Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Free Radic Biol Med ; 220: 271-287, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38734267

ABSTRACT

Bilirubin-induced brain damage is a serious clinical consequence of hyperbilirubinemia, yet the underlying molecular mechanisms remain largely unknown. Ferroptosis, an iron-dependent cell death, is characterized by iron overload and lipid peroxidation. Here, we report a novel regulatory mechanism of demethylase AlkB homolog 5 (ALKBH5) in acyl-coenzyme A synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis in hyperbilirubinemia. Hyperdifferential PC12 cells and newborn Sprague-Dawley rats were used to establish in vitro and in vivo hyperbilirubinemia models, respectively. Proteomics, coupled with bioinformatics analysis, first suggested the important role of ferroptosis in hyperbilirubinemia-induced brain damage. In vitro experiments showed that ferroptosis is activated in hyperbilirubinemia, and ferroptosis inhibitors (desferrioxamine and ferrostatin-1) treatment effectively alleviates hyperbilirubinemia-induced oxidative damage. Notably, we observed that the ferroptosis in hyperbilirubinemia was regulated by m6A modification through the downregulation of ALKBH5 expression. MeRIP-seq and RIP-seq showed that ALKBH5 may trigger hyperbilirubinemia ferroptosis by stabilizing ACSL4 mRNA via m6A modification. Further, hyperbilirubinemia-induced oxidative damage was alleviated through ACSL4 genetic knockdown or rosiglitazone-mediated chemical repression but was exacerbated by ACSL4 overexpression. Mechanistically, ALKBH5 promotes ACSL4 mRNA stability and ferroptosis by combining the 669 and 2015 m6A modified sites within 3' UTR of ACSL4 mRNA. Our findings unveil a novel molecular mechanism of ferroptosis and suggest that m6A-dependent ferroptosis could be an underlying clinical target for the therapy of hyperbilirubinemia.

2.
World J Pediatr ; 2024 May 07.
Article in English | MEDLINE | ID: mdl-38713366

ABSTRACT

BACKGROUND: SARS-CoV-2 continues to mutate over time, and reports on children infected with Omicron BA.5 are limited. We aimed to analyze the specific symptoms of Omicron-infected children and to improve patient care. METHODS: We selected 315 consecutively hospitalized children with Omicron BA.5 and 16,744 non-Omicron-infected febrile children visiting the fever clinic at our hospital between December 8 and 30, 2022. Specific convulsions and body temperatures were compared between the two cohorts. We analyzed potential associations between convulsions and vaccination, and additionally evaluated the brain damage among severe Omicron-infected children. RESULTS: Convulsion rates (97.5% vs. 4.3%, P < 0.001) and frequencies (median: 2.0 vs. 1.6, P < 0.001) significantly differed between Omicron-infected and non-Omicron-infected febrile children. The body temperatures of Omicron-infected children were significantly higher during convulsions than when they were not convulsing and those of non-Omicron-infected febrile children during convulsions (median: 39.5 vs. 38.2 and 38.6 °C, both P < 0.001). In the three Omicron-subgroups, the temperature during convulsions was proportional to the percentage of patients and significantly differed ( P < 0.001), while not in the three non-Omicron-subgroups ( P = 0.244). The convulsion frequency was lower in the 55 vaccinated children compared to the 260 non-vaccinated children (average: 1.8 vs. 2.1, P < 0.001). The vaccination dose and convulsion frequency in Omicron-infected children were significantly correlated ( P < 0.001). Fifteen of the 112 severe Omicron cases had brain damage. CONCLUSIONS: Omicron-infected children experience higher body temperatures and frequencies during convulsions than those of non-Omicron-infected febrile children. We additionally found evidence of brain damage caused by infection with omicron BA.5. Vaccination and prompt fever reduction may relieve symptoms.

3.
Immunol Lett ; 267: 106865, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38705483

ABSTRACT

PURPOSE: To investigate the relationship between the lipid profiles of patients with primary Sjögren's syndrome (pSS) and other clinical characteristics, laboratory examination, disease activity, and inflammatory factors. In addition, the risk factors for hyperlipidemia-related complications of pSS and the effect of hydroxychloroquine (HCQ) usage on the lipid profile were incorporated into this study. METHODS: This is a single-center, retrospective study that included 367 patients who were diagnosed with pSS at Tongji Hospital, School of Medicine, Tongji University, China from January 2010 to March 2022. Initially, demographic information, clinical characteristics, medication records, and complications of the patients were gathered. A case-control analysis compared the 12 systems involvement (ESSDAI domain), clinical symptoms, and laboratory tests between pSS patients with and without dyslipidemia. A simple linear regression model was employed to investigate the relationship between serum lipid profile and inflammatory factors. Logistics regression analysis was performed to assess variables for hyperlipidemia-related complications of pSS. The paired t-test was then used to evaluate the improvement in lipid profile among pSS patients. RESULTS: 48.7 % of all pSS patients had dyslipidemia, and alterations in lipid levels were related to gender, age, and smoking status but not body mass index (BMI). Dyslipidemia is more prevalent in pSS patients who exhibit heightened autoimmunity and elevated levels of inflammation. Higher concentrations of multiple highly inflammatory factors correlate with a more severe form of dyslipidemia. Non-traditional cardiovascular risk factors may contribute to hyperlipidemia-related complications of pSS, such as increased, low complement 3 (C3) and low C4. According to our study, HCQ usage may protect against lipid-related disease in pSS. CONCLUSION: Attention should be paid to the dyslipidemia of pSS. This research aims to clarify the population portrait of pSS patients with abnormal lipid profiles and provides insights into the correlation between metabolism and inflammation in individuals with pSS and the potential role they play in the advancement of the disease. These findings provide novel avenues for further understanding the underlying mechanisms of pSS pathogenesis.

4.
Sci Data ; 11(1): 444, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702302

ABSTRACT

With the rapid global warming in recent decades, the Tibetan Plateau (TP) has suffered severe impacts, such as glacier retreat, glacial lake expansion, and permafrost degradation, which threaten the lives and properties of the local and downstream populations. Regional Reanalysis (RR) is vital for TP due to the limitations of observations. In this work, a 62-year (1961-2022) long atmospheric regional reanalysis with spatial resolution of 9 km (convective gray-zone scale) and temporal resolution of 1 hour over the TP (TPRR) was developed using the Weather Research and Forecasting (WRF) model, combined with re-initialization method, spectral nudging (SN), and several optimizations. TPRR is forced by ERA5 at hourly intervals. TPRR outperforms ERA5, realistically capturing climatological characteristics and seasonal variations of precipitation and T2m (air temperature at 2m above ground level). Moreover, TPRR better reproduces the frequency and intensity of precipitation, as well as the diurnal cycle of precipitation. This study also quantifies the wetting trend of 0.0071 mm/year over the TP amid global warming using TPRR.

5.
Front Immunol ; 15: 1289492, 2024.
Article in English | MEDLINE | ID: mdl-38510251

ABSTRACT

Sjögren's syndrome (SjS) is a systemic, highly diverse, and chronic autoimmune disease with a significant global prevalence. It is a complex condition that requires careful management and monitoring. Recent research indicates that epigenetic mechanisms contribute to the pathophysiology of SjS by modulating gene expression and genome stability. DNA methylation, a form of epigenetic modification, is the fundamental mechanism that modifies the expression of various genes by modifying the transcriptional availability of regulatory regions within the genome. In general, adding a methyl group to DNA is linked with the inhibition of genes because it changes the chromatin structure. DNA methylation changes the fate of multiple immune cells, such as it leads to the transition of naïve lymphocytes to effector lymphocytes. A lack of central epigenetic enzymes frequently results in abnormal immune activation. Alterations in epigenetic modifications within immune cells or salivary gland epithelial cells are frequently detected during the pathogenesis of SjS, representing a robust association with autoimmune responses. The analysis of genome methylation is a beneficial tool for establishing connections between epigenetic changes within different cell types and their association with SjS. In various studies related to SjS, most differentially methylated regions are in the human leukocyte antigen (HLA) locus. Notably, the demethylation of various sites in the genome is often observed in SjS patients. The most strongly linked differentially methylated regions in SjS patients are found within genes regulated by type I interferon. This demethylation process is partly related to B-cell infiltration and disease progression. In addition, DNA demethylation of the runt-related transcription factor (RUNX1) gene, lymphotoxin-α (LTA), and myxovirus resistance protein A (MxA) is associated with SjS. It may assist the early diagnosis of SjS by serving as a potential biomarker. Therefore, this review offers a detailed insight into the function of DNA methylation in SjS and helps researchers to identify potential biomarkers in diagnosis, prognosis, and therapeutic targets.


Subject(s)
Autoimmune Diseases , Sjogren's Syndrome , Humans , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation
6.
Environ Pollut ; 349: 123875, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38548152

ABSTRACT

With the evidence emerging that abnormal expression of long noncoding RNAs (lncRNAs) are involved in onset of Parkinson's disease (PD), the role of NR_030777 contributing to this disease is of great interest. We recently found that a novel lncRNA "NR_030777" demonstrates protective effects on PQ-induced neurodegeneration. However, the underlying molecular mechanisms of NR_030777 in the regulation of mitochondrial fission and mitophagy involved in PQ-induced neuronal damage remain to be explored. NR_030777 brain conditional overexpressing mice as well as in vitro primary neuronal cells from cerebral cortex and Neuro2a cells were adopted. Immunofluorescence, Immunohistochemistry, qRT-PCR and Western blotting were used to evaluate the expression levels of RNA and proteins. RNA immunoprecipitation and RNA pulldown experiment were used to evaluate the interaction of NR_030777 with its target proteins. NR_030777 and mitophagy were increased, and tyrosine hydroxylase (TH) levels recovered after NR_030777 overexpression upon PQ treatment. The overexpression and knockdown of NR_030777 unveiled that NR_030777 positively regulated mitophagy such as the upregulation of LC3B-II:I, ATG12-ATG5, p62 and NBR1. Moreover, the application of mdivi-1, a DRP-1 inhibitor, in combination with NR_030777 genetic modified cells unveiled that NR_030777 promoted DRP1-mediated mitochondrial fission and mitophagy. Furthermore, NR_030777 were directly bound to CDK1 to increase p-DRP1 levels at the Ser616 site, leading to mitochondrial fission and mitophagy. On the other hand, NR_030777 acted directly on ATG12 within the ATG12-ATG5 complex in the 800-1400 nt region to modulate the membrane formation. Accordingly, NR_030777 deficiency in neuron cells compromised cell mitophagy. Finally, the above findings were confirmed using NR_030777-overexpressing mice. NR_030777 exerted a protective effect on PQ-exposed mice by enhancing mitophagy. Our data provide the first scientific evidence for the precise invention of PQ-induced PD. Our findings further propose a breakthrough for understanding the regulatory relationship between NR_030777, CDK1, ATG12 and mitophagy in PQ-induced PD.


Subject(s)
CDC2 Protein Kinase , Mitochondrial Dynamics , Mitophagy , Paraquat , Parkinson Disease , RNA, Long Noncoding , Animals , Mitophagy/drug effects , Mitochondrial Dynamics/drug effects , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/genetics , Paraquat/toxicity , Neurons/metabolism , Neurons/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
7.
Sci Data ; 10(1): 890, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38086806

ABSTRACT

U-Net, a deep-learning convolutional neural network, is used to downscale coarse meteorological data. Based on 19 models from the Coupled Model Intercomparison Project Phase 6 and the Multi-Source Weather (MSWX) dataset, bias correction and UNet downscaling approaches are used to develop high resolution dataset over the East Asian region, referred to as Climate Change for East Asia with Bias corrected UNet Dataset (CLIMEA-BCUD). CLIMEA-BCUD provides nine meteorological variables including 2-m air temperature, 2-m daily maximum air temperature, 2-m daily minimum air temperature, precipitation, 10-m wind speed, 2-m relative humidity, 2-m specific humidity, downward shortwave radiation and downward longwave radiation with 0.1° horizontal resolution at daily intervals over the historical period of 1950-2014 and three future scenarios (SSP1-2.6, SSP2-4.5 and SSP5-8.5) of 2015-2100. Validation against MSWX indicates that CLIMEA-BCUD shows reasonable performance in terms of climatology, and it is capable of simulating seasonal cycles and future changes well. It is suggested that CLIMEA-BCUD can promote the application of deep learning in climate research in the areas of climate change, hydrology, etc.

8.
BMC Pulm Med ; 23(1): 473, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38007449

ABSTRACT

INTRODUCTION: Tofacitinib, a selective inhibitor of JAK1 and/or JAK3, is considered to alleviate the pulmonary condition of primary Sjögren's syndrome (pSS)-associated interstitial lung disease (ILD) through its anti-inflammatory and antifibrotic effects. METHODS AND ANALYSIS: This is a single-center, prospective, randomized, open-label trial. The trial will compare a 52-week course of oral tofacitinib with traditional therapy cyclophosphamide (CYC) combined with azathioprine (AZA) in the treatment of pSS-ILD. A total of 120 patients will be randomly assigned into two treatment groups with a 1:1 ratio and followed for 52 weeks from the first dose. The primary endpoint of the study is the increase of forced vital capacity (FVC) at 52 weeks. Secondary endpoints include high-resolution computed tomography (HRCT), diffusion capacity for carbon monoxide of the lung (DLCO), the Mahler dyspnea index, the health-related quality of life (HARQoL) score, the cough symptom score, EULAR Sjögren's syndrome disease activity index (ESSDAI), and safety. DISCUSSION: This study will be the first randomized controlled trial to investigate tofacitinib compared to the traditional regimen of CYC in combination with AZA in the treatment of pSS-ILD, which will provide data on efficacy and safety and further elucidate the role of the JAK-STAT signaling pathway in the development of pSS-ILD. ETHICS AND DISSEMINATION: Before starting the experiment, the research proposal, informed consent (ICF) and relevant documents in accordance with the ethical principles of the Helsinki Declaration and the relevant requirements of the local GCP rules for ethical approval shall be submitted to the ethics committee of the hospital. The ethical approval of this study is reviewed by the Ethics Committee of Tongji Hospital and the ethical approval number is 2021-LCYJ-007. When the experiment is completed, the results will also be disseminated to patients and the public through publishing papers in international medical journals. TRIAL REGISTRATION: The study was registered on the Chinese Clinical Trial Registry, www.chictr.org.cn ; ID ChiCTR2000031389.


Subject(s)
Lung Diseases, Interstitial , Sjogren's Syndrome , Humans , Azathioprine , Cyclophosphamide/therapeutic use , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/complications , Prospective Studies , Quality of Life , Randomized Controlled Trials as Topic , Sjogren's Syndrome/complications , Sjogren's Syndrome/drug therapy
9.
Mol Med Rep ; 28(6)2023 Dec.
Article in English | MEDLINE | ID: mdl-37937619

ABSTRACT

Ferroptosis is driven by iron­dependent accumulation of lipid hydroperoxides, and hemolytic hyperbilirubinemia causes accumulation of unconjugated bilirubin and iron. The present study aimed to assess the role of ferroptosis in hemolytic hyperbilirubinemia­induced brain damage (HHIBD). Rats were randomly divided into the control, phenylhydrazine (PHZ) and deferoxamine (DFO) + PHZ groups, with 12 rats in each group. Ferroptosis­associated biochemical and protein indicators were measured in the brain tissue of rats. We also performed tandem mass tag­labeled proteomic analysis. The levels of iron and malondialdehyde were significantly higher and levels of glutathione (GSH) and superoxide dismutase activity significantly lower in the brain tissues of the PHZ group compared with those in the control group. HHIBD also resulted in significant increases in the expression of the ferroptosis­related proteins acyl­CoA synthetase long­chain family member 4, ferritin heavy chain 1 and transferrin receptor and divalent metal transporter 1, as well as a significant reduction in the expression of ferroptosis suppressor protein 1. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis demonstrated that the differentially expressed proteins of rat brain tissues between the control and PHZ groups were significantly involved in ferroptosis, GSH metabolism and fatty acid biosynthesis pathways. Pretreatment with DFO induced antioxidant activity and alleviated lipid peroxidation­mediated HHIBD. In addition, PC12 cells treated with ferric ammonium citrate showed shrinking mitochondria, high mitochondrial membrane density, and increased lipid reactive oxygen species and intracellular ferrous iron, which were antagonized by pretreatment with ferrostatin­1 or DFO, which was reversed by pretreatment with ferrostatin­1 or DFO. The present study demonstrated that ferroptosis is involved in HHIBD and provided novel insights into candidate proteins that are potentially involved in ferroptosis in the brain during hemolytic hyperbilirubinemia.


Subject(s)
Brain Injuries , Ferroptosis , Rats , Animals , Proteomics , Apoptosis , Iron/metabolism , Hyperbilirubinemia , Brain Injuries/metabolism , Brain/metabolism , Hemolysis , Glutathione/metabolism , Lipids
10.
Nat Commun ; 14(1): 7257, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37945564

ABSTRACT

To mitigate climate warming, many countries have committed to achieve carbon neutrality in the mid-21st century. Here, we assess the global impacts of changing greenhouse gases (GHGs), aerosols, and tropospheric ozone (O3) following a carbon neutrality pathway on climate and extreme weather events individually using the Community Earth System Model version 1 (CESM1). The results suggest that the future aerosol reductions significantly contribute to climate warming and increase the frequency and intensity of extreme weathers toward carbon neutrality and aerosol impacts far outweigh those of GHGs and tropospheric O3. It reverses the knowledge that the changing GHGs dominate the future climate changes as predicted in the middle of the road pathway. Therefore, substantial reductions in GHGs and tropospheric O3 are necessary to reach the 1.5 °C warming target and mitigate the harmful effects of concomitant aerosol reductions on climate and extreme weather events under carbon neutrality in the future.

11.
Front Psychol ; 14: 1201481, 2023.
Article in English | MEDLINE | ID: mdl-37705952

ABSTRACT

Introduction: In light of the COVID-19 pandemic, there is an increased need for potential travelers to gather information about their trips to mitigate perceived risks. This study aims to understand the relationship between the intensity of media use (both new and traditional), epidemic risk perception, and tourism protection behavior intention among potential tourists. Methods: A total of 491 valid questionnaires were collected in Shanghai, China. Factor analysis, path analysis, and effect analysis were conducted using SPSS and AMOS to examine the impact of different media types on epidemic risk perception and tourism protection behavior. Results: The findings indicate a positive association between new media use intensity and epidemic risk perception, as well as an intention to adopt safety-conscious tourism behaviors. In contrast, traditional media usage is inversely associated with risk perception but has no significant influence on protective behavior. The results also highlight the role of demographic factors, such as age, education level, occupation, and income, in modulating the relationship between media usage and risk perception. Discussion: The contrasting effects of new and traditional media suggest the need for a tailored approach in epidemic communication strategies. Public health officials should leverage new media to enhance risk perception and safety-oriented behaviors, while recognizing the role of traditional media in managing lower risk perceptions and assuaging panic. The study emphasizes the importance of personalized messaging based on demographic disparities in media usage and perception. The mediating role of risk perception in shaping protective behaviors offers insights for promoting adherence to safety protocols. Conclusion: This study contributes to a comprehensive understanding of media influences during health crises, emphasizing the responsibility of media platforms in transmitting accurate information. The findings call for a nuanced approach to epidemic communication, considering the strengths and weaknesses of different media types. Segmented and personalized messaging strategies can cater to demographic variations in media usage and perception. Enhancing risk perception through tailored messaging can promote protective behaviors and effectively manage public sentiment during health crises.

12.
Int J Rheum Dis ; 26(10): 1951-1959, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37550949

ABSTRACT

OBJECTIVE: To explore the clinical characteristics of rheumatic disease (RD) patients who suffered from moderate or severe coronavirus disease 2019 (COVID-19) infection and to evaluate risk factors of COVID-19 infection in RD patients. METHODS: A retrospective analysis was conducted on 148 moderate or severe COVID-19 patients admitted to the First People's Hospital of Suqian Affiliated to Nanjing Medical University, including 74 RD patients and 74 non-RD patients. Clinical data were collected including clinical characteristics and laboratory tests. RESULTS: The RD group showed a higher proportion of females with a higher incidence of interstitial lung disease and kidney disease than the non-RD group. Also, the incidence of fatigue, olfactory dysfunction and musculoskeletal pain was higher in the RD group, but the incidence of cough, wheezing, and fever was lower compared with non-RD patients. The hospitalized course of the RD group (12.7 days ± 6.55) was significantly longer than that in the non-RD group (8.07 days ± 3.40). Also, patients in the RD group had higher levels of erythrocyte sedimentation rate, interleukin (IL)-2, and IL-4 than the non-RD group. The logistic regression analysis showed that dizziness and headache, C-reactive protein (CRP) > 8 mg/L and lactate dehydrogenase (LDH) > 248 µ/L were independent risk factors for severe COVID-19 infections of RD patients. CONCLUSION: RD patients who suffered from moderate or severe COVID-19 infections have a higher risk of comorbidities, higher levels of inflammation, and longer hospitalized course. Dizziness and headache, CRP > 8 mg/L and LDH > 248 µ/L are risk factors for severe COVID-19 infections in RD patients.

13.
Ther Adv Chronic Dis ; 14: 20406223231181490, 2023.
Article in English | MEDLINE | ID: mdl-37485232

ABSTRACT

Background: Patients with primary Sjögren's syndrome (pSS) are at increased risk of cardiovascular morbidity as compared with the general population. Objectives: A retrospective study on 349 Chinese patients with pSS was conducted to identify potential risk factors for cardiovascular events and develop a cardiovascular risk nomogram. Design: This is a retrospective observational study. Methods: The study included 349 patients who were diagnosed with pSS at Tongji Hospital, School of Medicine, Tongji University, China from January 2010 to March 2022. The least absolute shrinkage and selection operator (LASSO) was used to select features for the cardiovascular risk model. The features selected in LASSO were used to build the cardiovascular risk model in a multivariate logistic regression analysis. C-index, receiver operating characteristic (ROC) curve, calibration plot, and decision curve analysis were used to assess the predictive model. Internal validation was performed by bootstrapping. Results: Sex, joint pain as an initial symptom, dry mouth, oral ulcers, dental caries, Raynaud's phenomenon, fatigue, diabetes, elevated thyroid-stimulating hormone (TSH) level, and elevated systolic blood pressure were included in the nomogram for the prediction of cardiovascular risk. Our model had good discrimination (C-index: 0.824, 95% confidence interval: 0.712-0.936) and good calibration (C-index in the interval validation: 0.8). Decision curve analysis indicated that our nomogram demonstrated clinical usefulness for intervention in a cardiovascular disease possibility threshold of 3%. Conclusion: The cardiovascular risk nomogram incorporating sex, initial joint pain, dry mouth, oral ulcer, dental caries, Raynaud's phenomenon, fatigue, diabetes, elevated TSH, and systolic blood pressure could be used in the prediction of cardiovascular risk in patients with pSS and the guidance of further treatment.

14.
Cytokine ; 168: 156227, 2023 08.
Article in English | MEDLINE | ID: mdl-37244248

ABSTRACT

Primary Sjögren's syndrome (pSS) is an autoimmune disease that targets exocrine glands, leading to exocrine dysfunction. Due to its propensity to infect epithelial and B cells, Epstein-Barr virus (EBV) is hypothesized to be related with pSS. Through molecular mimicry, the synthesis of specific antigens, and the release of inflammatory cytokines, EBV contributes to the development of pSS. Lymphoma is the most lethal outcome of EBV infection and the development of pSS. As a population-wide virus, EBV has had a significant role in the development of lymphoma in people with pSS. In the review, we will discuss the possible causes of the disease.


Subject(s)
Autoimmune Diseases , Epstein-Barr Virus Infections , Lymphoma , Sjogren's Syndrome , Humans , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Autoimmune Diseases/complications
15.
Sci Total Environ ; 881: 163429, 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37072102

ABSTRACT

Cobalt exposure, even at low concentrations, induces neurodegenerative damage, such as Alzheimer's disease (AD). The specific underlying mechanisms remain unclear. Our previous study demonstrated that m6A methylation alteration is involved in cobalt-induced neurodegenerative damage, such as in AD. However, the role of m6A RNA methylation and its underlying mechanisms are poorly understood. In this study, both epidemiological and laboratory studies showed that cobalt exposure could downregulate the expression of the m6A demethylase ALKBH5, suggesting a key role for ALKBH5. Moreover, Methylated RNA immunoprecipitation and sequencing (MeRIP-seq) analysis revealed that ALKBH5 deficiency is associated with neurodegenerative diseases. KEGG pathway and Gene ontology analyses further revealed that the differentially m6A-modified genes resulting from ALKBH5 downregulation and cobalt exposure were aggregated in the pathways of proliferation, apoptosis, and autophagy. Subsequently, ALKBH5 deficiency was shown to exacerbate cell viability decline, motivate cell apoptosis and attenuate cell autophagy induced by cobalt with experimental techniques of gene overexpression/inhibition. In addition, morphological changes in neurons and the expression of AD-related proteins, such as APP, P-Tau, and Tau, in the cerebral hippocampus of wild-type and ALKBH5 knockout mice after chronic cobalt exposure were also investigated. Both in vitro and in vivo results showed that lower expression of ALKBH5 aggravated cobalt-induced neurodegenerative damage. These results suggest that ALKBH5, as an epigenetic regulator, could be a potential target for alleviating cobalt-induced neurodegenerative damage. In addition, we propose a novel strategy for the prevention and treatment of environmental toxicant-related neurodegeneration from an epigenetic perspective.


Subject(s)
Cobalt , RNA , Mice , Animals , Cobalt/toxicity , Methylation
16.
Zhongguo Dang Dai Er Ke Za Zhi ; 25(4): 401-407, 2023 Apr 15.
Article in Chinese | MEDLINE | ID: mdl-37073846

ABSTRACT

A boy, aged 16 months, attended the hospital due to head and facial erythema for 15 months and vulva erythema for 10 months with aggravation for 5 days. The boy developed perioral and periocular erythema in the neonatal period and had erythema and papules with desquamation and erosion in the neck, armpit, and trigone of vulva in infancy. Blood gas analysis showed metabolic acidosis; the analysis of amino acid and acylcarnitine profiles for inherited metabolic diseases and the analysis of organic acid in urine suggested multiple carboxylase deficiency; genetic testing showed a homozygous mutation of c.1522C>T(p.R508W) in the HLCS gene. Finally the boy was diagnosed with holocarboxylase synthetase deficiency and achieved a good clinical outcome after oral biotin treatment. This article analyzes the clinical data of a child with holocarboxylase synthetase deficiency and summarizes the etiology, diagnosis, and treatment of this child, so as to provide ideas for clinicians to diagnose this rare disease.


Subject(s)
Holocarboxylase Synthetase Deficiency , Humans , Male , Biotin/genetics , Biotin/therapeutic use , Holocarboxylase Synthetase Deficiency/genetics , Holocarboxylase Synthetase Deficiency/diagnosis , Holocarboxylase Synthetase Deficiency/drug therapy , Homozygote , Mutation , Rare Diseases/drug therapy , Infant
17.
J Hazard Mater ; 453: 131354, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37054644

ABSTRACT

Cobalt is the most widely used heavy metal pollutant in medicine and industry. Excessive cobalt exposure can adversely affect human health. Neurodegenerative symptoms have been observed in cobalt-exposed populations; however, the underlying mechanisms remain largely unknown. In this study, we demonstrate that the N6-methyladenosine (m6A) demethylase fat mass and obesity-associated gene (FTO) mediates cobalt-induced neurodegeneration by impairing autophagic flux. Cobalt-induced neurodegeneration was exacerbated through FTO genetic knockdown or repression of demethylase activity, but was alleviated by FTO overexpression. Mechanistically, we showed that FTO regulates TSC1/2-mTOR signaling pathway by targeting TSC1 mRNA stability in an m6A-YTHDF2 manner, which resulted in autophagosome accumulation. Furthermore, FTO decreases lysosome-associated membrane protein-2 (LAMP2) to inhibit the integration of autophagosomes and lysosomes, leading to autophagic flux damage. In vivo experiments further identified that central nervous system (CNS)-Fto-specific knockout resulted in serious neurobehavioral and pathological damage as well as TSC1-related autophagy impairment in cobalt-exposed mice. Interestingly, FTO-regulated autophagy impairment has been confirmed in patients with hip replacement. Collectively, our results provide novel insights into m6A-modulated autophagy through FTO-YTHDF2 targeted TSC1 mRNA stability, revealing cobalt is a novel epigenetic hazard that induces neurodegeneration. These findings suggest the potential therapeutic targets for hip replacement in patients with neurodegenerative damage.


Subject(s)
Autophagy , Cobalt , Animals , Humans , Mice , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Cobalt/toxicity , Obesity , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism
18.
Ecotoxicol Environ Saf ; 255: 114804, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36948007

ABSTRACT

Paraquat (PQ) has been widely acknowledged as an environmental risk factor for Parkinson's disease (PD). However, the interaction between splicing factor and long non-coding RNA (lncRNA) in the process of PQ-induced PD has rarely been studied. Based on previous research, this study focused on splicing factor 3 subunit 3 (SF3B3) and lncRNA NR_030777. After changing the target gene expression level by lentiviral transfection technology, the related gene expression was detected by western blot and qRT-PCR. The expression of SF3B3 protein was reduced in Neuro-2a cells after PQ exposure, and the reactive oxygen species (ROS) scavenger N-acetylcysteine prevented this decline. Knockdown of SF3B3 reduced the PQ-triggered NR_030777 expression increase, and overexpression of NR_030777 reduced the transcriptional and translational level of Sf3b3. Then, knockdown of SF3B3 exacerbated the PQ-induced decrease in cell viability and aggravated the reduction of tyrosine hydroxylase (TH) protein expression. Overexpressing SF3B3 reversed the reduction of TH expression caused by PQ. Moreover, after intervention with the autophagy inhibitor Bafilomycin A1, LC3B-II protein expression was further increased in Neuro-2a cells with the knockdown of SF3B3, indicating that autophagy was enhanced. In conclusion, PQ modulated the interplay between NR_030777 and SF3B3 through ROS production, thereby impairing autophagic flux and causing neuronal damage.


Subject(s)
Paraquat , RNA, Long Noncoding , Acetylcysteine/pharmacology , Neurons/metabolism , Paraquat/toxicity , Reactive Oxygen Species/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA Splicing Factors/metabolism
19.
J Orthop Surg Res ; 18(1): 41, 2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36647153

ABSTRACT

OBJECTIVE: To identify primary Sjögren's syndrome (pSS) patients with arthralgia at risk for osteoarthritis (OA) or arthritis. METHODS: This study included 368 pSS patients admitted to a mono-centric from March 2010 to December 2020. Patients were divided into groups according to whether complicated with OA or arthritis. Data were analyzed to determine the differences in demographical characteristics, symptoms, and laboratory examination. RESULTS: The involvement of the OA joints was predominately knee and spine sites (including cervical and lumbar spine degeneration). When diagnosing arthritis, it was mainly peripheral symmetric polyarthritis, the most affected sites were the interphalangeal and metacarpophalangeal joints. There were significant differences in age, disease duration, uric acid (UA), and total cholesterol (TC) between pSS-OA and pSS-nOA patients (P < 0.050). Logistic regression analysis showed that age (OR = 1.965; P = 0.009) and joint pain (OR = 3.382; P < 0.001) were dangerous factors associated with OA. Interestingly, although the level of UA, TC, and triglycerides (TG) was shown to be positive with OA, there was no statistical significance after the OR was computed in the four-cell table. In pSS-arthritis, EULAR Sjögren's syndrome disease activity index (ESSDAI) (P = 0.011), the frequency of joint pain (P < 0.001), and muscular involvement (P = 0.037) were higher than non-arthritis group. In pSS patients only presenting with joint pain, arthritis patients had higher ESSDAI and system involvements, but lower UA and TG levels compared with OA group (P < 0.050). CONCLUSION: In pSS patients with arthralgia, OA accounted for the majority. pSS patients with advanced age and more pronounced metabolic characteristics, such as elevated blood lipids and uric acid, was a key factor in groups at risk for OA. However, arthritis patients had higher rates of dry mouth and eye, higher disease activity, antibodies positive, and more organs damage. In the future, it may be necessary to be more cautious in the diagnosis of joint manifestations in pSS patients in order to make the appropriate treatments.


Subject(s)
Osteoarthritis , Sjogren's Syndrome , Humans , Sjogren's Syndrome/complications , Sjogren's Syndrome/diagnosis , Uric Acid , Osteoarthritis/complications , Arthralgia/diagnosis , Arthralgia/etiology , Metacarpophalangeal Joint
20.
Sci Total Environ ; 857(Pt 2): 159432, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36243078

ABSTRACT

Cobalt is an environmental toxicant, and excessive bodily exposure can damage the nervous system. Particularly, our previous study reported that low-dose cobalt (significantly less than the safety threshold) is still able to induce neurodegenerative changes. However, the underlying molecular mechanism is still insufficient revealed. Herein, we further investigate the molecular mechanism between cobalt-induced neurodegeneration and autophagy, as well as explore the interplay between hypoxia-inducible factor-1α (HIF-1α), reactive oxygen species (ROS), and autophagy in cobalt-exposed mice and human neuroglioma cells. We first reveal cobalt as an environmental toxicant to severely induce ß amyloid (Aß) deposition, tau hyperphosphorylation, and dysregulated autophagy in the hippocampus and cortex of mice. In particular, we further identify that cobalt-induced neurotoxicity is triggered by the impairment of autophagic flux in vitro experiments. Moreover, the mechanistic study reveals that cobalt exposure extremely activates HIF-1α expression to facilitate the overproduction of ROS. Then, elevated ROS can target the amino-threonine kinase (AKT)-mammalian target of rapamycin (mTOR)-Unc-51 like autophagy activating kinase 1 (ULK1) signaling pathway to participate in cobalt-induced impairment of autophagic flux. Subsequently, defected autophagy further exacerbates cobalt-induced neurotoxicity for its unable to eliminate the deposition of pathological protein. Therefore, our data provide scientific evidence for cobalt safety evaluation and risk assessment and propose a breakthrough for understanding the regulatory relationship between HIF-1α, ROS, and autophagy in cobalt-induced neurodegeneration.


Subject(s)
Cobalt , Hypoxia-Inducible Factor 1, alpha Subunit , Reactive Oxygen Species , Animals , Humans , Mice , Autophagy/physiology , Cobalt/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...