Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.720.361
Filtrar
1.
Carbohydr Polym ; 323: 121368, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37940266

RESUMO

Hydrocolloids are important food additives and have potential regulatory effects on gut microbiota. The development of colitis is closely related to changes in gut microbiota. The effect of food hydrocolloids on the structure of the gut microbiota and their impact on colitis has not been well investigated. Therefore, this study investigated the effects of four hydrocolloids (carrageenan, guar gum, xanthan gum, and pectin) on colitis, and explored their regulatory effects on gut microbiota. The results indicated that pectin and guar effectively alleviated body weight loss and disease activity index, reduced inflammatory cytokine levels, and promoted short-chain fatty acids (SCFAs) production. They increased the abundance of Akkermansia muciniphila, Oscillospira, and Lactobacillus, and Akkermansia abundance had a negative correlation with the severity of colitis. In contrast, carrageenan and xanthan gum did not significantly improve colitis, and carrageenan reduced the production of SCFAs. Both carrageenan and xanthan gum increased the abundance of Ruminococcus gnavus, and Ruminococcus abundance was positively correlated with the severity of colitis. These findings suggest that food additives have an impact on host health and provide guidance for the diet of patients with colitis.


Assuntos
Colite , Microbioma Gastrointestinal , Humanos , Animais , Camundongos , Carragenina/química , Colite/induzido quimicamente , Colite/tratamento farmacológico , Aditivos Alimentares/química , Pectinas/farmacologia , Ácidos Graxos Voláteis , Coloides/farmacologia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
2.
Carbohydr Polym ; 323: 121425, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37940297

RESUMO

Islet transplantation to restore endogenous insulin secretion is a promising therapy for type 1 diabetes in clinic. However, host immune rejection seriously limits the survival of transplanted islets. Despite of the various encapsulation strategies and materials developed so far to provide immune isolation for transplanted islets, long-term blood glucose regulation is still difficult due to the inherent defects of the encapsulation materials. Herein, a novel islet-encapsulation composite material with low immunogenicity, good biocompatibility and excellent stability is reported. Specifically, chitosan (CS) microgels (diameter: ∼302 µm) are prepared via Michael addition reaction between maleimide grafted chitosan (CS-Mal) and thiol grafted chitosan (CS-NAC) in droplet-based microfluidic device, and then zwitterionic surface layer is constructed on CS microgel surface by covalent binding between maleimide groups on CS and thiol groups on thiol modified carboxymethyl cellulose (CMC-SH). The as-formed carboxymethyl cellulose coated chitosan (CS@CMC) microgels show not only long-term stability in vivo owing to the non-biodegradability of CMC, but also fantastic anti-adsorption and antifibrosis because of the stable zwitterionic surface layer. As a result, islets encapsulated in the CS@CMC microgels exhibit high viability and good insulin secretion function in vivo, and long-term blood glucose regulation is achieved for 180 days in diabetic mice post-transplantation.


Assuntos
Quitosana , Diabetes Mellitus Experimental , Ilhotas Pancreáticas , Microgéis , Camundongos , Animais , Glicemia/metabolismo , Quitosana/metabolismo , Carboximetilcelulose Sódica/metabolismo , Diabetes Mellitus Experimental/metabolismo , Maleimidas , Compostos de Sulfidrila , Ilhotas Pancreáticas/metabolismo
3.
Methods Mol Biol ; 2725: 121-129, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37856021

RESUMO

Volume electron microscopy technologies such as serial block face scanning electron microscopy (SBF-SEM) allow the characterization of tissue organization and cellular content in three dimensions at nanoscale resolution. Here, we describe the procedure to process and image an air-liquid interface culture of human or mouse airway epithelial cells for visualization of the multiciliated epithelium by SBF-SEM in vertical or horizontal cross section.


Assuntos
Imageamento Tridimensional , Microscopia Eletrônica de Volume , Animais , Humanos , Camundongos , Imageamento Tridimensional/métodos , Microscopia Eletrônica de Varredura , Epitélio , Células Epiteliais
4.
Methods Mol Biol ; 2725: 167-180, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37856024

RESUMO

Multiciliated cells (MCC) display on their apical surface hundreds of beating cilia that propel physiological fluids. They line brain ventricles where they propel the cerebrospinal liquid, airways where they clear mucus and pathogens and reproductive ducts where they concentrate the sperm in males or drive the egg along the oviducts in females. Motile cilia are nucleated from basal bodies which are modified centrioles. MCC therefore evade centriole archetypal duplication program to make several hundreds and nucleate an identical number of motile cilia. Defects in this centriole amplification process lead to severe human pathologies called "ciliary aplasia" or "acilia syndrome" and more recently renamed "reduced generation of motile cilia" (RGMC). Patients with this syndrome present frequent hydrocephaly, lung failure, and subfertility. In this manuscript, we describe the protocol we developed and optimized over the years to live image the centriole amplification dynamics. We explain why mouse brain MCC is a good model and provide the tips to enable successful spatially and temporally resolved monitoring of this massive organelle reorganization.


Assuntos
Centríolos , Sêmen , Feminino , Camundongos , Animais , Humanos , Masculino , Organelas , Cílios/fisiologia , Encéfalo
5.
Methods Mol Biol ; 2725: 199-212, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37856026

RESUMO

Biological complexity is achieved through elaborate interactions between relatively few individual components. Affinity purification (AP) has allowed these networks of protein-protein interactions that regulate key biological processes to be interrogated systematically. In order to perform these studies at the required scale, easily transfectable immortalized cell lines have typically been used. Gene-editing now affords the systematic creation of isogenic mouse models carrying endogenous tags for affinity proteomics. This may allow protein-protein interactions to be characterized in the appropriate tissue for a particular biological process or disease phenotype under physiological conditions, and for interaction landscapes to be compared across tissues. Here we demonstrate application to intraflagellar transport (IFT) proteins, which are WD40-domain-containing proteins that are essential for the formation and function of all types of cilia. We describe a method to generate mice with an endogenous C-terminal streptavidin/FLAG tag, using Ift80 as an example, and demonstrate the successful implementation of AP in this model. This method can easily be adapted for N- and C-terminal tagging of many other proteins in vivo.


Assuntos
Proteínas de Transporte , Proteínas , Animais , Camundongos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Estreptavidina/metabolismo , Proteínas/metabolismo , Transporte Biológico , Cílios/metabolismo
6.
Methods Mol Biol ; 2725: 239-250, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37856029

RESUMO

The wall of the ventricular system within the neuraxis is lined almost entirely by E1 ependymal cells, each of which projects multiple motile cilia from their apical surface into the cerebrospinal fluid (CSF). This specialized layer of E1 cells constitutes the border between the CSF and the brain interstitial fluid (BIF), and by controlling influx and efflux across the CSF to BIF interface, it is increasingly recognized to play an integral role in modulating and maintaining the brain microenvironment. The motile cilia have been shown to be responsive to changes in the CSF microenvironment, and while the physiological role of this mechanism remains incompletely understood, manipulating this control mechanism may influence the brain microenvironment potentially opening a new frontier in therapeutic intervention.In this paper, we describe our techniques for preparing organotypic slices from the murine brain parenchyma and establishing cell cultures of multiciliated ependymal cells from mouse and rat neonatal brain tissue. Our methodology generates a functional readout of ciliary function, specifically high-speed video microscopy (HSVM) enables the quantification of ciliary beat frequency (CBF), and characterization of ciliary beat pattern.


Assuntos
Cílios , Epêndima , Ratos , Camundongos , Animais , Cílios/metabolismo , Microscopia de Vídeo , Encéfalo , Técnicas de Cultura de Células
7.
Int J Cancer ; 154(1): 169-179, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37611176

RESUMO

Intracranial metastases are common in nonsmall-cell lung cancer (NSCLC) patients, whose prognosis is very poor. In addition, intracranial progression is common during systemic treatments due to the inability to penetrate central nervous system (CNS) barriers, whereas the intracranial effects of cancer immunotherapies remain unclear. We analyzed clinical data to evaluate the frequency of intracranial progression in advanced NSCLC patients treated with PD-1 blockade therapies compared with those treated without PD-1 blockade therapies, and found that the frequency of intracranial progression in advanced NSCLC patients treated with PD-1 blockade therapies was significantly lower than that in patients treated with cytotoxic chemotherapies. In murine models, intracranial rechallenged tumors after initial rejection by PD-1 blockade were suppressed. Accordingly, long-lived memory precursor effector T cells and antigen-specific T cells were increased by PD-1 blockade in intracranial lesions. However, intracranial rechallenged different tumors are not suppressed. Our results indicate that cancer immunotherapies can prevent intracranial progression, maintaining long-term effects intracranially as well as systemically. If intracranial recurrence occurs during the treatment with PD-1 blockade therapies, aggressive local therapies could be worthwhile.


Assuntos
Neoplasias Encefálicas , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Neoplasias Pulmonares/patologia , Receptor de Morte Celular Programada 1 , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Prognóstico , Imunoterapia/métodos , Neoplasias Encefálicas/terapia , Antígeno B7-H1
8.
Sci Total Environ ; 906: 167287, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37748599

RESUMO

Whether nanoplastics with differential charges cause intestinal impairment via distinct mechanisms remains unclear. We investigated the relationship between fecal metabolites and the gut microbiome, and potential biomarkers thereof, in mice following exposure to differentially charged polystyrene nanoplastics (PS-NPs). Metagenomic analysis revealed that exposure to differentially charged PS-NPs resulted in alterations in the abundances of Bilophila_wadsworthia, Helicobacter apodemus, and Helicobacter typhlonius. A total of 237 fecal metabolites were significantly altered in mice that exhibited intestinal impairment, and these included 10 gut microbiota-related fecal metabolites that accurately discriminated impaired intestinal samples from the control. Additionally, the specific gut microbiome-related fecal metabolite-based model approach for the prediction of intestinal impairment in mice had an area under the curve (AUC) of 1.0 in the PS (without charge) group, an AUC of 0.94 in the PS-NH2 (positive charge) group, and an AUC of 0.86 in the PS-COOH (negative charge) group. Thus, the model showed promising evaluable accuracy for the prediction of intestinal impairment induced by nanoplastics in a charge-specific manner. Our study demonstrates that the fecal metabolome of mice with intestinal impairment following exposure to differentially charged nanoplastics is associated with changes in the gut microbiome. The identified biomarkers have potential application for the detection of intestinal impairment after exposure to negative, positive, or noncharged nanomaterials.


Assuntos
Microbioma Gastrointestinal , Animais , Camundongos , Metagenoma , Microplásticos , Metaboloma , Poliestirenos , Biomarcadores , RNA Ribossômico 16S
9.
J Biomed Mater Res A ; 112(1): 110-120, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37772330

RESUMO

Natural killer (NK) cell functionality is a strong indicator of favorable prognosis in cancer patients, making NK cells an appealing therapeutic target to prevent lymph node dissemination. We engineered liposomes that are conjugated with anti-CD335 antibodies for NK cell targeting, and the apoptotic ligand TRAIL to kill cancer cells. Liposomes were made using a thin film hydration method followed by extrusion to approximately 100 nm in diameter and conjugation of proteins via thiol-maleimide click chemistry. TRAIL/anti-CD335 liposomes successfully bound to isolated NK cells. Once piggybacked to the surface of NK cells, these "Super Natural Killer Cells" were able to more effectively kill oxaliplatin-resistant SW620 cells and metastatic COLO205 colorectal cancer cells via TRAIL-mediated apoptosis compared to NK cells alone. Importantly, Super NK cells were more effective under physiological levels of fluid shear stress found in the lymphatics. Liposome biodistribution after intravenous administration confirmed the sustained presence of liposomes within the spleen and tumor draining mesenteric lymph nodes for at least 4 days. These results demonstrate the enhanced apoptotic effects of NK cells armored with liposomal TRAIL against clinically relevant colorectal cancer cells, providing the groundwork for in vivo treatment studies in mouse models of colorectal cancer metastasis.


Assuntos
Neoplasias do Colo , Lipossomos , Camundongos , Animais , Humanos , Citotoxicidade Imunológica , Distribuição Tecidual , Células Matadoras Naturais/metabolismo , Apoptose , Neoplasias do Colo/patologia
10.
Sci Total Environ ; 906: 167592, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37802340

RESUMO

Microplastics and nanoplastics are ubiquitous pollutants, widely spread in the living and natural environment. Although their potential impact on human health has been investigated, many doubts remain about their effects in carcinogenic processes. We investigated the potential effects and its molecular mechanisms of polystyrene nanoplastics (PS-NPs) on epithelial ovarian cancer (EOC) using the human EOC cell line HEY as an in vitro cell model and mice as a mammalian model. In vivo exposure to PS-NPs (100 nm; 10 mg/L) via drinking water significantly accelerated EOC tumor growth in mice. In in vitro tests the PS-NPs reduced the relative viability of EOC cells in a dose-dependent manner. Histological analysis showed increased mitotic counts in EOC tumor tissues of PS-NP exposed mice. PS-NP exposure significantly affected gene expression and disturbed many metabolic pathways in both cultured EOC cells and EOC tumor tissue in mice. Gene functional and pathway analysis indicated that immune-related responses and the tumor microenvironment pathway were significantly enriched, which may be attributed to disturbed expression of thrombomodulin (THBD) and its regulators. It may be concluded that PS-NP exposure caused a significant acceleration of EOC tumor growth in mice and a dose-dependent decrease in the relative viability of EOC cells by altering the tumor growth microenvironment. This offers new insights into the mechanisms underlying PS-NP effects on EOC.


Assuntos
Nanopartículas , Neoplasias Ovarianas , Poluentes Químicos da Água , Animais , Camundongos , Humanos , Feminino , Poliestirenos/toxicidade , Microambiente Tumoral , Microplásticos , Plásticos , Neoplasias Ovarianas/induzido quimicamente , Carcinoma Epitelial do Ovário/induzido quimicamente , Carcinoma Epitelial do Ovário/genética , Carcinoma Epitelial do Ovário/patologia , Nanopartículas/toxicidade , Mamíferos
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166906, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37802156

RESUMO

Sphingolipids are important for the physicochemical properties of cellular membranes and deregulated in tumors. In human colon cancer tissue ceramide synthase (CerS) 4 and CerS5 are reduced which correlates with a reduced survival probability of late-stage colon cancer patients. Both enzymes are reduced after hypoxia in advanced colorectal cancer (CRC) cells (HCT-116, SW620) but not in non-metastatic CRC cells (SW480, Caco-2). Downregulation of CerS4 or CerS5 in advanced CRC cells enhanced tumor formation in nude mice and organoid growth in vitro. This was accompanied by an enhanced proliferation rate and metabolic changes leading to a shift towards the Warburg effect. In contrast, CerS4 or CerS5 depletion in Caco-2 cells reduced tumor growth in vivo. Lipidomic and proteomic analysis of membrane fractions revealed significant changes in tumor-promoting cellular pathways and cellular transporters. This study identifies CerS4 and CerS5 as prognostic markers for advanced colon cancer patients and provides a comprehensive overview about the associated cellular metabolic changes. We propose that the expression level of CerS4 and CerS5 in colon tumors could serve as a basis for decision-making for personalized treatment of advanced colon cancer patients. Trial registration: The study was accredited by the study board of the Deutsche Krebsgesellschaft (Registration No: St-D203, 2017/06/30, retrospectively registered).


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Animais , Camundongos , Humanos , Esfingolipídeos/metabolismo , Células CACO-2 , Camundongos Nus , Prognóstico , Proteômica , Neoplasias do Colo/metabolismo , Neoplasias Colorretais/patologia , Hipóxia
12.
Phytomedicine ; 122: 155141, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37837898

RESUMO

BACKGROUND: Pneumonia is one of the major causes of death after pathogens infection. Zingiberis rhizoma (GAN JIANG) is a herb that used in combination with other Chinese medicines to treat pathogen such as virus induced pneumonia. However, the affect of hexahydrocurcumin (HHC), a component from Zingiberis rhizoma, on pneumonia remains unknown. PURPOSE: This study aims to explore the effects of HHC on lipopolysaccharide (LPS)-induced acute pneumonia, and to clarify the underlying mechanism. METHODS: The pneumonia model of C57BL/6 mice was established by intratracheal injection of LPS to evaluate the therapeutic effect of HHC on lung injury and inflammation in vivo. RAW264.7 macrophages were utilized to illustrate the cellular mechanism of HHC in vitro. RESULTS: HHC alleviated lung injury, ROS and inflammatory cytokine IL-6 production in pneumonia mice in vivo. Molecular docking results disclosed the binding of HHC to JAK1 protein. The study further showed that HHC suppressed the inflammatory cytokines such as IL-6, TNF-α, IL-1ß gene expression, inhibited the phosphorylation of JAK1 but not JAK3, and the subsequent STAT3 phosphorylation in LPS-activated macrophages. HHC exhibited no effects on the protein levels of JAK1 and STAT3 in vitro. Consistently, HHC also attenuated the JAK1, STAT3 phosphorylation in pneumonia mice in vivo. CONCLUSION: The results reveal that HHC attenuates pneumonia by targeted inhibition of JAK1/STAT3 signaling pathway. It indicates the novel role of HHC to treat pneumonia, and its potential applications for JAK inhibitor-treated diseases.


Assuntos
Lesão Pulmonar , Pneumonia , Camundongos , Animais , Lipopolissacarídeos/farmacologia , Interleucina-6/metabolismo , Simulação de Acoplamento Molecular , Camundongos Endogâmicos C57BL , Transdução de Sinais , Citocinas/metabolismo , Fator de Transcrição STAT3/metabolismo
13.
Behav Brain Res ; 457: 114715, 2024 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-37838243

RESUMO

Calorie restriction (CR) has been shown to extend the mean and maximum lifespan in both preclinical and clinical settings. We have previously demonstrated that CR attenuates lipopolysaccharide (LPS)-induced fever and sickness behavior. CR also leads to reductions in pro-inflammatory and increases in anti-inflammatory profiles. LPS is a bacterial mimetic; however, few studies have explored this phenomenon utilizing a viral mimetic, such as polyinosinic:polycytidylic acid (poly I:C). Dose-dependently, poly I:C induced an increase in core body temperature (Tb), with the largest dose (5000 µg/kg) resulting in a 1.62 °C ( ± 0.23 °C) Tb increase at 7 h post-injection in ad libitum mice and was associated with reduced home-cage locomotor activity. We then investigated the effect of 50% CR for 28 days to attenuate fever and sickness behavior induced by a poly I:C (5000 µg/kg) viral immune challenge. CR resulted in the partial attenuation of fever and sickness behavior measures post-poly I:C. The freely fed, control mice demonstrated a 2.02 °C ( ± 0.22 °C) increase in Tb at 7 h post-injection compared to the CR poly I:C group which demonstrated an increase in Tb of 0.94 °C ( ± 0.27 °C). Locomotor patterns post-injection were different, CR mice displayed a reduction in activity during the light phase, and the control group displayed a reduction during the dark phase. CR moderately attenuated the neuroinflammatory response with a reduction in microglial density in the ventromedial nucleus of the hypothalamus. The fever and sickness behavior attenuation seen after CR may be driven by similar anti-inflammatory processes as after LPS; however, further investigation is required.


Assuntos
Restrição Calórica , Comportamento de Doença , Camundongos , Animais , Lipopolissacarídeos/efeitos adversos , Febre/induzido quimicamente , Poli I-C/efeitos adversos , Anti-Inflamatórios/efeitos adversos
14.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166918, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37838353

RESUMO

Thiol molecules have been recently re-considered as drug candidates in viral infections because of their ability to induce redox changes which interfere with virus life cycle and modulate the host immune response. Little is known about the molecular mechanisms of their immunomodulatory properties. Here we show that I-152, a thiol molecule metabolized to release N-acetyl-l-cysteine and cysteamine and acting as a pro-glutathione agent, causes early up-regulation of immunoproteasome subunits in the lymph nodes of murine leukemia virus infected mice. This evidence suggests that the immunoproteasome may be modulated by thiol-based compounds with important implications in understanding redox-controlled immunoregulation.


Assuntos
Leucemia , Síndrome de Imunodeficiência Adquirida Murina , Camundongos , Animais , Acetilcisteína/farmacologia , Acetilcisteína/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Cisteamina/farmacologia , Compostos de Sulfidrila , Regulação para Cima , Retroviridae/metabolismo , Linfonodos/patologia
15.
Phytomedicine ; 122: 155128, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37839227

RESUMO

BACKGROUND: Pancreatic cancer is an extremely malignant digestive tumor, however, owing to its high drug resistance of pancreatic cancer, the search for more effective anti-pancreatic cancer drugs is urgently needed. Lycorine, an alkaloid of natural plant origin, exerts antitumor effects on a variety of tumors. PURPOSE: This study aimed to investigate the therapeutic effect of lycorine on pancreatic cancer and elucidate its potential molecular mechanism. METHODS: Two pancreatic cancer cell lines, PANC-1 and BxPC-3, were used to investigate the therapeutic effects of lycorine on pancreatic cancer in vitro using the CCK8 assay, colony formation assay, 5-Ethynyl-2'- deoxyuridine (EdU) incorporation assay, flow cytometry, and western blotting. Transcriptome sequencing and gene set enrichment analysis (GSEA) were used to analyze the differentially expressed genes and pathways after lycorine treatment. Molecular docking, quantitative real-time PCR (qRT-PCR), oil red O staining, small interfering RNA (siRNA) transfection, and other experiments were performed to further validate the differentially expressed genes and pathways. In vivo experiments were conducted to investigate lycorine's inhibitory effects and toxicity on pancreatic cancer using a tumor-bearing mouse model. RESULTS: Lycorine inhibited the proliferation of pancreatic cancer cells, caused G2/M phase cycle arrest and induced apoptosis. Transcriptome sequencing and GSEA showed that lycorine inhibition of pancreatic cancer was associated with fatty acid metabolism, and aldehyde dehydrogenase 3A1 (ALDH3A1) was a significantly enriched target in the fatty acid metabolism process. ALDH3A1 expression was significantly upregulated in pancreatic cancer and was closely associated with prognosis. Molecular docking showed that lycorine binds strongly to ALDH3A1. Further studies revealed that lycorine inhibited the fatty acid oxidation (FAO) process in pancreatic cancer cells and induced cell growth inhibition and apoptosis through ALDH3A1. Lycorine also showed significant suppressive effects in tumor-bearing mice. Importantly, it did not result in significant toxicity to liver and kidney of mice, demonstrating its therapeutic potential as a safe antitumor agent. CONCLUSION: Lycorine inhibited pancreatic cancer cell proliferation, blocked the cell cycle, and induced apoptosis by targeting ALDH3A1. FAO inhibition was identified for the first time as a possible mechanism for the anticancer effects of lycorine. These findings enrich the theory of targeted therapy for pancreatic cancer, expand our understanding of the pharmacological targets of lycorine, and provide a reference for exploring its natural components.


Assuntos
Antineoplásicos , Neoplasias Pancreáticas , Animais , Camundongos , Simulação de Acoplamento Molecular , Linhagem Celular Tumoral , Transcriptoma , Proliferação de Células , Antineoplásicos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Apoptose , RNA Interferente Pequeno/farmacologia , Ácidos Graxos
16.
Phytomedicine ; 122: 155095, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37844381

RESUMO

BACKGROUND: Mycotoxins, such as aflatoxin and ochratoxin A (OTA), are found at measurable levels in many staple foods; the health implications of long-term exposure of such toxins are poorly understood. Increasing evidence has confirmed the important role of OTA in upregulation of oxidative stress- and inflammatory response-induced tissue injury. However, it remains unknown whether ochratoxin A can promote chronic colitis and its associated colon cancer (CRC) development, and potential molecular mechanism. Additionally, RING finger-interacting protein with C kinase (RINCK) is a ubiquitin ligase and mediates immune response. Unfortunately, the potential molecular function of RINCK on regulation of colitis is still largely unknown. PURPOSE: This study aims to provide mechanistic evidence that the role of RINCK in colitis and early colorectal cancer progression in response to OTA treatment via targeting nuclear factor erythroid 2-related factor 2 (NRF2). METHODS: The Cancer Genome Atlas (TCGA) database, GEO database, human subjects with CC phenotype and CC cell lines were used in this work. Pathological links between OTA, RINCK and treatment of CC are revealed through comprehensive means such as biological information analysis, clinical experiments, RNA-seq, and verification experiments. RESULTS: In this study, under oxidative stress in setting of colitis, we first identified RINCK as a key regulatory factor and a novel endogenous suppressor of nuclear factor erythroid 2-related factor 2 (NRF2), and we also confirm that RINCK is a NRF2 partner protein that catalyses its ubiquitination and degradation in intestinal epithelial cells (IECs). Notably, in vivo study, pathological phenotypes triggered by OTA pretreatment, accompanied by post-treatment of dextran sulfate sodium (DSS)-induced colitis was significantly mitigated by IEC-specific deficiency of Rinck, IEC-Rinck(KO) and adenovirus-associated virus (AAV)-triggered suppression of Rinck in rodent model, and lentivirus (LV)-mediated downregulation of Rinck (LV-shRinck) in rabbit model, as determined by decreased endogenous reactive oxygen species (ROS) production, pro-inflammatory cytokines contents, improved body weights, reduced survival rates, restored colon length, assuasive DAI and histological scores. Inversely, transgenic mice by IEC-specific Rinck overexpression, IEC-Rinck(OE) accelerated colitis in acute or chronic colitis rodent models and in vitro experiments. Moreover, we found that OTA pretreatment-promoted azoxymethane (AOM)/DSS-induced colitis-associated early colorectal cancer (CRC) was also dramatically reduced by IEC-Rinck(KO), indicated by the decreased tumor number and corresponding KI-67 levels. Clinical samples analysis revealed that RINCK levels were greatly increased in tumor tissues of patients with CRC phenotypes. In parallel, RINCK deletion remarkably retarded the proliferation of colon cancer and tumor growth in vitro and in vivo, respectively. Mechanistically, in response to onset of colitis, RINCK directly interacts with NRF2 and promotes ubiquitin-proteasome degradation via increasing K48-linkage ubiquitin chain, thus leads in suppression of NRF2 nuclear translocation and its downstream cascade inactivation, which retards antioxidant defense. CONCLUSION: The findings suggested that oral sub-chronic exposure of OTA significantly facilitates DSS-induced colitis and colitis-associated CRC development. These results further elucidated the potential role of RINCK in colitis progression by mediating NRF2 degradation, and could be considered as a therapeutic target for the treatment of such disease.


Assuntos
Colite , Neoplasias do Colo , Neoplasias Colorretais , Enterite , Doenças Inflamatórias Intestinais , Camundongos , Animais , Humanos , Coelhos , Fator 2 Relacionado a NF-E2 , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Camundongos Transgênicos , Doença Crônica , Enterite/complicações , Ubiquitinas/uso terapêutico , Sulfato de Dextrana/efeitos adversos , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
17.
Phytomedicine ; 122: 155132, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37844379

RESUMO

BACKGROUND: White adipose tissues (WAT) release large amounts of inflammatory mediators, which are responsible for the pathology of rheumatoid arthritis (RA). PURPOSE: The current study investigated the involvement of WAT in the treatments of antigen-induced arthritis (AIA) mice with the herbal formula Qing-Luo-Yin (QLY). METHODS: Cytokines and biochemical/metabolic indicators were determined by ELISA and colorimetry methods, respectively. Monocytes were analyzed by flow cytometry. Tissues were subjected to PCR, western-blot and histological analyses. Pre-adipocytes were cultured in the different mouse serum from the in vivo experiment, and some of them were treated by certain compounds or/and lipopolysaccharide. Afterwards, the catalytic activity and thermostability of SIRT1 were tested. Gene/protein expression and cytokine production were investigated too. NAMPT and SIRT1 were silenced in some cells by siRNA. RESULTS: AIA mice suffered from inflammatory adipokines-mediated metabolism and immune disorders. Besides joint protective effects, QLY therapies favored adipocyte differentiation and suppressed inflammatory adipokines release. The up-regulation of fatty acid oxidation and inflammatory monocyte polarization was therefore inhibited in peripheral tissues. PPARγ expression was generally promoted by QLY. Whereas, SIRT1 activity was always impaired, indicated by the declined NAD+ levels and the increased ace-p65 expression. QLY effectively inhibited eNAMPT release in AIA mouse serum-cultured pre-adipocytes. This effect was antagonized by resveratrol (a SIRT1 agonist) and overshadowed by NAMPT silencing. QLY-related compounds berberine, dioscin and sophocarpine showed high binding affinities to SIRT1, stabilized this protein, and inhibited its deacetylation activity in vitro. Their effects on ace-p65 expression were weakened when SIRT1 was silenced. CONCLUSION: SIRT1 inhibitors in QLY reduced eNAMPT production and up-regulated PPARγ in AIA mice, leading to inflammation remission. These clues show that except for the well-known anti-inflammatory functions, SIRT1 participates in inflammatory reactions too and could be a potential anti-rheumatic target.


Assuntos
Artrite Reumatoide , Sirtuína 1 , Camundongos , Animais , Sirtuína 1/metabolismo , PPAR gama , Citocinas/metabolismo , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Adipocinas , Inflamação/tratamento farmacológico
18.
Phytomedicine ; 122: 155135, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37856990

RESUMO

BACKGROUND: Ferroptosis is an emerging iron-dependent programmed cell death mode characterized by lipid peroxidation and iron accumulation, closely associated with Hepatocellular Carcinoma (HCC) progression. Although the impact of Polyphyllin I (PPI), a prominent bioactive constituent derived from Paris polyphylla, on diverse malignancies has been established, the specific role and potential mechanistic pathways through which PPI modulates ferroptosis in HCC remain elusive. PURPOSE: This study aimed to elucidate the anti-cancer properties and potential mechanisms of PPI in inducing ferroptosis and triggering mitochondrial injury in HCC. METHODS: Cell viability was assessed using CCK-8 assays. EdU proliferation and colony formation assays were employed to evaluate cell proliferation. A wound-healing assay was performed to assess cell migration. Transwell assay was utilized to evaluate cell invasion. Ferroptosis was evaluated through the utilization of a FerroOrange fluorescent probe, malondialdehyde (MDA) and reduced glutathione (GSH) assay kits, DCFH-DA fluorescent probe, western blotting, and transmission electron microscopy (TEM) analysis. Molecular docking, immunofluorescence, and western blotting were employed to predict and validate the binding and interaction of PPI with Nrf2, HO-1, xCT, and GPX4. Mitochondrial structure and membrane potential changes were evaluated using JC-1 and Mito Tracker Green fluorescent probes. A nude mice xenograft model was constructed to determine the inhibitory effects and the levels of ferroptosis of PPI on HCC through hematoxylin and eosin (H&E), Prussian blue reaction, immunofluorescence staining, immunohistochemistry, and western blotting analysis, in vivo. RESULTS: PPI exhibited dose-dependent inhibitory effects on the proliferation, invasion, and metastasis of HCC cells mediated by increasing reactive oxygen species (ROS) and MDA levels, promoting Fe2+ accumulation, depleting GSH, and suppressing the expression of xCT and GPX4, thereby inducing ferroptosis in HCC. The induction of ferroptosis by PPI was associated with the binding of PPI to Nrf2, HO-1, and GPX4 proteins, modulating the Nrf2/HO-1/GPX4 antioxidant axis. PPI also induced mitochondrial structural damage and decreased mitochondrial membrane potential (MMP). Inhibition of ferroptosis by ferrostatin-1 (Fer-1) mitigated the mitochondrial disruption induced by PPI. In vivo, PPI inhibited Nrf2/HO-1/GPX4 axis-induced ferroptosis, impeding HCC growth similar to the effects of sorafenib. CONCLUSION: These results demonstrated that PPI intervention can suppress the proliferation, invasion, and metastasis of HCC cells by enhancing mitochondrial disruption and inducing ferroptosis via the Nrf2/HO-1/GPX4 axis. Consequently, our research advances the frontiers of pharmacodynamics and deepens our comprehension of the intricate mechanisms underpinning PPI. Furthermore, it has yielded an innovative treatment stratagem rooted in the tenets of Traditional Chinese Medicine (TCM), thereby furnishing a novel therapeutic avenue for addressing HCC.


Assuntos
Carcinoma Hepatocelular , Ferroptose , Neoplasias Hepáticas , Animais , Camundongos , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Fator 2 Relacionado a NF-E2 , Corantes Fluorescentes , Camundongos Nus , Simulação de Acoplamento Molecular , Neoplasias Hepáticas/tratamento farmacológico , Ferro , Espécies Reativas de Oxigênio
19.
Phytomedicine ; 122: 155121, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37856988

RESUMO

BACKGROUND: Raddeanin A is a triterpenoid isolated from Anemone raddeana Regel. It exhibits a broad spectrum of biological activities such as anti-tumor and anti-inflammatory, however, its neuroprotective effect in targeting Alzheimer's disease (AD) remains uninvestigated. PURPOSE: To provide scientific base for the development of novel AD drug by clarifying the neuroprotective effect and molecular mechanisms of raddeanin A in both in vitro and in vivo AD model. STUDY DESIGN: To confirm the neuroprotective role of raddeanin A in the treatment of AD, its mechanisms and effects on ß-amyloidosis and Aß fibrillation was studied in U87 cells. Besides, the improvement on cognitive deficit, pathological defects, reactive astrocyte clusters, inhibition on neuronal inflammation and apoptosis were further studied in 3 x Tg-AD mice model of AD. METHODS: Real-time PCR, western blot, dot blot, biolayer interferometry and bioinformatics analysis were used to confirm the in vitro effect and targets of raddeanin A on ß-amyloidosis and its associated protein network. A series of experiments including Morris water maze, H&E staining, nissl staining and immunofluorescence analysis were conducted to confirm the protective behavioral effect of raddeanin A in the in vivo AD mice model. RESULTS: Raddeanin A was identified to reduce ß-amyloidosis in U87 cells and 3 x Tg-AD mice model of AD by decreasing level of BACE1, APP, APP-ß and Aß. Raddeanin A improved behavioral, spatial memory and learning ability in the AD mice. In the cortex and hippocampus, raddeanin A improved the morphology and arrangement of neurons, lower the level of reactive astrocyte marker GFAP and apoptotic marker proteins Bax/Bcl2 ratio. Moreover, raddeanin A upregulated the mRNA and protein level of Prkcα in the hippocampus of AD mice whose neuroprotective effect was exerted possibly via the activation of protein kinase C. CONCLUSION: As a novel natural agent targeting ß-amyloidosis, our results provide the first evidence of the multiple in vitro and in vivo neuroprotective effect of raddeanin A, suggesting its potential therapeutic application in preventing or alleviating the symptoms of AD.


Assuntos
Doença de Alzheimer , Amiloidose , Anemone , Fármacos Neuroprotetores , Camundongos , Animais , Doença de Alzheimer/metabolismo , Anemone/metabolismo , Secretases da Proteína Precursora do Amiloide , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ácido Aspártico Endopeptidases , Cognição , Camundongos Transgênicos , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças , Precursor de Proteína beta-Amiloide/genética
20.
Phytomedicine ; 122: 155122, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37863002

RESUMO

BACKGROUND: Colchicine (COL) is a well-known plant-derived mitogenic toxin that has been widely applied for the treatment of immune system diseases and various cancers. However, its clinical use is severely limited by frequent occurrence of poisoning accidents, and the mechanism of COL poisoning is not clear yet. PURPOSE: The present study aimed to unveil how COL works as a toxin based on untargeted metabolomics analysis of animal models and clinical human case. METHODS: KM mice orally administered COL were used to establish poisoning models, and plasma samples were collected for untargeted metabolomics analysis. The data mining was performed to screen dose-dependent differences and disturbed metabolic pathways. The blood samples collected from clinical COL poisoning human case at various time points during treatment period were further analyzed to investigate the temporal changes in the metabolic disposition of COL in vivo and also verify the findings from mice. Finally, the expression of key pathways was evaluated by ELISA and Western blotting analysis. RESULTS: Histological examination demonstrated systemic toxicity of COL poisoning in mice. Metabolite profiling analysis of plasma samples from model mice and clinical case both revealed that COL poisoning could significantly disturb in vivo metabolism of amino acid and lipid metabolism by the FXR/AMPK signal pathway. Quantitative monitoring of the metabolic process of COL further demonstrated that it could be greatly ameliorated with the rapid metabolic transformation of COL in vivo, which thus may be an effective detoxification pathway for COL poisoning. CONCLUSION: The findings of the present study provided new insight into the molecular mechanism of COL poisoning, thus helpful for guiding reasonable application of this phytotoxin.


Assuntos
Colchicina , Metabolômica , Camundongos , Humanos , Animais , Colchicina/toxicidade , Redes e Vias Metabólicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...