Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Mol Biol Rep ; 51(1): 593, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38683404

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a common central nervous system neurodegenerative disease. Neuroinflammation is one of the significant neuropathological hallmarks. As a traditional Chinese medicine, Safranal exerts anti-inflammatory effects in various diseases, however, whether it plays a similar effect on PD is still unclear. The study was to investigate the effects and mechanism of Safranal on PD. METHODS: The PD mouse model was established by 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine MPTP firstly. Next, the degree of muscle stiffness, neuromuscular function, motor retardation and motor coordination ability were examined by observing and testing mouse movement behavior. Immunofluorescence staining was used to observe the expression of tyrosine hydroxylase (TH). The dopamine (DA) content of the striatum was detected by High-performance liquid chromatography (HPLC). The expression of TH and NLRP3 inflammasome-related markers NLRP3, IL-1ß, and Capase-1 were detected by Real-time Polymerase Chain Reaction (qRT-PCR) and western blotting (WB) respectively. RESULTS: Through behavioral testing, Parkinson's mouse showed a higher muscle stiffness and neuromuscular tension, a more motor retardation and activity disorders, together with a worse motor coordination compared with sham group. Simultaneously, DA content and TH expression in the striatum were decreased. However, after using Safranal treatment, the above pathological symptoms of Parkinson's mouse all improved compared with Safranal untreated group, the DA content and TH expression were also increased to varying degrees. Surprisingly, it observed a suppression of NLRP3 inflammation in the striatum of Parkinson's mouse. CONCLUSIONS: Safranal played a neuroprotective effect on the Parkinson's disease and its mechanism was related to the inhibition of NLRP3 inflammasome activation.


Subject(s)
Cyclohexenes , Disease Models, Animal , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroprotective Agents , Parkinson Disease , Terpenes , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Terpenes/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Male , Cyclohexenes/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Mice, Inbred C57BL , Inflammation/drug therapy , Inflammation/metabolism , Dopamine/metabolism , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Interleukin-1beta/metabolism , Tyrosine 3-Monooxygenase/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Caspase 1/metabolism
2.
J Virol ; 98(4): e0014624, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38440983

ABSTRACT

Peste des petits ruminants is an acute and highly contagious disease caused by the Peste des petits ruminants virus (PPRV). Host proteins play a crucial role in viral replication. However, the effect of fusion (F) protein-interacting partners on PPRV infection is poorly understood. In this study, we found that the expression of goat plasminogen activator urokinase (PLAU) gradually decreased in a time- and dose-dependent manner in PPRV-infected goat alveolar macrophages (GAMs). Goat PLAU was subsequently identified using co-immunoprecipitation and confocal microscopy as an F protein binding partner. The overexpression of goat PLAU inhibited PPRV growth and replication, whereas silencing goat PLAU promoted viral growth and replication. Additionally, we confirmed that goat PLAU interacted with a virus-induced signaling adapter (VISA) to antagonize F-mediated VISA degradation, increasing the production of type I interferon. We also found that goat PLAU reduced the inhibition of PPRV replication in VISA-knockdown GAMs. Our results show that the host protein PLAU inhibits the growth and replication of PPRV by VISA-triggering RIG-I-like receptors and provides insight into the host protein that antagonizes PPRV immunosuppression.IMPORTANCEThe role of host proteins that interact with Peste des petits ruminants virus (PPRV) fusion (F) protein in PPRV replication is poorly understood. This study confirmed that goat plasminogen activator urokinase (PLAU) interacts with the PPRV F protein. We further discovered that goat PLAU inhibited PPRV replication by enhancing virus-induced signaling adapter (VISA) expression and reducing the ability of the F protein to degrade VISA. These findings offer insights into host resistance to viral invasion and suggest new strategies and directions for developing PPR vaccines.


Subject(s)
Goat Diseases , Goats , Host-Pathogen Interactions , Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Urokinase-Type Plasminogen Activator , Viral Fusion Proteins , Animals , Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/metabolism , Goat Diseases/immunology , Goat Diseases/metabolism , Goat Diseases/virology , Goats/immunology , Goats/virology , Macrophages, Alveolar , Peste-des-Petits-Ruminants/immunology , Peste-des-Petits-Ruminants/metabolism , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/growth & development , Peste-des-petits-ruminants virus/immunology , Peste-des-petits-ruminants virus/metabolism , Protein Binding , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Viral Fusion Proteins/metabolism
3.
J Virol ; 98(1): e0159923, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38169281

ABSTRACT

African swine fever virus (ASFV) causes a highly contagious and deadly disease in domestic pigs and European wild boars, posing a severe threat to the global pig industry. ASFV CP204L, a highly immunogenic protein, is produced during the early stages of ASFV infection. However, the impact of CP204L protein-interacting partners on the outcome of ASFV infection is poorly understood. To accomplish this, coimmunoprecipitation and mass spectrometry analysis were conducted in ASFV-infected porcine alveolar macrophages (PAMs). We have demonstrated that sorting nexin 32 (SNX32) is a CP204L-binding protein and that CP204L interacted and colocalized with SNX32 in ASFV-infected PAMs. ASFV growth and replication were promoted by silencing SNX32 and suppressed by overexpressing SNX32. SNX32 degraded CP204L by recruiting the autophagy-related protein Ras-related protein Rab-1b (RAB1B). RAB1B overexpression inhibited ASFV replication, while knockdown of RAB1B had the opposite effect. Additionally, RAB1B, SNX32, and CP204L formed a complex upon ASFV infection. Taken together, this study demonstrates that SNX32 antagonizes ASFV growth and replication by recruiting the autophagy-related protein RAB1B. This finding extends our understanding of the interaction between ASFV CP204L and its host and provides new insights into exploring the relationship between ASFV infection and autophagy.IMPORTANCEAfrican swine fever (ASF) is a highly contagious and acute hemorrhagic viral disease with a high mortality near 100% in domestic pigs. ASF virus (ASFV), which is the only member of the family Asfarviridae, is a dsDNA virus of great complexity and size, encoding more than 150 proteins. Currently, there are no available vaccines against ASFV. ASFV CP204L represents the most abundantly expressed viral protein early in infection and plays an important role in regulating ASFV replication. However, the mechanism by which the interaction between ASFV CP204L and host proteins affects ASFV replication remains unclear. In this study, we demonstrated that the cellular protein SNX32 interacted with CP204L and degraded CP204L by upregulating the autophagy-related protein RAB1B. In summary, this study will help us understand the interaction mechanism between CP204L and its host upon infection and provide new insights for the development of vaccines and antiviral drugs.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antiviral Restriction Factors , Autophagy , Sorting Nexins , rab1 GTP-Binding Proteins , Animals , Autophagy-Related Proteins/metabolism , Sus scrofa/virology , Swine/virology , Sorting Nexins/metabolism , Antiviral Restriction Factors/metabolism , rab1 GTP-Binding Proteins/metabolism , Macrophages/virology , Virus Replication
4.
Mol Neurobiol ; 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38008889

ABSTRACT

Central sensitization is an important pathophysiological mechanism underlying chronic migraine (CM). Previous studies have shown that microglial activation and subsequent inflammation in the trigeminal nucleus caudalis (TNC) contribute to central sensitization. Toll-like receptor 2 (TLR2) is a receptor expressed on the membrane of microglia and participates in central sensitization in inflammatory and chronic pain; however, its role in CM is unclear. Therefore, this study investigated TLR2 involvement in CM in detail. Mice treated with recurrent nitroglycerin (NTG) were used as a CM model. Hyperalgesia was assessed using a 50% paw mechanical threshold and a 50% periorbital threshold on a Von Frey filament pain meter. Western blotting and immunofluorescence analyses were used to detect the expression of TLR2, microglia, c-fos and CGRP in TNC. The expression of inflammatory factors (IL-6, IL-1ß、 IL-10、TNF-α and IFN-ß1) was detected using quantitative real-time polymerase chain reaction (qRT-PCR). A selective TLR2 antagonist (C29) was systematically administered to observe its effect on hyperalgesia, microglia activation and the expression of c-fos, CGRP and inflammatory factors. Recurrent administration of NTG resulted in acute and chronic hypersensitivity, accompanied by upregulation of TLR2 expression and microglial activation in TNC. C29 partially inhibited pain hypersensitivity. C29 suppressed microglial activation induced by NTG administration. Inhibition of TLR2 reduced the expression of c-fos and CGRP in TNC after NTG treatment. C29 inhibited the expression of inflammatory mediators in TNC. These data showed that microglial TLR2 plays a critical role in the pathogenesis of CM by regulating microglial activation in TNC.

5.
Vet Microbiol ; 284: 109836, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37574636

ABSTRACT

African swine fever (ASF) is an acute, severe, and highly contagious disease caused by the African swine fever virus (ASFV), which infects domestic pigs and wild boars. The incidence and mortality rates of swine infected with virulent strains of ASFV can reach up to 100%. The large genome, its complex structure, multiple genotypes, and a lack of understanding regarding ASFV gene function are serious obstacles to the development of safe and effective vaccines. Here, ASFV I329L was identified as a relatively conserved gene that is expressed during the late stage of infection. A recombinant virus with I329L gene deletion (ASFV CN/GS/2018-ΔI329L) was produced by replacing I329L with an enhanced green fluorescent protein (EGFP) cassette. In order to explore the function of the ASFV I329L gene, transcriptome sequencing (RNA-seq) was performed on porcine alveolar macrophages (PAMs) infected with ASFV CN/GS/2018 and ASFV CN/GS/2018-ΔI329L. GO functional and KEGG pathway analyses were performed to analyze differentially expressed genes, and different alternative splicing (AS) events were also analyzed. We compared the sequencing data for each sample with the ASFV CN/GS/2018 reference sequence. Interestingly, we found 3 and 1 up-regulated genes and 12 and 19 down-regulated genes at 12 and 24 h post-infection, respectively. In addition, we verified the expression of 5 up-regulated and 5 down-regulated genes by RT-qPCR, and the results were consistent with those obtained based on RNA-seq. In summary, the results obtained in this study provide new insights for further elucidation of ASFV proteins and ASFV-host interactions. These findings will contribute to implementing a comprehensive strategy for controlling the spread of ASF.


Subject(s)
African Swine Fever Virus , African Swine Fever , Swine Diseases , Swine , Animals , Sus scrofa , Genotype , Gene Expression Profiling/veterinary
6.
Front Microbiol ; 14: 1190052, 2023.
Article in English | MEDLINE | ID: mdl-37396386

ABSTRACT

Organic fertilizers can partially replace chemical fertilizers to improve agricultural production and reduce negative environmental impacts. To study the effect of organic fertilizer on soil microbial carbon source utilization and bacterial community composition in the field of rain-fed wheat, we conducted a field experiment from 2016 to 2017 in a completely randomized block design with four treatments: the control with 100% NPK compound fertilizer (N: P2O5: K2O = 20:10:10) of 750 kg/ha (CK), a combination of 60% NPK compound fertilizer with organic fertilizer of 150 kg/ha (FO1), 300 kg/ha (FO2), and 450 kg/ha (FO3), respectively. We investigated the yield, soil property, the utilization of 31 carbon sources by soil microbes, soil bacterial community composition, and function prediction at the maturation stage. The results showed that (1) compared with CK, organic fertilizer substitution treatments improved ear number per hectare (13%-26%), grain numbers per spike (8%-14%), 1000-grain weight (7%-9%), and yield (3%-7%). Organic fertilizer substitution treatments increased the total nitrogen, available nitrogen, available phosphorus, and soil organic matter contents by 26%, 102%, 12%, and 26%, respectively, compared with CK treatments. Organic fertilizer substitution treatments significantly advanced the partial productivity of fertilizers. (2) Carbohydrates and amino acids were found to be the most sensitive carbon sources for soil microorganisms in different treatments. Particularly for FO3 treatment, the utilization of ß-Methyl D-Glucoside, L-Asparagine acid, and glycogen by soil microorganisms was higher than other treatments and positively correlated with soil nutrients and wheat yield. (3) Compared with CK, organic fertilizer substitution treatments increased the relative abundance of Proteobacteria, Acidobacteria, and Gemmatimonadetes and decreased the relative abundance of Actinobacteria and Firmicutes. Interestingly, FO3 treatment improved the relative abundance of Nitrosovibrio, Kaistobacter, Balneimonas, Skermanella, Pseudomonas, and Burkholderia belonging to Proteobacteria and significantly boosted the relative abundance of function gene K02433 [the aspartyl-tRNA (Asn)/glutamyl-tRNA (Gln)]. Based on the abovementioned findings, we suggest FO3 as the most appropriate organic substitution method in rain-fed wheat fields.

7.
Environ Pollut ; 332: 121958, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37286026

ABSTRACT

Hydrogen sulfide (H2S) is a multifunctional gaseous signaling molecule involved in the regulation of Cr stress responses. In the present study, we combined transcriptomic and physiological analyses to elucidate the mechanism underlying the mitigation of Cr toxicity by H2S in maize (Zea mays L.). We showed that treatment with sodium hydrosulfide (NaHS, a donor of H2S) partially alleviated Cr-induced growth inhibition. However, Cr uptake was not affected. RNA sequencing suggested that H2S regulates the expression of many genes involved in pectin biosynthesis, glutathione metabolism, and redox homeostasis. Under Cr stress, NaHS treatment significantly increased pectin content and pectin methylesterase activity; thus, more Cr was retained in the cell wall. NaHS application also increased the content of glutathione and phytochelatin, which chelate Cr and transport it into vacuoles for sequestration. Furthermore, NaHS treatment mitigated Cr-induced oxidative stress by enhancing the capacity of enzymatic and non-enzymatic antioxidants. Overall, our results strongly support that H2S alleviates Cr toxicity in maize by promoting Cr sequestration and re-establishing redox homeostasis rather than by reducing Cr uptake from the environment.


Subject(s)
Hydrogen Sulfide , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Zea mays/metabolism , Chromium/toxicity , Glutathione/metabolism , Oxidation-Reduction , Homeostasis
8.
J Virol ; 97(5): e0022823, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37162350

ABSTRACT

African swine fever (ASF), caused by the African swine fever virus (ASFV), is a transboundary infectious disease of domestic pigs and wild boars, resulting in significant swine production losses. Currently, no effective commercial ASF vaccines or therapeutic options are available. A previous study has shown that deletions of ASFV MGF110-9L and MGF505-7R genes (ASFV-Δ110-9L/505-7R) attenuated virulence in pigs and provided complete protection against parental lethal ASFV CN/GS/2018 (wild-type ASFV [ASFV-WT]) challenge, but the underlying mechanism is unclear. This study found that ASFV-Δ110-9L/505-7R weakened TBK1 degradation compared with ASFV-WT through RNA sequencing (RNA-seq) and Western blotting analyses. Furthermore, we confirmed that ASFV-Δ110-9L/505-7R blocked the degradation of TBK1 through the autophagy pathway. We also identified that the downregulation of an autophagy-related protein PIK3C2B was involved in the inhibition of TBK1 degradation induced by ASFV-Δ110-9L/505-7R. Additionally, we also confirmed that PIK3C2B promoted ASFV-Δ110-9L/505-7R replication in vitro. Together, this study elucidated a novel mechanism of virulence change of ASFV-Δ110-9L/505-7R, revealing a new mechanism of ASF live attenuated vaccines (LAVs) and providing theoretical guidance for the development of ASF vaccines. IMPORTANCE African swine fever (ASF) is a contagious and lethal hemorrhagic disease of pigs caused by the African swine fever virus (ASFV), leading to significant economic consequences for the global pig industry. The development of an effective and safe ASF vaccine has been unsuccessful. Previous studies have shown that live attenuated vaccines (LAVs) of ASFV are the most effective vaccine candidates to prevent ASF. Understanding the host responses caused by LAVs of ASFV is important in optimizing vaccine design and diversifying the resources available to control ASF. Recently, our laboratory found that the live attenuated ASFV-Δ110-9L/505-7R provided complete protection against parental ASFV-WT challenge. This study further demonstrated that ASFV-Δ110-9L/505-7R inhibits TBK1 degradation mediated by an autophagy activator PIK3C2B to increase type I interferon production. These results revealed an important mechanism for candidate vaccine ASFV-Δ110-9L/505-7R, providing strategies for exploring the virulence of multigene-deleted live attenuated ASFV strains and the development of vaccines.


Subject(s)
African Swine Fever Virus , African Swine Fever , Interferon Type I , Viral Vaccines , Animals , African Swine Fever/prevention & control , African Swine Fever Virus/genetics , Interferon Type I/metabolism , Sus scrofa , Swine , Vaccines, Attenuated , Genes, Viral
9.
FASEB J ; 37(6): e22934, 2023 06.
Article in English | MEDLINE | ID: mdl-37144880

ABSTRACT

African swine fever (ASF) caused by African swine fever virus (ASFV) is a devastating disease for the global pig industry and economic benefit. The limited knowledge on the pathogenesis and infection mechanisms of ASF restricts progress toward vaccine development and ASF control. Previously, we illustrated that deletion of the MGF-110-9L gene from highly virulent ASFV CN/GS/2018 strains (ASFV∆9L) results in attenuated virulence in swine, but the underlying mechanism remains unclear. In this study, we found that the difference in virulence between wild-type ASFV (wt-ASFV) and ASFV∆9L strains was mainly caused by the difference in TANK Binding Kinase 1 (TBK1) reduction. TBK1 reduction was further identified to be mediated by the autophagy pathway and this degradative process requires the up-regulation of a positive autophagy regulation molecule- Phosphatidylinositol-4-Phosphate 3-Kinase Catalytic Subunit Type 2 Beta (PIK3C2B). Moreover, TBK1 over-expression was confirmed to inhibit ASFV replication in vitro. In summary, these results indicate that wt-ASFV counteracts type I interferon (IFN) production by degrading TBK1, while ASFVΔ9L enhanced type I IFN production by weakening TBK1 reduction, clarifying the mechanism that ASFVΔ9L present the attenuated virulence in vitro.


Subject(s)
African Swine Fever Virus , African Swine Fever , Interferon Type I , Swine , Animals , African Swine Fever Virus/genetics , African Swine Fever/genetics , African Swine Fever/prevention & control , Virulence , Gene Expression , Interferon Type I/metabolism , Gene Deletion
10.
J Biol Chem ; 299(6): 104767, 2023 06.
Article in English | MEDLINE | ID: mdl-37142221

ABSTRACT

African swine fever, caused by a large icosahedral DNA virus (African swine fever virus, ASFV), is a highly contagious disease in domestic and feral swine, thus posing a significant economic threat to the global swine industry. Currently, there are no effective vaccines or the available methods to control ASFV infection. Attenuated live viruses with deleted virulence factors are considered to be the most promising vaccine candidates; however, the mechanism by which these attenuated viruses confer protection is unclear. Here, we used the Chinese ASFV CN/GS/2018 as a backbone and used homologous recombination to generate a virus in which MGF110-9L and MGF360-9L, two genes antagonize host innate antiviral immune response, were deleted (ASFV-ΔMGF110/360-9L). This genetically modified virus was highly attenuated in pigs and provided effective protection of pigs against parental ASFV challenge. Importantly, we found ASFV-ΔMGF110/360-9L infection induced higher expression of Toll-like receptor 2 (TLR2) mRNA compared with parental ASFV as determined by RNA-Seq and RT-PCR analysis. Further immunoblotting results showed that parental ASFV and ASFV-ΔMGF110/360-9L infection inhibited Pam3CSK4-triggered activating phosphorylation of proinflammatory transcription factor NF-κB subunit p65 and phosphorylation of NF-κB inhibitor IκBα levels, although NF-κB activation was higher in ASFV-ΔMGF110/360-9L-infected cells compared with parental ASFV-infected cells. Additionally, we show overexpression of TLR2 inhibited ASFV replication and the expression of ASFV p72 protein, whereas knockdown of TLR2 had the opposite effect. Our findings suggest that the attenuated virulence of ASFV-ΔMGF110/360-9L might be mediated by increased NF-κB and TLR2 signaling.


Subject(s)
African Swine Fever Virus , African Swine Fever , Viral Proteins , Animals , African Swine Fever/immunology , African Swine Fever/virology , African Swine Fever Virus/genetics , African Swine Fever Virus/pathogenicity , Antibody Formation/immunology , Gene Deletion , NF-kappa B/genetics , Swine , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Transcriptome , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication/immunology
11.
Small ; 19(2): e2204782, 2023 01.
Article in English | MEDLINE | ID: mdl-36412068

ABSTRACT

Nanozymes and amorphous nanomaterials attract great attention owing to their extraordinary properties. However, the requirements for special synthesis conditions become the bottleneck of their development. Herein, a new strategy involving the DNA-based coordination-driven self-assembly is reported for the synthesis of a novel amorphous/crystalline hetero-phase nanozyme (Fe-DNA). For the synthesis of both nanozymes and amorphous materials, this strategy is simple and controllable, avoiding the traditionally employed harsh conditions. Benefitting from the amorphous structure and the superior physicochemical properties, the synthesized Fe-DNA nanozyme is subsequently found to exhibit a smaller Michaelis constant value for hydrogen peroxide (H2 O2 ) (0.81 mm) than that of horseradish peroxidase (HRP) (3.70 mm), demonstrating the stronger affinity of the Fe-DNA nanozyme toward H2 O2 . The Fe-DNA nanozyme also shows significant peroxidase-like activity but only negligible oxidase-like activity, a characteristic which releases the corresponding assay system from oxygen interference, thereby improving the performance of the nanozyme-based sensing platform. In addition, compared with other nanozymes, the novel Fe-DNA nanozyme is degradable via phosphate; thus, mitigating potential environmental threat. This work provides novel amorphous/crystalline hetero-phase nanozymes and opens a new avenue for the design of amorphous nanomaterials and nanozymes.


Subject(s)
Biosensing Techniques , Peroxidase , Peroxidases/chemistry , Oxidoreductases , DNA , Hydrogen Peroxide
12.
Materials (Basel) ; 15(19)2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36234376

ABSTRACT

In order to obtain the optimal electrode layout and ice melting effect of cast conductive asphalt concrete steel bridge deck pavement, firstly, pouring conductive asphalt concrete was prepared; secondly, different electrode materials and layout methods were selected to test the heating rate of the specimen from start to 120 min, and the electrode materials and layout methods were optimized. Then, the finite element analysis software ANSYS was used to build the model for heating and ice melting simulation, and the indoor test was used to further verify the ice melting effect of the cast conductive asphalt coagulation with or without the insulation layer. Finally, the thermal-structural coupling analysis of cast conductive asphalt concrete steel bridge deck pavement was carried out using ANSYS finite element software. The results showed that the stainless steel electrode material had the best heating effect, and the electrode thickness in the range of 0.1~3 mm had no effect on the heating effect. The intermediate heating rate of the upper surface of the stainless steel sheet electrode cast conductive asphalt concrete in the left and right external electrodes was 8 ∘C/h, while the intermediate heating rate of the upper surface of the stainless steel mesh electrode cast conductive asphalt concrete was 12.9 ∘C/h. The layout of the left and right buried stainless steel metal mesh was able to effectively improve the snow melting efficiency; ANSYS finite element ice melting simulation was used to obtain the variation law of ice melting efficiency and a temperature field of cast conductive asphalt concrete. The indoor ice melting test showed that when melting the same thickness ice layer at 50 V voltage, it took 240 min with an insulation layer and 720 min without an insulation layer, which was three times that of the ice with an insulation layer, which further verifies the superiority of its ice melting effect. The most unfavorable load position of pavement under load and temperature field was determined. The maximum tensile stress and compressive stress of the pavement surface were transverse, and the maximum shear stress of the pavement bottom was transverse.

13.
Front Immunol ; 13: 947180, 2022.
Article in English | MEDLINE | ID: mdl-35935977

ABSTRACT

African swine fever (ASF) is an acute, hemorrhagic and highly contagious infectious disease caused by African swine fever virus (ASFV), which infects domestic pigs or wild boars. It is characterized by short course of disease, high fever and hemorrhagic lesions, with mortality of up to 100% from acute infection. Up to now, the lack of commercial vaccines and effective drugs has seriously threatened the healthy economic development of the global pig industry. ASFV is a double-stranded DNA virus and genome varies between about 170-194 kb, which encodes 150-200 viral proteins, including 68 structural proteins and more than 100 non-structural proteins. In recent years, although the research on structure and function of ASFV-encoded proteins has been deepened, the structure and infection process of ASFV are still not clear. This review summarizes the main process of ASFV infection, replication and functions of related viral proteins to provide scientific basis and theoretical basis for ASFV research and vaccine development.


Subject(s)
African Swine Fever Virus , African Swine Fever , African Swine Fever Virus/genetics , Animals , Sus scrofa , Swine , Viral Proteins/metabolism
14.
Mol Brain ; 15(1): 73, 2022 08 20.
Article in English | MEDLINE | ID: mdl-35987639

ABSTRACT

Migraine is a complex neurological disease of unknown etiology involving both genetic and environmental factors. It has previously been reported that persistent pain may be mediated by the immune and inflammatory systems. Toll-like receptors (TLRs) play a significant role in immune and inflammatory responses and are expressed by microglia and astrocytes. One of the fundamental mechanisms of the innate immune system in coordinating inflammatory signal transduction is through TLRs, which protect the host organism by initiating inflammatory signaling cascades in response to tissue damage or stress. TLRs reside at the neuroimmune interface, and accumulating evidence has suggested that the inflammatory consequences of TLR activation on glia (mainly microglia and astrocytes), sensory neurons, and other cell types can influence nociceptive processing and lead to pain. Several studies have shown that TLRs may play a key role in neuropathic pain and migraine etiology by activating the microglia. The pathogenesis of migraine may involve a TLR-mediated crosstalk between neurons and immune cells. Innate responses in the central nervous system (CNS) occur during neuroinflammatory phenomena, including migraine. Antigens found in the environment play a crucial role in the inflammatory response, causing a broad range of diseases, including migraines. These can be recognized by several innate immune cells, including macrophages, microglia, and dendritic cells, and can be activated through TLR signaling. Given the prevalence of migraine and the insufficient efficacy and safety of current treatment options, a deeper understanding of TLRs is expected to provide novel therapies for managing chronic migraine. This review aimed to justify the view that TLRs may be involved in migraine.


Subject(s)
Migraine Disorders , Neuralgia , Central Nervous System/metabolism , Humans , Microglia/metabolism , Migraine Disorders/complications , Migraine Disorders/metabolism , Migraine Disorders/pathology , Neuralgia/metabolism , Toll-Like Receptors/metabolism
15.
J Virol ; 96(10): e0030922, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35502911

ABSTRACT

Inflammatory responses play a central role in host defense against invading pathogens. Peste des petits ruminants virus (PPRV) causes highly contagious acute or subacute disease of small ruminants. However, the precise mechanism by which PPRV regulates inflammatory responses remains unknown. Here, we revealed a novel mechanism by which PPRV induces inflammation. Our study showed that PPRV induced the secretion of interleukin 1ß (IL-1ß) by activating the NF-κB signaling pathway and the NLRP3 inflammasome. Moreover, PPRV replication and protein synthesis were essential for NLRP3 inflammasome activation. Importantly, PPRV N protein promoted NF-κB signaling pathway and NLRP3 inflammasome via direct binding of MyD88 and NLPR3, respectively, and induced caspase-1 cleavage and IL-1ß maturation. Biochemically, N protein interacted with MyD88 to potentiate the assembly of MyD88 complex and interacted with NLPR3 to facilitate NLRP3 inflammasome complex assembly by forming an N-NLRP3-ASC ring-like structure, leading to IL-1ß secretion. These findings demonstrate a new function of PPRV N protein as an important proinflammation factor and identify a novel underlying mechanism modulating inflammasome assembly and function induced by PPRV. IMPORTANCE An important part of the innate immune response is the activation of NF-κB signaling pathway and NLPR3 inflammasome, which is induced upon exposure to pathogens. Peste des petits ruminants virus (PPRV) is a highly contagious virus causing fever, stomatitis, and pneumoenteritis in goats by inducing many proinflammatory cytokines. Although the NF-κB signaling pathway and NLRP3 inflammasome play an important role in regulating host immunity and viral infection, the precise mechanism by which PPRV regulates inflammatory responses remains unknown. This study demonstrates that PPRV induces inflammatory responses. Mechanistically, PPRV N protein facilitates the MyD88 complex assembly by directly binding to MyD88 and promotes the NLRP3 inflammasome complex assembly by directly binding to NLRP3 to form ring-like structures of N-NLRP3-ASC. These findings provide insights into the prevention and treatment of PPRV infection.


Subject(s)
Myeloid Differentiation Factor 88 , NLR Family, Pyrin Domain-Containing 3 Protein , Nucleocapsid Proteins , Peste-des-petits-ruminants virus , Animals , Goats , Inflammasomes/metabolism , Inflammation/virology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleocapsid Proteins/metabolism , Peste-des-Petits-Ruminants
16.
Sheng Wu Gong Cheng Xue Bao ; 38(5): 1837-1846, 2022 May 25.
Article in Chinese | MEDLINE | ID: mdl-35611732

ABSTRACT

We researched the mechanism of African swine fever virus (ASFV) protein E248R in regulating the cGAS-STING pathway. First, we verified via the dual-luciferase reporter assay system that E248R protein inhibited the secretion of IFN-ß induced by cGAS-STING or HT-DNA in a dose-dependent manner. The relative quantitative PCR analysis indicated that the overexpression of E248R inhibited HT-DNA-induced transcription of IFN-b1, RANTES, IL-6, and TNF-α in PK-15 cells. Next, we found that E248R interacted with STING by co-immunoprecipitation assay and laser confocal microscopy. Finally, we demonstrated that E248R inhibited the expression of STING protein by using Western blotting. We demonstrated for the first time that the E248R protein of ASFV suppressed the host innate immune response via inhibiting STING expression. The results are pivotal in extending the understanding of the ASFV immune escape and can guide the design of vaccines against ASFV.


Subject(s)
African Swine Fever Virus , African Swine Fever Virus/genetics , Animals , DNA , Immunity, Innate , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Signal Transduction , Swine
17.
Article in English | MEDLINE | ID: mdl-34648929

ABSTRACT

The present study was conducted to characterize the full-length cDNA of c-Jun N-terminal kinase (JNK) in Procambarus clarkii (Pcjnk) and evaluate its potential function under different molt cycle. The full-length cDNA of Pcjnk covered 2937 bp with an open reading frame of 1320 bp, encoding 439 amino acids. A typical conserved TPY motif (118Thr-Pro-120Tyr) was found in Pcjnk. Quantitative real-time PCR (qRT-PCR) analysis revealed a constitutive expression of Pcjnk in the tested tissue, with the highest expression occurring in the hepatopancreas. Additionally, the present study initially revealed that relative mRNA expression of Pcjnk and apoptosis level were significantly higher in the premolt stage (D1/D2 and D3/D4 stage) as compared to other molt stages. In contrast to the levels of superoxide dismutase (SOD) and malondialdehyde (MDA), catalase (CAT) and glutathione peroxidase (GPX) level decreased significantly from the intermolt stage (C stage) to the premolt stage (D1/D2 and D3/D4 stage), then increased from the premolt stage to the postmolt stage (A and B stage). The results obtained in the present study indicated that molt could cause apoptosis induced by oxidative stress through the activation of JNK in Procambarus clarkii.


Subject(s)
Astacoidea , Molting , Animals , Apoptosis , JNK Mitogen-Activated Protein Kinases , Oxidative Stress
18.
J Virol ; 96(1): e0150021, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34613824

ABSTRACT

African swine fever (ASF), a devastating infectious disease in swine, severely threatens the global pig farming industry. Disease control has been hampered by the unavailability of vaccines. Here, we report that deletion of the QP509L and QP383R genes (ASFV-ΔQP509L/QP383R) from the highly virulent ASF virus (ASFV) CN/GS/2018 strain results in complete viral attenuation in swine. Animals inoculated with ASFV-ΔQP509L/QP383R at a 104 50% hemadsorbing dose (HAD50) remained clinically normal during the 17-day observational period. All ASFV-ΔQP509L/QP383R-infected animals had low viremia titers and developed a low-level p30-specific antibody response. However, ASFV-ΔQP509L/QP383R did not induce protection against challenge with the virulent parental ASFV CN/GS/2018 isolate. RNA-sequencing analysis revealed that innate immune-related genes (Ifnb, Traf2, Cxcl10, Isg15, Rantes, and Mx1) were significantly lower in ASFV-ΔQP509L/QP383R-infected than in ASFV-infected porcine alveolar macrophages. In addition, ASFV-ΔQP509L/QP383R-infected pigs had low levels of interferon-ß (IFN-ß) based on enzyme-linked immunosorbent assay (ELISA). These data suggest that deletion of ASFV QP509L/383R reduces virulence but does not induce protection against lethal ASFV challenge. IMPORTANCE African swine fever (ASF) is endemic to several parts of the word, with outbreaks of the disease devastating the swine farming industry; currently, no commercially available vaccine exists. Here, we report that deletion of the previously uncharacterized QP509L and QP383R viral genes completely attenuates virulence in the ASF virus (ASFV) CN/GS/2018 isolate. However, ASFV-ΔQP509L/QP383R-infected animals were not protected from developing an ASF infection after challenge with the virulent parental virus. ASFV-ΔQP509L/QP383R induced lower levels of innate immune-related genes and IFN-ß than the parental virus. Our results increase our knowledge of developing an effective and live ASF attenuated vaccine.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever/virology , Host-Pathogen Interactions , Sequence Deletion , Viral Proteins/genetics , African Swine Fever/immunology , African Swine Fever Virus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cells, Cultured , Disease Resistance , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunization , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mutagenesis , Swine , Transcriptome , Virulence/genetics , Virulence Factors/genetics , Virus Replication
19.
J Environ Manage ; 299: 113650, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34481370

ABSTRACT

The partial substitution of chemical fertilizers with organic manure has positive effects on crop productivity and sustainable development. Nevertheless, few studies have focused on major grain crops. Herein, we report the short-term effects of the partial substitution of chemical fertilizers with organic manure on the physicochemical properties, microbial community, and enzyme activities in the rhizosphere soil of a maize (Zea mays L.) field. A decrease in soil bulk density, pH, and electrical conductivity, concomitant with an increase in soil urease, invertase, and alkaline phosphatase activities, and a high level of nutrients were observed in organic manure-treated soil. The influence of the organic substitution treatment on bacterial diversity was greater than that on fungal diversity, particularly on alpha diversity. Among dominant bacterial phyla, Actinobacteria abundance changed the most, with significantly increase under organic manure treatment. In turn, among fungi, only Ascomycota responded substantially to organic substitution. Binding spatial ordination analysis revealed that relative soil water content and soil organic carbon, and nitrate and total nitrogen contents had a stronger effect on bacteria and fungi, respectively, than any other soil physicochemical property. Additionally, the changes in bacterial and fungal communities influenced soil enzymatic activities. Moreover, partial least squares path model revealed that soil physicochemical properties indirectly affected soil enzymatic activities by their direct effects on microbial (both bacteria and fungi) community. Overall, our results indicate that the substitution of chemical fertilizers by organic manure changed the composition of the soil microbial community, and that the effects of the substitution were more significant on bacteria than on fungi.


Subject(s)
Fertilizers , Microbiota , Carbon , Fertilizers/analysis , Manure , Nitrogen , Rhizosphere , Soil , Soil Microbiology
20.
Front Immunol ; 12: 718548, 2021.
Article in English | MEDLINE | ID: mdl-34526993

ABSTRACT

Ras-GTPase-activating protein (SH3 domain)-binding protein (G3BP) is an RNA binding protein. G3BP is a key component of stress granules (SGs) and can interact with many host proteins to regulate the expression of SGs. As an antiviral factor, G3BP can interact with viral proteins to regulate the assembly of SGs and thus exert antiviral effects. However, many viruses can also use G3BP as a proximal factor and recruit translation initiation factors to promote viral proliferation. G3BP regulates mRNA translation and attenuation to regulate gene expression; therefore, it is closely related to diseases, such as cancer, embryonic death, arteriosclerosis, and neurodevelopmental disorders. This review discusses the important discoveries and developments related G3BP in the biological field over the past 20 years, which includes the formation of SGs, interaction with viruses, stability of RNA, and disease progression.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/chemistry , RNA Helicases/metabolism , RNA Recognition Motif Proteins/chemistry , RNA Recognition Motif Proteins/metabolism , Animals , Cytoplasmic Granules/metabolism , DNA Helicases/genetics , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA Viruses/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Research , Stress, Physiological , Structure-Activity Relationship , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...