver index and attenuated liver histological injury in MASH rats. PA alleviated hepatic injury by inhibiting steatosis and inflammation. These effects are associated with the improvement of SREBP-1c- and PPARα-mediated lipid metabolism and inhibition of the STING-signaling pathway-mediated inflammatory response. Moreover, PA-inhibited hepatic endoplasmic reticulum (ER) stress and mitochondrial dysfunction, reducing SREBP-1c and STING expressions and enhance PPARα expression. PA treatment had the strongest effect on the regulation of mitogen fusion protein 2 (Mfn2) in inhibiting mitochondrial dysfunction. Mfn2 is an important structural protein for binding ERs and mitochondria to form mitochondria-associated ER membranes (MAMs). MASH-mediated disruption of MAMs was inhibited after PA treatment-induced Mfn2 activation. Therefore, the pharmacological effect of PA on MASH is mainly attributed to the inhibition of MAM disruption-induced hepatic steatosis and inflammation. The findings of this study may have implications for MASH treatment that do not neglect the role of Mfn2-mediated MAMs.


Subject(s)
Diet, High-Fat , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , PPAR alpha , Rats, Sprague-Dawley , Sesquiterpenes , Animals , Male , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/drug effects , Diet, High-Fat/adverse effects , Rats , Sesquiterpenes/therapeutic use , Sesquiterpenes/pharmacology , PPAR alpha/metabolism , Endoplasmic Reticulum Stress/drug effects , Liver/pathology , Liver/drug effects , Liver/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Fatty Liver/drug therapy , Fatty Liver/metabolism , Fatty Liver/pathology , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Lipid Metabolism/drug effects , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Pogostemon , Signal Transduction/drug effects
3.
Int Immunopharmacol ; 138: 112632, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38986300

ABSTRACT

Uric acid nephropathy (UAN), caused by a common metabolic disorder resulting from hyperuricemia (HUA), has an increasing incidence. Previous studies have shown that berberine (BBR) has clear urate-lowering and anti-inflammatory effects in UAN mice, but its mechanism needs to be further clarified. Therefore, Potassium Oxonate (PO) combined with hypoxanthine (HX) induced UAN mice model and MSU induced THP-1 cells polarization model were adopted to investigate the mechanism of BBR on UAN in terms of tissue distribution and molecular pharmacology. Study unveiled that BBR was first found to bind to red blood cells (RBCs), which were recognized and phagocytosed by monocytes, then recruited by the injured kidney. Subsequently, BBR was enriched and functional in damaged kidney. The results of in vivo experiments revealed that, BBR reduced UA, BUN, CRE levels as well as the release of TNF-α, IL-1ß, IL-18 and IL-6, and alleviated renal injury in UAN mice, as consistent with previous studies. Additionally, BBR decreased MCP-1 expression, while diminishing macrophage infiltration and decreasing M1 proportion as determined by RT-qPCR. In vitro experiments, demonstrated that MSU promoted inflammatory polarization of THP-1 cells, while BBR reduced synthesis of inflammatory factors and inhibited MSU-induced inflammatory polarization. These effects of BBR were dependent on AMPK activation along with indirect inhibition of NF-κB signaling pathway mediated. However, the anti-inflammatory and macrophage polarization regulation effects of BBR were completely reversed upon administration of Compound C, an AMPK inhibitor. Therefore, BBR ameliorated kidney injury via regulating macrophage polarization through AMPK, which has therapeutic potential for UAN patients.


Subject(s)
AMP-Activated Protein Kinases , Berberine , Cytokines , Kidney , Macrophages , Signal Transduction , Uric Acid , Animals , Humans , Male , Mice , AMP-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/pharmacology , Berberine/pharmacology , Cytokines/metabolism , Disease Models, Animal , Hyperuricemia/drug therapy , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Kidney Diseases/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Signal Transduction/drug effects , THP-1 Cells
4.
Cancer Cell Int ; 24(1): 195, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38835070

ABSTRACT

BACKGROUND: Investigating the unexplored territory of lncRNA m6A modification in colorectal cancer (CRC) vasculature, this study focuses on LINC01106 and YTHDF1. METHODS: Clinical assessments reveal upregulated LINC01106 promoting vascular generation via the miR-449b-5p-VEGFA pathway. RESULTS: YTHDF1, elevated in CRC tissues, emerges as an adverse prognostic factor. Functional experiments showcase YTHDF1's inhibitory effects on CRC cell dynamics. Mechanistically, Me-CLIP identifies m6A-modified LINC01106, validated as a YTHDF1 target through Me-RIP. CONCLUSIONS: This study sheds light on the YTHDF1-mediated m6A modification of LINC01106, presenting it as a key player in suppressing CRC vascular generation.

5.
Int J Nanomedicine ; 19: 5297-5316, 2024.
Article in English | MEDLINE | ID: mdl-38859955

ABSTRACT

Propose: Oxyberberine (OBB), one of the main metabolites of berberine derived from intestinal and erythrocyte metabolism, exhibits appreciable anti-hyperuricemic activity. However, the low water solubility and poor plasma concentration-effect relationship of OBB hamper its development and utilization. Therefore, an OBB-hydroxypropyl-ß-cyclodextrin (HP-ß-CD) supersaturated drug delivery system (SDDS) was prepared and characterized in this work. Methods: OBB-HP-ß-CD SDDS was prepared using the ultrasonic-solvent evaporation method and characterized. Additionally, the in vitro and in vivo release experiments were conducted to assess the release kinetics of OBB-HP-ß-CD SDDS. Subsequently, the therapeutic efficacy of OBB-HP-ß-CD SDDS on hyperuricemia (HUA) was investigated by means of histopathological examination and evaluation of relevant biomarkers. Results: The results of FT-IR, DSC, PXRD, NMR and molecular modeling showed that the crystallized form of OBB was transformed into an amorphous OBB-HP-ß-CD complex. Dynamic light scattering indicated that this system was relatively stable and maintained by formation of nanoaggregates with an average diameter of 23 nm. The dissolution rate of OBB-HP-ß-CD SDDS was about 5 times higher than that of OBB raw material. Furthermore, the AUC0-t of OBB-HP-ß-CD SDDS (10.882 µg/mL*h) was significantly higher than that of the raw OBB counterpart (0.701 µg/mL*h). The oral relative bioavailability of OBB-HP-ß-CD SDDS was also enhanced by 16 times compared to that of the raw material. Finally, in vivo pharmacodynamic assay showed the anti-hyperuricemic potency of OBB-HP-ß-CD SDDS was approximately 5-10 times higher than that of OBB raw material. Conclusion: Based on our findings above, OBB-HP-ß-CD SDDS proved to be an excellent drug delivery system for increasing the solubility, dissolution, bioavailability, and anti-hyperuricemic potency of OBB.


Subject(s)
Berberine , Animals , Berberine/pharmacokinetics , Berberine/chemistry , Berberine/administration & dosage , Berberine/pharmacology , Male , 2-Hydroxypropyl-beta-cyclodextrin/chemistry , 2-Hydroxypropyl-beta-cyclodextrin/pharmacokinetics , Hyperuricemia/drug therapy , Hyperuricemia/blood , Drug Delivery Systems/methods , Solubility , Nanoparticles/chemistry , Rats , Rats, Sprague-Dawley , Drug Liberation , Particle Size , Biological Availability , Uric Acid/chemistry , Uric Acid/blood
6.
Eur J Pharmacol ; 968: 176433, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38369273

ABSTRACT

The promotion of excess low-density lipoprotein (LDL) clearance stands as an effective clinical approach for treating hyperlipidemia. Tetrahydroberberine, a metabolite of berberine, exhibits superior bioavailability compared to berberine and demonstrates a pronounced hypolipidemic effect. Despite these characteristics, the impact of tetrahydroberberine on improving excessive LDL clearance in hyperlipidemia has remained unexplored. Thus, this study investigates the potential effects of tetrahydroberberine on high-fat diet-induced hyperlipidemia in mice. The findings reveal that tetrahydroberberine exerts a more potent lipid-lowering effect than berberine, particularly concerning LDL-cholesterol in hyperlipidemic mice. Notably, tetrahydroberberine significantly reduces serum levels of upstream lipoproteins, including intermediate-density lipoprotein (IDL) and very low-density lipoprotein, by promoting their conversion to LDL. This reduction is further facilitated by the upregulation of hepatic LDL receptor expression induced by tetrahydroberberine. Intriguingly, tetrahydroberberine enhances the apolipoprotein E (ApoE)/apolipoprotein B100 (ApoB100) ratio, influencing lipoprotein assembly in the serum. This effect is achieved through the activation of the efflux of ApoE-containing cholesterol in the liver. The ApoE/ApoB100 ratio exhibits a robust negative correlation with serum levels of LDL and IDL, indicating its potential as a diagnostic indicator for hyperlipidemia. Moreover, tetrahydroberberine enhances hepatic lipid clearance without inducing lipid accumulation in the liver and alleviates existing liver lipid content. Importantly, no apparent hepatorenal toxicity is observed following tetrahydroberberine treatment for hyperlipidemia. In summary, tetrahydroberberine demonstrates a positive impact against hyperlipidemia by modulating lipoprotein assembly-induced clearance of LDL and IDL. The ApoE/ApoB100 ratio emerges as a promising diagnostic indicator for hyperlipidemia, showcasing the potential clinical significance of tetrahydroberberine in lipid management.


Subject(s)
Berberine , Berberine/analogs & derivatives , Hyperlipidemias , Mice , Animals , Lipoproteins, IDL/metabolism , Berberine/pharmacology , Berberine/therapeutic use , Diet, High-Fat/adverse effects , Triglycerides , Cholesterol/metabolism , Apolipoproteins E/genetics , Cholesterol, LDL , Liver/metabolism
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167051, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38336103

ABSTRACT

Currently, it is acknowledged that gout is caused by uric acid (UA). However, some studies have revealed no correlation between gout and UA levels, and growing evidence suggests that 2,8-dihydroxyadenine (2,8-DHA), whose structural formula is similar to UA but is less soluble, may induce gout. Hence, we hypothesized that uroliths from hyperuricemia (HUA) patients, which is closely associated with gout, may contain 2,8-DHA. In this study, 2,8-DHA in uroliths and serum of HUA patients were determined using HPLC. Moreover, bioinformatics was used to investigate the pathogenic mechanisms of 2,8-DHA nephropathy. Subsequently, a mouse model of 2,8-DHA nephropathy established by the gavage administration of adenine, as well as a model of injured HK-2 cells induced by 2,8-DHA were used to explore the pathogenesis of 2,8-DHA nephropathy. Interestingly, 2,8-DHA could readily deposit in the cortex of the renal tubules, and was found in the majority of these HUA patients. Additionally, the differentially expressed genes between 2,8-DHA nephropathy mice and control mice were found to be involved in inflammatory reactions. Importantly, CCL2 and IL-1ß genes had the maximum degree, closeness, and betweenness centrality scores. The expressions of CCL2 and IL-1ß genes were significantly increased in the serum of 24 HUA patients with uroliths, indicating that they may be significant factors for 2,8-DHA nephropathy. Further analysis illustrated that oxidative damage and inflammation were the crucial processes of 2,8-DHA renal injury, and CCL2 and IL-1ß genes were verified to be essential biomarkers for 2,8-DHA nephropathy. These findings revealed further insights into 2,8-DHA nephropathy, and provided new ideas for its diagnosis and treatment.


Subject(s)
Adenine/analogs & derivatives , Gout , Hyperuricemia , Kidney Diseases , Humans , Mice , Animals , Hyperuricemia/metabolism , Kidney/metabolism , Uric Acid/metabolism
8.
Neural Netw ; 172: 106075, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38278092

ABSTRACT

The SSVEP-based paradigm serves as a prevalent approach in the realm of brain-computer interface (BCI). However, the processing of multi-channel electroencephalogram (EEG) data introduces challenges due to its non-Euclidean characteristic, necessitating methodologies that account for inter-channel topological relations. In this paper, we introduce the Dynamic Decomposition Graph Convolutional Neural Network (DDGCNN) designed for the classification of SSVEP EEG signals. Our approach incorporates layerwise dynamic graphs to address the oversmoothing issue in Graph Convolutional Networks (GCNs), employing a dense connection mechanism to mitigate the gradient vanishing problem. Furthermore, we enhance the traditional linear transformation inherent in GCNs with graph dynamic fusion, thereby elevating feature extraction and adaptive aggregation capabilities. Our experimental results demonstrate the effectiveness of proposed approach in learning and extracting features from EEG topological structure. The results shown that DDGCNN outperforms other state-of-the-art (SOTA) algorithms reported on two datasets (Dataset 1: 54 subjects, 4 targets, 2 sessions; Dataset 2: 35 subjects, 40 targets). Additionally, we showcase the implementation of DDGCNN in the context of synchronized BCI robotic fish control. This work represents a significant advancement in the field of EEG signal processing for SSVEP-based BCIs. Our proposed method processes SSVEP time domain signals directly as an end-to-end system, making it easy to deploy. The code is available at https://github.com/zshubin/DDGCNN.


Subject(s)
Brain-Computer Interfaces , Humans , Evoked Potentials, Visual , Neural Networks, Computer , Algorithms , Electroencephalography/methods , Photic Stimulation
9.
Org Lett ; 26(3): 676-680, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38235713

ABSTRACT

A palladium-catalyzed divergent cascade decarboxylative annulation of aryl iodides and α-oxocarboxylic acids using norbornene (NBE) derivatives as a controlled switch is reported. When NBE is used as a mediator, fluorenones are synthesized with moderate to excellent yields via a Catellani reaction that involves sequential ortho-C-H arylation and ipso-decarboxylative acylation of aryl iodides. Employing oxanorbornadiene (ONBD) instead of NBE enables the assembly of dibenzo[a,c]cycloheptenones by a retro-Diels-Alder reaction rather than the release of an ONBD. Additionally, the synthetic utility of this method is demonstrated by the diversification of the products.

10.
Clin. transl. oncol. (Print) ; 25(10): 2972-2982, oct. 2023.
Article in English | IBECS | ID: ibc-225078

ABSTRACT

Objective Great success has been achieved in CAR-T cell immunotherapy in the treatment of hematological tumors. However, it is particularly difficult in solid tumors, because CAR-T is difficult to enter interior and exert long-term stable immune effects. Dendritic cells (DCs) can not only present tumor antigens but also promote the infiltration of T cells. Therefore, CAR-T cells with the help of DC vaccines are a reliable approach to treat solid tumors. Methods To test whether DC vaccine could promote CAR-T cell therapy in solid tumors, DC vaccine was co-cultured with MSLN CAR-T cells. The in vitro effects of DC vaccine on CAR-T were assessed by measuring cell proliferation, cell differentiation, and cytokine secretion. Effects of DC vaccine on CAR-T were evaluated using mice with subcutaneous tumors in vivo. The infiltration of CAR-T was analyzed using immunofluorescence. The persistence of CAR-T in mouse blood was analyzed using real-time quantitative PCR. Results The results showed that DC vaccine significantly enhanced the proliferation potential of MSLN CAR-T cells in vitro. DC vaccines not only promoted the infiltration of CAR-T cells, but also significantly improved the persistence of CAR-T in solid tumors in vivo. Conclusion In conclusion, this study has demonstrated that DC vaccine can promote CAR-T therapy in solid tumors, which provides the possibility of widespread clinical application of CAR-T cells in the future (AU)


Subject(s)
Animals , Mice , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Chimeric Antigen , Vaccines , T-Lymphocytes
11.
Article in English | MEDLINE | ID: mdl-37756172

ABSTRACT

The classification problem for short time-window steady-state visual evoked potentials (SSVEPs) is important in practical applications because shorter time-window often means faster response speed. By combining the advantages of the local feature learning ability of convolutional neural network (CNN) and the feature importance distinguishing ability of attention mechanism, a novel network called AttentCNN is proposed to further improve the classification performance for short time-window SSVEP. Considering the frequency-domain features extracted from short time-window signals are not obvious, this network starts with the time-domain feature extraction module based on the filter bank (FB). The FB consists of four sixth-order Butterworth filters with different bandpass ranges. Then extracted multimodal features are aggregated together. The second major module is a set of residual squeeze and excitation blocks (RSEs) that has the ability to improve the quality of extracted features by learning the interdependence between features. The final major module is time-domain CNN (tCNN) that consists of four CNNs for further feature extraction and followed by a fully connected (FC) layer for output. Our designed networks are validated over two large public datasets, and necessary comparisons are given to verify the effectiveness and superiority of the proposed network. In the end, in order to demonstrate the application potential of the proposed strategy in the medical rehabilitation field, we design a novel five-finger bionic hand and connect it to our trained network to achieve the control of bionic hand by human brain signals directly. Our source codes are available on Github: https://github.com/JiannanChen/AggtCNN.git.

13.
J Air Waste Manag Assoc ; 73(8): 638-648, 2023 08.
Article in English | MEDLINE | ID: mdl-37431990

ABSTRACT

Leachate from landfills can be a significant challenge to manage and treat due to conventional contaminants. The addition of emerging contaminants such as per- and polyfluorinatedalkyl substances (PFASs) makes treatment even more complex. PFASs enter landfills through consumer waste and have been detected in landfill leachates at varying concentrations. The design and decision-making on leachate treatment require essential information since it depends on local factors, e.g. climate, proximity to wastewater treatment plants, and waste type. This study conducted a survey on actively operated public municipal solid waste (MSW) landfills in the Eastern and Northwestern regions of the US to understand the current leachate treatment practices and views from public MSW landfill managers on PFAS treatment. The survey aims to explore the possible adaptations from the industry to the pending regulatory guidelines for the potential PFASs treatment. Results show the majority of the landfills are currently using off-site disposal (72% of the responses), followed by complete onsite treatment (18% of the responses) and pre-treatment onsite and off-site disposal methods (10% of the responses). The factors that guided the selection of treatment methods included climate, economics, and future regulations. Evaporation and recirculation were the most prevalent onsite treatment technologies for public landfills, which reduced the leachate quantity for treatment. The public landfills expressed awareness of the potential impact of PFASs on the changes in leachate treatment. The current state-level regulation, potential federal PFAS regulation, and treatment costs are raising awareness of the onsite treatment for PFASs. The results of this study will benefit the improvement of PFAS awareness and provide critical information that will directly affect the leachate treatment process for PFASs.Implications: This study presents a survey on the current leachate treatment process in the public municipal solid waste landfills in the eastern and northwestern U.S. and their potential process improvement on the impact of PFASs. This study is relevant to the topic of the JA&WMA because the research falls directly within the scope of this journal, and it documents the leachate treatment of landfills, and the results of this study will immediately contribute to our understanding of the waste treatment, benefiting the improvement of PFASs awareness, and providing critical information that will directly affect the leachate treatment process.


Subject(s)
Fluorocarbons , Refuse Disposal , Water Pollutants, Chemical , Solid Waste/analysis , Refuse Disposal/methods , Waste Disposal Facilities , Water Pollutants, Chemical/analysis , Fluorocarbons/analysis
14.
Immunology ; 170(3): 388-400, 2023 11.
Article in English | MEDLINE | ID: mdl-37501391

ABSTRACT

It is well known that chimeric antigen receptor T-cell immunotherapy (CAR-T-cell immunotherapy) has excellent therapeutic effect in haematological tumours, but it still faces great challenges in solid tumours, including inefficient T-cell tumour infiltration and poor functional persistence. Flap structure-specific endonuclease 1 (FEN1), highly expressed in a variety of cancer cells, plays an important role in both DNA replication and repair. Previous studies have reported that FEN1 inhibition is an effective strategy for cancer treatment. Therefore, we hypothesized whether FEN1 inhibitors combined with CAR-T-cell immunotherapy would have a stronger killing effect on solid tumours. The results showed that low dose of FEN1 inhibitors SC13 could induce an increase of double-stranded broken DNA (dsDNA) in the cytoplasm. Cytosolic dsDNA can activate the cyclic GMP-AMP synthase-stimulator of interferon gene signalling pathway and increase the secretion of chemokines. In vivo, under the action of FEN1 inhibitor SC13, more chemokines were produced at solid tumour sites, which promoted the infiltration of CAR-T cells and improved anti-tumour immunity. These findings suggest that FEN1 inhibitors could enable CAR-T cells to overcome poor T-cell infiltration and improve the treatment of solid tumours.


Subject(s)
Neoplasms , Humans , Signal Transduction , DNA , T-Lymphocytes/metabolism , Nucleotidyltransferases/genetics , Chemokines , Flap Endonucleases/genetics , Flap Endonucleases/metabolism
15.
Front Immunol ; 14: 1175920, 2023.
Article in English | MEDLINE | ID: mdl-37359558

ABSTRACT

Chimeric antigen receptor T cell therapy has become an important immunotherapeutic tool for overcoming cancers. However, the efficacy of CAR-T cell therapy in solid tumors is relatively poor due to the complexity of the tumor microenvironment and inhibitory immune checkpoints. TIGIT on the surface of T cells acts as an immune checkpoint by binding to CD155 on the tumor cells' surface, thereby inhibiting tumor cell killing. Blocking TIGIT/CD155 interactions is a promising approach in cancer immunotherapy. In this study, we generated anti-MLSN CAR-T cells in combination with anti-α-TIGIT for solid tumors treatment. The anti-α-TIGIT effectively enhanced the efficacy of anti-MLSN CAR-T cells on the killing of target cells in vitro. In addition, we genetically engineered anti-MSLN CAR-T cells with the capacity to constitutively produce TIGIT-blocking single-chain variable fragments. Our study demonstrated that blocking TIGIT significantly promoted cytokine release to augment the tumor-killing effect of MT CAR-T cells. Moreover, the self-delivery of TIGIT-blocking scFvs enhanced the infiltration and activation of MT CAR-T cells in the tumor microenvironments to achieve better tumor regression in vivo. These results suggest that blocking TIGIT effectively enhances the anti-tumor effect of CAR-T cells and suggest a promising strategy of combining CAR-T with immune checkpoints blockade in the treatment of solid tumors.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Immunotherapy/methods , T-Lymphocytes , Immunotherapy, Adoptive/methods , Tumor Microenvironment , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
16.
J Agric Food Chem ; 71(27): 10338-10348, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37389466

ABSTRACT

Hepatic endothelial function is central to the development of nonalcoholic steatohepatitis (NASH). Curcumin (Cur) is reportedly hepatoprotective, however, it remains unknown whether Cur improves hepatic endothelial function in NASH. Additionally, the poor bioavailability of Cur renders it difficult to elucidate its hepatoprotective effect, hence, its biotransformation should be considered. Herein, we investigated the effects and mechanisms of Cur and its bioconversion on hepatic endothelial function against high-fat diet-induced NASH in rats. The results revealed that Cur improved hepatic lipid accumulation, inflammation, and endothelial dysfunction by inhibiting NF-κB and PI3K/Akt/HIF-1α pathways, however, these effects were weakened via antibiotic addition, which was closely related to reduced tetrahydrocurcumin (THC) produce in the liver and intestinal content. Moreover, THC exerted a better effect than Cur on restoring liver sinusoidal endothelial cells function to attenuate steatosis and injury in L02 cells. Thus, these findings indicate that the effect of Cur on NASH is closely related to hepatic endothelial function improvement with intestinal microbial biotransformation.


Subject(s)
Curcumin , Non-alcoholic Fatty Liver Disease , Rats , Animals , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Curcumin/metabolism , Diet, High-Fat/adverse effects , Phosphatidylinositol 3-Kinases/metabolism , Endothelial Cells/metabolism , Liver/metabolism , Biotransformation , Mice, Inbred C57BL
17.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2405-2416, 2023 10.
Article in English | MEDLINE | ID: mdl-37193772

ABSTRACT

Hyperuricemia (HUA) is a common chronic metabolic disease that can cause renal failure and even death in severe cases. Berberine (BBR) is an isoquinoline alkaloid derived from Phellodendri Cortex with strong antioxidant, anti-inflammatory, and anti-apoptotic properties. The purpose of this study was to investigate the protective effects of berberine (BBR) against uric acid (UA)-induced HK-2 cells and unravel their regulatory potential mechanisms. The CCK8 assay was carried out to detect cell viability. The expression levels of inflammatory factors interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) and Lactate dehydrogenase (LDH) were measured using Enzyme-linked immunosorbent assays (ELISA). The expression of the apoptosis-related protein (cleaved-Caspase3, cleaved-Caspase9, BAX, BCL-2) was detected by western blot. The effects of BBR on the activities of the NOD-like receptor family pyrin domain containing 3 (NLRP3) and the expression of the downstream genes were determined by RT-PCR and western blot in HK-2 cells. From the data, BBR significantly reversed the up-regulation of inflammatory factors (IL-1ß, IL-18) and LDH. Furthermore, BBR down-regulated protein expression of pro-apoptotic proteins BAX, cleaved caspase3 (cl-Caspase3), cleaved caspase9 (cl-Caspase9), and enhanced the expression of antiapoptotic protein BCL-2. Simultaneously, BBR inhibited the activated NLPR3 and reduced the mRNA levels of NLRP3, Caspase1, IL-18, and IL-1ß. Also, BBR attenuated the expression of NLRP3 pathway-related proteins (NLRP3, ASC, Caspase1, cleaved-Caspase1, IL-18, IL-1ß, and GSDMD). Furthermore, specific NLRP3-siRNA efficiently blocked UA-induced the level of inflammatory factors (IL-1ß, IL-18) and LDH and further inhibited activated NLRP3 pathway. Collectively, our results suggested that BBR can alleviate cell injury induced by UA. The underlying unctionary mechanism may be through the NLRP3 signaling pathway.


Subject(s)
Berberine , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/pharmacology , Uric Acid/metabolism , Inflammasomes/genetics , Berberine/pharmacology , bcl-2-Associated X Protein , Signal Transduction
18.
Clin Transl Oncol ; 25(10): 2972-2982, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37079211

ABSTRACT

OBJECTIVE: Great success has been achieved in CAR-T cell immunotherapy in the treatment of hematological tumors. However, it is particularly difficult in solid tumors, because CAR-T is difficult to enter interior and exert long-term stable immune effects. Dendritic cells (DCs) can not only present tumor antigens but also promote the infiltration of T cells. Therefore, CAR-T cells with the help of DC vaccines are a reliable approach to treat solid tumors. METHODS: To test whether DC vaccine could promote CAR-T cell therapy in solid tumors, DC vaccine was co-cultured with MSLN CAR-T cells. The in vitro effects of DC vaccine on CAR-T were assessed by measuring cell proliferation, cell differentiation, and cytokine secretion. Effects of DC vaccine on CAR-T were evaluated using mice with subcutaneous tumors in vivo. The infiltration of CAR-T was analyzed using immunofluorescence. The persistence of CAR-T in mouse blood was analyzed using real-time quantitative PCR. RESULTS: The results showed that DC vaccine significantly enhanced the proliferation potential of MSLN CAR-T cells in vitro. DC vaccines not only promoted the infiltration of CAR-T cells, but also significantly improved the persistence of CAR-T in solid tumors in vivo. CONCLUSION: In conclusion, this study has demonstrated that DC vaccine can promote CAR-T therapy in solid tumors, which provides the possibility of widespread clinical application of CAR-T cells in the future.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Vaccines , Mice , Animals , T-Lymphocytes , T-Cell Exhaustion , Neoplasms/therapy , Immunotherapy, Adoptive/methods
19.
Front Pharmacol ; 14: 1136076, 2023.
Article in English | MEDLINE | ID: mdl-36895947

ABSTRACT

Background: Brucea javanica (L.) Merr, has a long history to be an anti-dysentery medicine for thousand of years, which is commonly called "Ya-Dan-Zi" in Chinese. The common liquid preparation of its seed, B. javanica oil (BJO) exerts anti-inflammatory action in gastrointestinal diseases and is popularly used as an antitumor adjuvant in Asia. However, there is no report that BJO has the potential to treat 5-Fluorouracil (5-FU)-induced chemotherapeutic intestinal mucosal injury (CIM). Aim of the study: To test the hypothesis that BJO has potential intestinal protection on intestinal mucosal injury caused by 5-FU in mice and to explore the mechanisms. Materials and methods: Kunming mice (half male and female), were randomly divided into six groups: normal group, 5-FU group (5-FU, 60 mg/kg), LO group (loperamide, 4.0 mg/kg), BJO group (0.125, 0.25, 0.50 g/kg). CIM was induced by intraperitoneal injection of 5-FU at a dose of 60 mg/kg/day for 5 days (from day 1 to day 5). BJO and LO were given orally 30 min prior to 5-FU administration for 7 days (from day 1 to day 7). The ameliorative effects of BJO were assessed by body weight, diarrhea assessment, and H&E staining of the intestine. Furthermore, the changes in oxidative stress level, inflammatory level, intestinal epithelial cell apoptosis, and proliferation, as well as the amount of intestinal tight junction proteins were evaluated. Finally, the involvements of the Nrf2/HO-1 pathway were tested by western blot. Results: BJO effectively alleviated 5-FU-induced CIM, as represented by the improvement of body weight, diarrhea syndrome, and histopathological changes in the ileum. BJO not only attenuated oxidative stress by upregulating SOD and downregulating MDA in the serum, but also reduced the intestinal level of COX-2 and inflammatory cytokines, and repressed CXCL1/2 and NLRP3 inflammasome activation. Moreover, BJO ameliorated 5-FU-induced epithelial apoptosis as evidenced by the downregulation of Bax and caspase-3 and the upregulation of Bcl-2, but enhanced mucosal epithelial cell proliferation as implied by the increase of crypt-localized proliferating cell nuclear antigen (PCNA) level. Furthermore, BJO contributed to the mucosal barrier by raising the level of tight junction proteins (ZO-1, occludin, and claudin-1). Mechanistically, these anti-intestinal mucositis pharmacological effects of BJO were relevant for the activation of Nrf2/HO-1 in the intestinal tissues. Conclusion: The present study provides new insights into the protective effects of BJO against CIM and suggests that BJO deserves to be applied as a potential therapeutic agent for the prevention of CIM.

20.
Food Funct ; 14(6): 2822-2835, 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36866793

ABSTRACT

As the final hydrogenated metabolite of curcumin, octahydrocurcumin (OHC) exhibits increased powerful bioactivities. The chiral and symmetric chemical structure indicated that there were two OHC stereoisomers, (3R,5S)-octahydrocurcumin (Meso-OHC) and (3S,5S)-octahydrocurcumin ((3S,5S)-OHC), which may induce different effects on metabolic enzymes and bioactivities. Thus, we detected OHC stereoisomers from rat metabolites (blood, liver, urine and feces) after oral administration of curcumin. In addition, OHC stereoisomers were prepared and then their different influences on cytochrome P450 enzymes (CYPs) and UDP-glucuronyltransferases (UGTs) in L-02 cells were tested to explore the potential interaction and different bioactivities. Our results proved that curcumin could be metabolised into OHC stereoisomers first. In addition, Meso-OHC and (3S,5S)-OHC exhibited slight induction or inhibition effects on CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP3A4 and UGTs. Furthermore, Meso-OHC exhibited more intensive inhibition toward CYP2E1 expression than (3S,5S)-OHC, ascribed to the different mode of binding to the enzyme protein (P < 0.05), which finally induced more effective liver protection effects in acetaminophen-induced L-02 cell injury.


Subject(s)
Curcumin , Cytochrome P-450 CYP2E1 , Rats , Animals , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Curcumin/chemistry , Stereoisomerism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Microsomes, Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL