Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 227.165
Filter
1.
Sci Rep ; 14(1): 12838, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834634

ABSTRACT

Disorders related to gut health are a significant cause of morbidity among athletes in wheelchair. This pilot feasibility trial aims to investigate whether probiotics compared to prebiotics can improve inflammatory status and gut microbiome composition in elite athletes in wheelchair. We conducted a 12-week, randomized, cross-over controlled trial involving 14 elite Swiss athletes in wheelchair. Participants were given a multispecies-multistrain probiotic or prebiotic (oat bran) daily for 4 weeks (Clinical trials.gov NCT04659408 09/12/2020). This was followed by a 4-week washout and then crossed over. Thirty inflammatory markers were assessed using bead-based multiplex immunoassays (LegendPlex) from serum samples. The gut microbiome was characterized via 16S rRNA sequencing of stool DNA samples. Statistical analyses were conducted using linear mixed-effect models (LMM). At baseline, most athletes (10/14) exhibited low levels of inflammation which associated with higher gut microbiome alpha diversity indices compared to those with high inflammation levels. The use of probiotic had higher decrease in 25 (83%) inflammatory markers measured compared to prebiotic use. Probiotic has the potential in lowering inflammation status and improving the gut microbiome diversity. The future trial should focus on having sufficient sample sizes, population with higher inflammation status, longer intervention exposure and use of differential abundance analysis.


Subject(s)
Athletes , Cross-Over Studies , Gastrointestinal Microbiome , Inflammation , Prebiotics , Probiotics , Humans , Probiotics/administration & dosage , Probiotics/therapeutic use , Prebiotics/administration & dosage , Male , Pilot Projects , Adult , Female , Wheelchairs , Young Adult , RNA, Ribosomal, 16S/genetics , Biomarkers , Feces/microbiology
2.
Sci Rep ; 14(1): 12829, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834647

ABSTRACT

The present cross-sectional study aimed to explore the relationship between systemic inflammatory indices (SIIs) and anthropometric measures, metabolic, and liver function biomarkers in patients with non-alcoholic fatty liver disease (NAFLD). This study was carried out on 238 NAFLD patients with overweight or obesity, aged 18-55 years. Anthropometric measurements were done and body mass index (BMI), waist-to-hip ratio (WHR), and waist-to-height ratio (WHtR) were estimated. Metabolic factors including serum glucose, lipid profile, liver function biomarkers, and complete blood cell count were assessed after a 24-h fasting state. SIIs including the ratios of neutrophil to lymphocyte (NLR), monocytes to lymphocyte (MLR), platelet to lymphocyte (PLR), and monocytes to high-density lipoprotein cholesterol (MHR) were calculated. Results indicate that apart from PLR, all of the SIIs significantly changed by increasing steatosis severity (all p < 0.05). Moreover, changes in NLR showed a significant association with anthropometric indices including waist circumference (p = 0.032), BMI (p = 0.047), and WHtR (p = 0.002), as well as levels of fasting blood sugar (p = 0.045), triglycerides, (p = 0.025) and low-density lipoprotein cholesterol (p = 0.006). The findings also indicate the relations between lipid profile and all studied SIIs, notably MHR and MLR. All of the SIIs exhibited associations with some liver function indices as well. MHR was positively correlated with the metabolic risk factors of NAFLD while, oppositely, PLR was considered as a preventive marker of NAFLD.


Subject(s)
Body Mass Index , Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/metabolism , Adult , Male , Female , Middle Aged , Cross-Sectional Studies , Adolescent , Young Adult , Inflammation/blood , Inflammation/metabolism , Biomarkers/blood , Liver/metabolism , Liver/pathology , Anthropometry , Obesity/complications , Obesity/metabolism , Obesity/blood , Liver Function Tests , Blood Glucose/metabolism , Waist-Hip Ratio
3.
Sci Rep ; 14(1): 12827, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834834

ABSTRACT

Gut microbiota plays a crucial role in gastrointestinal tumors. Additionally, gut microbes influence the progression of esophageal cancer. However, the major bacterial genera that affect the invasion and metastasis of esophageal cancer remain unknown, and the underlying mechanisms remain unclear. Here, we investigated the gut flora and metabolites of patients with esophageal squamous cell carcinoma and found abundant Bacteroides and increased secretion and entry of the surface antigen lipopolysaccharide (LPS) into the blood, causing inflammatory changes in the body. We confirmed these results in a mouse model of 4NQO-induced esophageal carcinoma in situ and further identified epithelial-mesenchymal transition (EMT) occurrence and TLR4/Myd88/NF-κB pathway activation in mouse esophageal tumors. Additionally, in vitro experiments revealed that LPS from Bacteroides fragile promoted esophageal cancer cell proliferation, migration, and invasion, and induced EMT by activating the TLR4/Myd88/NF-κB pathway. These results reveal that Bacteroides are closely associated with esophageal cancer progression through a higher inflammatory response level and signaling pathway activation that are both common to inflammation and tumors induced by LPS, providing a new biological target for esophageal cancer prevention or treatment.


Subject(s)
Epithelial-Mesenchymal Transition , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lipopolysaccharides , Myeloid Differentiation Factor 88 , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Myeloid Differentiation Factor 88/metabolism , Animals , NF-kappa B/metabolism , Humans , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/microbiology , Mice , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/microbiology , Epithelial-Mesenchymal Transition/drug effects , Cell Line, Tumor , Neoplasm Invasiveness , Inflammation/metabolism , Inflammation/pathology , Bacteroidetes , Gastrointestinal Microbiome , Cell Movement/drug effects , Male , Neoplasm Metastasis , Cell Proliferation , Female
4.
Zhonghua Nei Ke Za Zhi ; 63(6): 605-612, 2024 Jun 01.
Article in Chinese | MEDLINE | ID: mdl-38825930

ABSTRACT

Objective: To observe the characteristics and differences of gut microbiota in asthma patients with different inflammatory types through metagenomic analysis. Methods: Adults aged ≥18 years who visited the Respiratory Clinic of Peking University Third Hospital from August 1, 2021 to August 31, 2022 and were primarily diagnosed with asthma were selected as the study subjects. Finally, 29 patients with stable asthma were included. Fresh fecal samples were collected and the fecal DNA was extracted for high-throughput 16sRNA sequencing of gut microbiota. The diversity and community structure of gut microbiota in different groups of asthma patients were compared, and the species differences were analyzed through random forest and LEfSe analysis. Results: There were sex-based differences in asthma patients with different types of inflammation, and the proportion of female patients was higher in neutrophilic asthma patients (χ2=4.14, P=0.042). There was no significant intergroup difference in the alpha diversity of gut microbiota among asthma patients with different inflammatory types, but there were significant differences in the microbiome. Patients with neutrophilic asthma had higher relative abundance of Bacillales (P=0.029) and Oscillospiraceae (P=0.015). In species LEfSe analysis, patients with eosinophilic asthma had a higher relative abundance of fungi. Conclusion: There are intergroup differences in the gut microbiota of asthma patients with different inflammation types, and fungi are biomarkers that distinguish the differences in gut microbiota between patients with eosinophilic asthma and neutrophilic asthma.


Subject(s)
Asthma , Feces , Gastrointestinal Microbiome , Inflammation , Humans , Asthma/microbiology , Feces/microbiology , Inflammation/microbiology , Female , Male , RNA, Ribosomal, 16S/genetics , Adult
5.
Nanotechnology ; 35(33)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829163

ABSTRACT

Dry eye disease (DED) is a major global eye disease leading to severe eye discomfort and even vision impairment. The incidence of DED has been gradually increasing with the high frequency of use of electronic devices. It has been demonstrated that celastrol (Cel) has excellent therapeutic efficacy in ocular disorders. However, the poor water solubility and short half-life of Cel limit its further therapeutic applications. In this work, a reactive oxygen species (ROS) sensitive polymeric micelle was fabricated for Cel delivery. The micelles improve the solubility of Cel, and the resulting Cel loaded micelles exhibit an enhanced intervention effect for DED. Thein vitroresults demonstrated that Cel-nanomedicine had a marked ROS responsive release behavior. The results ofin vitroandin vivoexperiments demonstrated that Cel has excellent biological activities to alleviate inflammation in DED by inhibiting TLR4 signaling activation and reducing pro-inflammatory cytokine expression. Therefore, the Cel nanomedicine can effectively eliminate ocular inflammation, promote corneal epithelial repair, and restore the number of goblet cells and tear secretion, providing a new option for the treatment of DED.


Subject(s)
Dry Eye Syndromes , Micelles , Nanomedicine , Pentacyclic Triterpenes , Reactive Oxygen Species , Triterpenes , Dry Eye Syndromes/drug therapy , Pentacyclic Triterpenes/pharmacology , Animals , Reactive Oxygen Species/metabolism , Mice , Nanomedicine/methods , Triterpenes/pharmacology , Triterpenes/chemistry , Inflammation/drug therapy , Toll-Like Receptor 4/metabolism , Humans , Tears/metabolism , Tears/drug effects
6.
Food Res Int ; 188: 114489, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823872

ABSTRACT

Solanum nigrum L. (SN) berry is an edible berry containing abundant polyphenols and bioactive compounds, which possess antioxidant and antiinflammatory properties. However, the effects of SN on alcohol-induced biochemical changes in the enterohepatic axis remain unclear. In the current study, a chronic ethanol-fed mice ALD model was used to test the protective mechanisms of SN berries. Microbiota composition was determined via 16S rRNA sequencing, we found that SN berries extract (SNE) improved intestinal imbalance by reducing the Firmicutes to Bacteroides ratio, restoring the abundance of Akkermansia microbiota, and reducing the abundance of Allobaculum and Shigella. SNE restored the intestinal short-chain fatty acids content. In addition, liver transcriptome data analysis revealed that SNE primarily affected the genes involved in lipid metabolism and inflammatory responses. Furthermore, SNE ameliorated hepatic steatosis in alcohol-fed mice by activating AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase (ACC), peroxisome proliferator-activated receptor α (PPAR-α). SNE reduced the expression of toll-like receptor 4 (TLR4), myeloid differentiation factor-88 (MyD88) nuclear factor kappa-B (NF-κB), which can indicate that SNE mainly adjusted LPS/TLR4/MyD88/NF-κB pathway to reduce liver inflammation. SNE enhanced hepatic antioxidant capacity by regulating NRF2-related protein expression. SNE alleviates alcoholic liver injury by regulating of gut microbiota, lipid metabolism, inflammation, and oxidative stress. This study may provide a reference for the development and utilization of SN resources.


Subject(s)
Fruit , Gastrointestinal Microbiome , Lipid Metabolism , Liver Diseases, Alcoholic , Oxidative Stress , Plant Extracts , Solanum nigrum , Animals , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Lipid Metabolism/drug effects , Plant Extracts/pharmacology , Mice , Fruit/chemistry , Solanum nigrum/chemistry , Male , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/prevention & control , Mice, Inbred C57BL , Inflammation , Liver/drug effects , Liver/metabolism , Toll-Like Receptor 4/metabolism , Disease Models, Animal , PPAR alpha/metabolism , Antioxidants/pharmacology , Ethanol
7.
J Toxicol Sci ; 49(6): 281-288, 2024.
Article in English | MEDLINE | ID: mdl-38825487

ABSTRACT

Nitric oxide (NO) plays a physiological role in signal transduction and excess or chronic NO has toxic effects as an inflammatory mediator. NO reversibly forms protein S-nitrosylation and exerts toxicological functions related to disease progression. DNA methyltransferases, epigenome-related enzymes, are inhibited in enzymatic activity by S-nitrosylation. Therefore, excess or chronic NO exposure may cause disease by altering gene expression. However, the effects of chronic NO exposure on transcriptome are poorly understood. Here, we performed transcriptome analysis of A549, AGS, HEK293T, and SW48 cells exposed to NO (100 µM) for 48 hr. We showed that the differentially expressed genes were cell-specific. Gene ontology analysis showed that the functional signature of differentially expressed genes related to cell adhesion or migration was upregulated in several cell lines. Gene set enrichment analysis indicated that NO stimulated inflammation-related gene expression in various cell lines. This finding supports previous studies showing that NO is closely involved in inflammatory diseases. Overall, this study elucidates the pathogenesis of NO-associated inflammatory diseases by focusing on changes in gene expression.


Subject(s)
Gene Expression Profiling , Nitric Oxide , Transcriptome , Humans , Nitric Oxide/metabolism , Transcriptome/drug effects , Cell Adhesion/drug effects , Cell Adhesion/genetics , HEK293 Cells , Cell Movement/drug effects , Cell Movement/genetics , Inflammation/genetics , Inflammation/chemically induced , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Int Heart J ; 65(3): 498-505, 2024.
Article in English | MEDLINE | ID: mdl-38825494

ABSTRACT

This study aimed to explore the expression of long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) in patients with acute myocardial infarction (AMI) and its inflammatory regulation mechanism through miR-211/interleukin 10 (IL-10) axis.A total of 75 participants were enrolled in this study: 25 healthy people in the control group, 25 patients with stable angina pectoris (SAP) in the SAP group, and 25 patients with AMI in the AMI group. Real-time qPCR was used to detect mRNA expression levels of NEAT1, miR-211, and IL-10. The interaction between miR-211, NEAT1, and IL-10 was confirmed by dual-luciferase reporter assay, and protein expression was detected using western blot.High expression of NEAT1 in peripheral blood mononuclear cells (PBMCs) of patients with AMI was negatively related to serum creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), tumor necrosis factor-α (TNF-α), IL-6, and IL-1ß and was positively correlated with left ventricular ejection fraction (LVEF). In THP-1 cells, miR-211 was confirmed to target and inhibit IL-10 expression. NEAT1 knockdown and miR-211-mimic markedly decreased IL-10 protein levels, whereas anti-miR-211 markedly increased IL-10 protein levels. Importantly, miR-211 level was negatively related to NEAT1 and IL-10 levels, whereas IL-10 level was positively related to the level of NEAT1 expression in PBMCs of patients with AMI.LncRNA NEAT1 was highly expressed in PBMCs of patients with AMI, and NEAT1 suppressed inflammation via miR-211/IL-10 axis in PBMCs of patients with AMI.


Subject(s)
Interleukin-10 , Leukocytes, Mononuclear , MicroRNAs , Myocardial Infarction , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/blood , MicroRNAs/blood , MicroRNAs/genetics , Interleukin-10/blood , Interleukin-10/metabolism , Myocardial Infarction/blood , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Leukocytes, Mononuclear/metabolism , Male , Female , Middle Aged , Aged , Inflammation/genetics , Inflammation/blood , Inflammation/metabolism , Case-Control Studies
9.
Parasite Immunol ; 46(6): e13039, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38838041

ABSTRACT

Ticks are notorious blood-sucking ectoparasites that affect both humans and animals. They serve as a unique vector of various deadly diseases. Here, we have shown the roles of the receptor for advanced glycation end products (RAGE) during repeated infestations by the tick Haemaphysalis longicornis using RAGE-/- mice. In primary infestation, a large blood pool developed, which was flooded with numerous RBCs, especially during the rapid feeding phase of the tick both in wild-type (wt) and RAGE-/- mice. Very few inflammatory cells were detected around the zones of haemorrhage in the primary infestations. However, the number of inflammatory cells gradually increased in the subsequent tick infestations, and during the third infestations, the number of inflammatory cells reached to the highest level (350.3 ± 16.8 cells/focus). The site of attachment was totally occupied by the inflammatory cells in wt mice, whereas very few cells were detected at the ticks' biting sites in RAGE-/- mice. RAGE was highly expressed during the third infestation in wt mice. In the third infestation, infiltration of CD44+ lymphocytes, eosinophils and expression of S100A8 and S100B significantly increased at the biting sites of ticks in wt, but not in RAGE-/- mice. In addition, peripheral eosinophil counts significantly increased in wt but not in RAGE-/- mice. Taken together, our study revealed that RAGE-mediated inflammation and eosinophils played crucial roles in the tick-induced inflammatory reactions.


Subject(s)
Inflammation , Ixodidae , Mice, Knockout , Receptor for Advanced Glycation End Products , Tick Infestations , Animals , Ixodidae/genetics , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/genetics , Mice , Tick Infestations/immunology , Mice, Inbred C57BL , Female , Feeding Behavior , Haemaphysalis longicornis
10.
Cell Metab ; 36(6): 1269-1286.e9, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38838640

ABSTRACT

Patients with metabolic dysfunction-associated steatotic liver disease (MASLD), especially advanced metabolic dysfunction-associated steatohepatitis (MASH), have an increased risk of cardiovascular diseases (CVDs). Whether CVD events will, in turn, influence the pathogenesis of MASLD remains unknown. Here, we show that myocardial infarction (MI) accelerates hepatic pathological progression of MASLD. Patients with MASLD who experience CVD events after their diagnosis exhibit accelerated liver fibrosis progression. MI promotes hepatic fibrosis in mice with MASH, accompanied by elevated circulating Ly6Chi monocytes and their recruitment to damaged liver tissues. These adverse effects are significantly abrogated when deleting these cells. Meanwhile, MI substantially increases circulating and cardiac periostin levels, which act on hepatocytes and stellate cells to promote hepatic lipid accumulation and fibrosis, finally exacerbating hepatic pathological progression of MASH. These preclinical and clinical results demonstrate that MI alters systemic homeostasis and upregulates pro-fibrotic factor production, triggering cross-disease communication that accelerates hepatic pathological progression of MASLD.


Subject(s)
Disease Progression , Mice, Inbred C57BL , Myocardial Infarction , Animals , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Humans , Mice , Male , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Monocytes/metabolism , Female , Middle Aged , Inflammation/pathology , Inflammation/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/pathology , Liver/metabolism , Cell Adhesion Molecules/metabolism
11.
Sci Rep ; 14(1): 12637, 2024 06 02.
Article in English | MEDLINE | ID: mdl-38825605

ABSTRACT

Osteoporosis (OP) is a bone metabolism disease that is associated with inflammatory pathological mechanism. Nonetheless, rare studies have investigated the diagnostic effectiveness of immune-inflammation index in the male population. Therefore, it is interesting to achieve early diagnosis of OP in male population based on the inflammatory makers from blood routine examination. We developed a prediction model based on a training dataset of 826 Chinese male patients through a retrospective study, and the data was collected from January 2022 to May 2023. All participants underwent the dual-energy X-ray absorptiometry (DXEA) and blood routine examination. Inflammatory markers such as systemic immune-inflammation index (SII) and platelet-to-lymphocyte ratio (PLR) was calculated and recorded. We utilized the least absolute shrinkage and selection operator (LASSO) regression model to optimize feature selection. Multivariable logistic regression analysis was applied to construct a predicting model incorporating the feature selected in the LASSO model. This predictive model was displayed as a nomogram. Receiver operating characteristic (ROC) curve, C-index, calibration curve, and clinical decision curve analysis (DCA) to evaluate model performance. Internal validation was test by the bootstrapping method. This study was approved by the Ethic Committee of the First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine (Ethic No. JY2023012) and conducted in accordance with the relevant guidelines and regulations. The predictive factors included in the prediction model were age, BMI, cardiovascular diseases, cerebrovascular diseases, neuropathy, thyroid diseases, fracture history, SII, PLR, C-reactive protein (CRP). The model displayed well discrimination with a C-index of 0.822 (95% confidence interval: 0.798-0.846) and good calibration. Internal validation showed a high C-index value of 0.805. Decision curve analysis (DCA) showed that when the threshold probability was between 3 and 76%, the nomogram had a good clinical value. This nomogram can effectively predict the incidence of OP in male population based on SII and PLR, which would help clinicians rapidly and conveniently diagnose OP with men in the future.


Subject(s)
Inflammation , Nomograms , Osteoporosis , Humans , Male , Osteoporosis/diagnosis , Osteoporosis/blood , Middle Aged , Retrospective Studies , Aged , Inflammation/blood , Inflammation/diagnosis , China/epidemiology , Risk Factors , Biomarkers/blood , Absorptiometry, Photon , ROC Curve , Adult , Risk Assessment/methods
12.
Anal Chim Acta ; 1312: 342747, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38834275

ABSTRACT

BACKGROUND: Lipid droplets (LDs) polarity is intricately linked to diverse biological processes and diseases. The visualization of LDs-polarity is of vital importance but challenging due to the lack of high-specificity, high-sensitivity and large-Stokes shift probes for real-time tracking LDs-polarity in biological systems. RESULTS: Four D-π-A based fluorescent probes (TPA-TCF1-TPA-TCF4) have been developed by combining tricyanofuran (an electron acceptor, A) and triphenylamine (an electron donor, D) derivatives with different terminal groups. Among them, TPA-TCF1 and TPA-TCF4 exhibit excellent polar sensitivity, large Stokes shift (≥182 nm in H2O), and efficient LDs targeting ability. In particular, TPA-TCF4 is capable of monitoring the change of LDs-polarity during ferroptosis, inflammation, apoptosis of cancer cell, and fatty liver. SIGNIFICANCE: All these features render TPA-TCF4 a versatile tool for pharmacodynamic evaluation of anti-cancer drugs, in-depth understanding of the biological effect of LDs on ferroptosis, and medical diagnosis of LDs-polarity related diseases.


Subject(s)
Fatty Liver , Ferroptosis , Fluorescent Dyes , Inflammation , Lipid Droplets , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Humans , Ferroptosis/drug effects , Fatty Liver/drug therapy , Fatty Liver/metabolism , Fluorescent Dyes/chemistry , Inflammation/drug therapy , Inflammation/metabolism , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Molecular Structure
13.
J Vet Sci ; 25(3): e35, 2024 May.
Article in English | MEDLINE | ID: mdl-38834505

ABSTRACT

IMPORTANCE: Experimental autoimmune encephalomyelitis (EAE) is an animal model of multiple sclerosis characterized by inflammation within the central nervous system. However, inflammation in non-neuronal tissues, including the lungs, has not been fully evaluated. OBJECTIVE: This study evaluated the inflammatory response in lungs of EAE mice by immunohistochemistry and histochemistry. METHODS: Eight adult C57BL/6 mice were injected with myelin oligodendrocyte glycoprotein35-55 to induce the EAE. Lungs and spinal cords were sampled from the experimental mice at the time of sacrifice and used for the western blotting, histochemistry, and immunohistochemistry. RESULTS: Histopathological examination revealed inflammatory lesions in the lungs of EAE mice, characterized by infiltration of myeloperoxidase (MPO)- and galectin-3-positive cells, as determined by immunohistochemistry. Increased numbers of collagen fibers in the lungs of EAE mice were confirmed by histopathological analysis. Western blotting revealed significantly elevated level of osteopontin (OPN), cluster of differentiation 44 (CD44), MPO and galectin-3 in the lungs of EAE mice compared with normal controls (p < 0.05). Immunohistochemical analysis revealed both OPN and CD44 in ionized calcium-binding adapter molecule 1-positive macrophages within the lungs of EAE mice. CONCLUSIONS AND RELEVANCE: Taken together, these findings suggest that the increased OPN level in lungs of EAE mice led to inflammation; concurrent increases in proinflammatory factors (OPN and galectin-3) caused pulmonary impairment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Lung , Mice, Inbred C57BL , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Lung/pathology , Female , Immunohistochemistry , Osteopontin/metabolism , Galectin 3/metabolism , Peroxidase/metabolism , Hyaluronan Receptors/metabolism , Spinal Cord/pathology , Inflammation/pathology , Blotting, Western
15.
Neurosurg Rev ; 47(1): 256, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38834876

ABSTRACT

OBJECTIVE: White blood cells (WBC) play an important role in the inflammatory response of the body. Elevated WBC counts on admission in patients with subarachnoid hemorrhage (SAH) correlate with a poor prognosis. However, the role of longitudinal WBC trajectories based on repeated WBC measurements during hospitalization remains unclear. We explored the association between different WBC trajectory patterns and in-hospital mortality. METHODS: We analyzed a cohort of consecutive patients with SAH between 2012 and 2020. Group-based trajectory modeling (GBTM) was used to group the patients according to their white blood cell patterns over the first 4 days. Stabilized inverse probability treatment weighting (sIPTW) was used to balance baseline demographic and clinical characteristics. We analyzed the association between the WBC trajectory groups and in-hospital mortality using a Cox proportional hazards model. RESULTS: In total, 506 patients with SAH were included in this retrospective cohort. The final model identified two distinct longitudinal WBC trajectories. After adjusting for confounding factors, multivariate regression analysis suggested that an elevated longitudinal WBC trajectory increased the risk of in-hospital mortality (hazard ratio [HR], 2.476; 95% confidence interval [CI] 1.081-5.227; P = 0.024) before sIPTW, and (HR, 2.472; 95%CI 1.489-4.977; P = 0.018) after sIPTW. CONCLUSION: In patients with SAH, different clinically relevant groups could be identified using WBC trajectory analysis. The WBC count trajectory-initially elevated and then decreased- may lead to an increased risk of in-hospital mortality following SAH.


Subject(s)
Hospital Mortality , Subarachnoid Hemorrhage , Humans , Subarachnoid Hemorrhage/mortality , Subarachnoid Hemorrhage/blood , Male , Female , Middle Aged , Aged , Leukocyte Count , Retrospective Studies , Inflammation , Adult , Prognosis , Cohort Studies
16.
J Nanobiotechnology ; 22(1): 314, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840113

ABSTRACT

Osteoporosis is the most common bone metabolic disease that affects the health of middle-aged and elderly people, which is hallmarked by imbalanced bone remodeling and a deteriorating immune microenvironment. Magnesium and calcium are pivotal matrix components that participate in the bone formation process, especially in the immune microenvironment regulation and bone remodeling stages. Nevertheless, how to potently deliver magnesium and calcium to bone tissue remains a challenge. Here, we have constructed a multifunctional nanoplatform composed of calcium-based upconversion nanoparticles and magnesium organic frameworks (CM-NH2-PAA-Ald, denoted as CMPA), which features bone-targeting and pH-responsive properties, effectively regulating the inflammatory microenvironment and promoting the coordination of osteogenic functions for treating osteoporosis. The nanoplatform can efficaciously target bone tissue and gradually degrade in response to the acidic microenvironment of osteoporosis to release magnesium and calcium ions. This study validates that CMPA possessing favorable biocompatibility can suppress inflammation and facilitate osteogenesis to treat osteoporosis. Importantly, high-throughput sequencing results demonstrate that the nanoplatform exerts a good inflammatory regulation effect through inhibition of the nuclear factor kappa-B signaling pathway, thereby normalizing the osteoporotic microenvironment. This collaborative therapeutic strategy that focuses on improving bone microenvironment and promoting osteogenesis provides new insight for the treatment of metabolic diseases such as osteoporosis.


Subject(s)
Calcium , Magnesium , Nanoparticles , Osteogenesis , Osteoporosis , Osteogenesis/drug effects , Osteoporosis/drug therapy , Magnesium/pharmacology , Magnesium/chemistry , Calcium/metabolism , Animals , Nanoparticles/chemistry , Mice , Inflammation/drug therapy , Bone and Bones/drug effects , Bone and Bones/metabolism , Humans , Cellular Microenvironment/drug effects , Female , NF-kappa B/metabolism
17.
J Transl Med ; 22(1): 535, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840216

ABSTRACT

BACKGROUND: Inflammation and endothelial barrier dysfunction are the major pathophysiological changes in acute respiratory distress syndrome (ARDS). Sphingosine-1-phosphate receptor 3 (S1PR3), a G protein-coupled receptor, has been found to mediate inflammation and endothelial cell (EC) integrity. However, the function of S1PR3 in ARDS has not been fully elucidated. METHODS: We used a murine lipopolysaccharide (LPS)-induced ARDS model and an LPS- stimulated ECs model to investigate the role of S1PR3 in anti-inflammatory effects and endothelial barrier protection during ARDS. RESULTS: We found that S1PR3 expression was increased in the lung tissues of mice with LPS-induced ARDS. TY-52156, a selective S1PR3 inhibitor, effectively attenuated LPS-induced inflammation by suppressing the expression of proinflammatory cytokines and restored the endothelial barrier by repairing adherens junctions and reducing vascular leakage. S1PR3 inhibition was achieved by an adeno-associated virus in vivo and a small interfering RNA in vitro. Both the in vivo and in vitro studies demonstrated that pharmacological or genetic inhibition of S1PR3 protected against ARDS by inhibiting the NF-κB pathway and improving mitochondrial oxidative phosphorylation. CONCLUSIONS: S1PR3 inhibition protects against LPS-induced ARDS via suppression of pulmonary inflammation and promotion of the endothelial barrier by inhibiting NF-κB and improving mitochondrial oxidative phosphorylation, indicating that S1PR3 is a potential therapeutic target for ARDS.


Subject(s)
Lipopolysaccharides , Mice, Inbred C57BL , Mitochondria , NF-kappa B , Oxidative Phosphorylation , Respiratory Distress Syndrome , Sphingosine-1-Phosphate Receptors , Animals , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , NF-kappa B/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Male , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/antagonists & inhibitors , Humans , Lung/pathology , Lung/drug effects , Lung/metabolism , Mice , Inflammation/pathology , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Protective Agents/pharmacology , Cytokines/metabolism
18.
J Cell Mol Med ; 28(11): e18476, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38842136

ABSTRACT

Osteoarthritis (OA) is a complicated disease that involves apoptosis and mitophagy. MST1 is a pro-apoptotic factor. Hence, decreasing its expression plays an anti-apoptotic effect. This study aims to investigate the protective effect of MST1 inhibition on OA and the underlying processes. Immunofluorescence (IF) was used to detect MST1 expression in cartilage tissue. Western Blot, ELISA and IF were used to analyse the expression of inflammation, extracellular matrix (ECM) degradation, apoptosis and mitophagy-associated proteins. MST1 expression in chondrocytes was inhibited using siRNA and shRNA in vitro and in vivo. Haematoxylin-Eosin, Safranin O-Fast Green and alcian blue staining were used to evaluate the therapeutic effect of inhibiting MST1. This study discovered that the expression of MST1 was higher in OA patients. Inhibition of MST1 reduced inflammation, ECM degradation and apoptosis and enhanced mitophagy in vitro. MST1 inhibition slows OA progression in vivo. Inhibiting MST1 suppressed apoptosis, inflammation and ECM degradation via promoting Parkin-mediated mitophagy and the Nrf2-NF-κB axis. The results suggest that MST1 is a possible therapeutic target for the treatment of osteoarthritis as its inhibition delays the progression of OA through the Nrf2-NF-κB axis and mitophagy.


Subject(s)
Apoptosis , Chondrocytes , Disease Progression , Mitophagy , NF-E2-Related Factor 2 , NF-kappa B , Osteoarthritis , Signal Transduction , Ubiquitin-Protein Ligases , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/genetics , Mitophagy/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NF-kappa B/metabolism , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Apoptosis/genetics , Male , Mice , Extracellular Matrix/metabolism , Gene Knockdown Techniques , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins
19.
Crit Rev Eukaryot Gene Expr ; 34(5): 45-57, 2024.
Article in English | MEDLINE | ID: mdl-38842203

ABSTRACT

Inflammation-mediated dysfunction of cardiomyocytes is the main cause of diabetic cardiomyopathy (DCM). The present study aimed to investigate the roles of siah E3 ubiquitin protein ligase 1 (SIAH1) in DCM. The online dataset GSE4172 was used to analyze the differentially expressed genes in myocardial inflammation of DCM patients. RT-qPCR was conducted to detect mRNA levels. Enzyme-Linked Immunosorbent Assay (ELISA) was performed to detect cytokine release. Western blot was used to detect protein expression. Lactate dehydrogenase (LDH) assay was used to determine cytotoxicity. In vitro ubiquitination assay was applied to determine the ubiquitination of nuclear factor kappa B inhibitor alpha (1κВ-α). Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was used to detect the death of cardiomyocytes. Flow cytometry was applied for determining cardiomyocyte pyroptosis. The results showed that SIAH1 was overexpressed in human inflammatory cardiomyopathy. High expression of SIAH1 was associated with inflammatory response. SIAH1 was also overexpressed lipopolysaccharide (LPS)-induced inflammatory cardiomyopathy model in vitro. However, SIAH1 knockdown suppressed the inflammatory-related pyroptosis of cardiomyocytes. SIAH1 promoted the ubiquitination of 1κВ-α and activated nuclear factor kappa В (NF-κВ) signaling, which promoted the pyroptosis of cardiomyocytes. In conclusion, SIAH1 exacerbated the progression of human inflammatory cardiomyopathy via inducing the ubiquitination of 1κВ-α and activation of NF-κВ signaling. Therefore, SIAHI/IκB-α/NF-κB signaling may be a potential target for human inflammatory cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies , Myocytes, Cardiac , NF-kappa B , Pyroptosis , Signal Transduction , Ubiquitin-Protein Ligases , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Humans , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , NF-KappaB Inhibitor alpha/metabolism , NF-KappaB Inhibitor alpha/genetics , Ubiquitination , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics
20.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 92-96, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836675

ABSTRACT

The currest study aimed to measure the effects of laparoscopic radical gastrectomy on inflammatory response along with immune function in gastric cancer (GC) patients. Seventy patients with GC in our hospital were retrospectively chosen to be the study objects and separated into control group (CG, 35 cases) and observation group (OG, 35 cases). Patients in the OG received radical laparotomy. Patients in the OG received laparoscopic radical gastrectomy. The surgical indicators, postoperative recovery indicators, inflammatory factors, immune function, incidence of adverse reactions along with quality of life of patients in both groups were compared. In contrast to the CG, the operation time of the OG presented as shorter (P<0.05), and the amount of intraoperative blood loss together with postoperative VAS score in the OG presented lower (P<0.05), but the number of lymph nodes dissection presented not statistically significant between 2 groups (P>0.05). The postoperative exhaust time, feeding time as well as hospital stay in the OG presented shorter relative to the CG (P<0.05). The serum levels of CRP, and IL-6 together with TNF-α presented elevated in both groups after surgery, and those in the OG presented lower when compared with the CG (P<0.05). The serum levels of IgA, and IgG together with IgM presented declined in both groups after surgery, and those in the OG presented higher when compared with the CG (P<0.05). The incidence of postoperative complications in the OG presented reduction relative to the CG (P<0.05). The GLQI scores of the OG presented significantly higher relative to the CG at discharge (P<0.05). Compared with radical gastrectomy, laparoscopic radical gastrectomy is more suitable for the treatment of GC, which can reduce the inflammatory response and promote the immune function of GC patients.


Subject(s)
Gastrectomy , Inflammation , Laparoscopy , Stomach Neoplasms , Humans , Stomach Neoplasms/surgery , Stomach Neoplasms/immunology , Gastrectomy/methods , Gastrectomy/adverse effects , Laparoscopy/adverse effects , Laparoscopy/methods , Male , Female , Middle Aged , Inflammation/immunology , Aged , Quality of Life , Retrospective Studies , C-Reactive Protein/metabolism , Postoperative Complications/immunology , Postoperative Complications/etiology , Postoperative Complications/epidemiology , Tumor Necrosis Factor-alpha/blood , Interleukin-6/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...