Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Virol ; 98(1): e0162523, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38084960

RESUMO

Porcine epidemic diarrhea virus (PEDV) is an enteric coronavirus that causes high mortality in piglets, thus posing a serious threat to the world pig industry. Porcine epidemic diarrhea (PED) is related to the imbalance of sodium absorption by small intestinal epithelial cells; however, the etiology of sodium imbalanced diarrhea caused by PEDV remains unclear. Herein, we first proved that PEDV can cause a significant decrease in Na+/H+ exchanger 3 (NHE3) expression on the cell membrane, in a viral dose-dependent manner. Further study showed that the PEDV nucleocapsid (N) protein participates in the regulation of NHE3 activity through interacting with Ezrin. Flame atomic absorption spectroscopy results indicated a serious imbalance in Na+ concentration inside and outside cells following overexpression of PEDV N. Meanwhile, molecular docking technology identified that the small molecule drug Pemetrexed acts on the PEDV N-Ezrin interaction region. It was confirmed that Pemetrexed can alleviate the imbalanced Na+ concentration in IPEC-J2 cells and the diarrhea symptoms of Rongchang pigs caused by PEDV infection. Overall, our data suggest that the interaction between PEDV N and Ezrin reduces the level of phosphorylated Ezrin, resulting in a decrease in the amount of NHE3 protein on the cell membrane. This leads to an imbalance of intracellular and extracellular Na+, which causes diarrhea symptoms in piglets. Pemetrexed is effective in relieving diarrhea caused by PEDV. Our results provide a reference to screen for anti-PEDV targets and to develop drugs to prevent PED.IMPORTANCEPorcine epidemic diarrhea (PED) has caused significant economic losses to the pig industry since its initial outbreak, and the pathogenic mechanism of porcine epidemic diarrhea virus (PEDV) is still under investigation. Herein, we found that the PEDV nucleocapsid protein interacts with Ezrin to regulate Na+/H+ exchanger 3 activity. In addition, we screened out Pemetrexed, a small molecule drug, which can effectively alleviate pig diarrhea caused by PEDV. These results provide support for further exploration of the pathogenesis of PEDV and the development of drugs to prevent PED.


Assuntos
Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Animais , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/veterinária , Diarreia/tratamento farmacológico , Diarreia/veterinária , Simulação de Acoplamento Molecular , Proteínas do Nucleocapsídeo/metabolismo , Pemetrexede/metabolismo , Vírus da Diarreia Epidêmica Suína/fisiologia , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio/metabolismo , Suínos , Doenças dos Suínos/tratamento farmacológico
2.
Front Microbiol ; 14: 1237913, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38029193

RESUMO

As part of the genus Enteropathogenic Coronaviruses, Porcine Epidemic Diarrhea Virus (PEDV) is an important cause of early diarrhea and death in piglets, and one of the most difficult swine diseases to prevent and control in the pig industry. Previously, we found that PEDV can block Na+ absorption and induce diarrhea in piglets by inhibiting the activity of the sodium-hydrogen ion transporter NHE3 in pig intestinal epithelial cells, but the mechanism needs to be further explored. The epidermal growth factor receptor (EGFR) has been proved to be one of the co-receptors involved in many viral infections and a key protein involved in the regulation of NHE3 activity in response to various pathological stimuli. Based on this, our study used porcine intestinal epithelial cells (IPEC-J2) as an infection model to investigate the role of EGFR in regulating NHE3 activity after PEDV infection. The results showed that EGFR mediated viral invasion by interacting with PEDV S1, and activated EGFR regulated the downstream EGFR/ERK signaling pathway, resulting in decreased expression of NHE3 and reduced NHE3 mobility at the plasma membrane, which ultimately led to decreased NHE3 activity. The low level of NHE3 expression in intestinal epithelial cells may be a key factor leading to PEDV-induced diarrhea in newborn piglets. This study reveals the importance of EGFR in the regulation of NHE3 activity by PEDV and provides new targets and clues for the prevention and treatment of PEDV-induced diarrhea in piglets.

3.
Front Microbiol ; 14: 1251275, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37840713

RESUMO

Porcine epidemic diarrhea virus (PEDV) mainly invades the small intestine and promotes an inflammatory response, eventually leading to severe diarrhea, vomiting, dehydration, and even death of piglets, which seriously threatens the economic development of pig farming. In recent years, researchers have found that probiotics can improve the intestinal microenvironment and reduce diarrhea. At the same time, certain probiotics have been shown to have antiviral effects; however, their mechanisms are different. Herein, we aimed to investigate the inhibitory effect of Lactiplantibacillus plantarum supernatant (LP-1S) on PEDV and its mechanism. We used IPEC-J2 cells as a model to assess the inhibitory effect of LP-1S on PEDV and to further investigate the relationship between LP-1S, Ca2+, and PEDV. The results showed that a divalent cation chelating agent (EGTA) and calcium channel inhibitors (Bepridil hydrochloride and BAPTA-acetoxymethylate) could inhibit PEDV proliferation while effectively reducing the intracellular Ca2+ concentration. Furthermore, LP-1S could reduce PEDV-induced loss of calcium channel proteins (TRPV6 and PMCA1b), alleviate intracellular Ca2+ accumulation caused by PEDV infection, and promote the balance of intra- and extracellular Ca2+ concentrations, thereby inhibiting PEDV proliferation. In summary, we found that LP-1S has potential therapeutic value against PEDV, which is realized by modulating Ca2+. This provides a potential new drug to treat PEDV infection.

4.
Front Cell Infect Microbiol ; 13: 1129172, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37143745

RESUMO

Globally, brucellosis is a widespread zoonotic disease. It is prevalent in more than 170 countries and regions. It mostly damages an animal's reproductive system and causes extreme economic losses to the animal husbandry industry. Once inside cells, Brucella resides in a vacuole, designated the BCV, which interacts with components of the endocytic and secretory pathways to ensure bacterial survival. Numerous studies conducted recently have revealed that Brucella's ability to cause a chronic infection depends on how it interacts with the host. This paper describes the immune system, apoptosis, and metabolic control of host cells as part of the mechanism of Brucella survival in host cells. Brucella contributes to both the body's non-specific and specific immunity during chronic infection, and it can aid in its survival by causing the body's immune system to become suppressed. In addition, Brucella regulates apoptosis to avoid being detected by the host immune system. The BvrR/BvrS, VjbR, BlxR, and BPE123 proteins enable Brucella to fine-tune its metabolism while also ensuring its survival and replication and improving its ability to adapt to the intracellular environment.


Assuntos
Brucella , Brucelose , Animais , Infecção Persistente , Macrófagos/microbiologia , Vacúolos/microbiologia
5.
Viruses ; 15(4)2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37112936

RESUMO

Various coronaviruses have emerged as a result of cross-species transmission among humans and domestic animals. Porcine epidemic diarrhea virus (PEDV; family Coronaviridae, genus Alphacoronavirus) causes acute diarrhea, vomiting, dehydration, and high mortality in neonatal piglets. Porcine small intestinal epithelial cells (IPEC-J2 cells) can be used as target cells for PEDV infection. However, the origin of PEDV in pigs, the host range, and cross-species infection of PEDV remain unclear. To determine whether PEDV has the ability to infect human cells in vitro, human small intestinal epithelial cells (FHs 74 Int cells) were inoculated with PEDV LJX and PEDV CV777 strains. The results indicated that PEDV LJX, but not PEDV CV777, could infect FHs 74 Int cells. Furthermore, we observed M gene mRNA transcripts and N protein expression in infected FHs 74 Int cells. A one-step growth curve showed that the highest viral titer of PEDV occurred at 12 h post infection. Viral particles in vacuoles were observed in FHs 74 Int cells at 24 h post infection. The results proved that human small intestinal epithelial cells are susceptible to PEDV infection, suggesting the possibility of cross-species transmission of PEDV.


Assuntos
Infecções por Coronavirus , Vírus da Diarreia Epidêmica Suína , Doenças dos Suínos , Humanos , Animais , Suínos , Linhagem Celular , Vírus da Diarreia Epidêmica Suína/genética , Intestinos , Células Epiteliais , Infecções por Coronavirus/veterinária , Diarreia
6.
Mol Phylogenet Evol ; 181: 107704, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36657625

RESUMO

The present study aimed to apply bioinformatic methods to analyze the structure of the S protein of human respiratory coronaviruses, including severe respiratory disease syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), human coronavirus HKU1 (HCoV-HKU1), and severe respiratory disease syndrome coronavirus type 2 (SARS-CoV-2). We predicted and analyzed the physicochemical properties, hydrophilicity and hydrophobicity, transmembrane regions, signal peptides, phosphorylation and glycosylation sites, epitopes, functional domains, and motifs of the S proteins of human respiratory coronaviruses. All four S proteins contain a transmembrane region, which enables them to bind to host cell surface receptors. All four S proteins contain a signal peptide, phosphorylation sites, glycosylation sites, and epitopes. The predicted phosphorylation sites might mediate S protein activation, the glycosylation sites might affect the cellular orientation of the virus, and the predicted epitopes might have implications for the design of antiviral inhibitors. The S proteins of all four viruses have two structural domains, S1 (C-terminal and N-terminal domains) and S2 (homology region 1 and 2). Our bioinformatic analysis of the structural and functional domains of human respiratory coronavirus S proteins provides a basis for future research to develop broad-spectrum antiviral drugs, vaccines, and antibodies.


Assuntos
COVID-19 , Coronavírus da Síndrome Respiratória do Oriente Médio , Humanos , SARS-CoV-2 , Filogenia , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Biologia Computacional
7.
J Virol ; 96(22): e0147322, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36342433

RESUMO

Transmissible gastroenteritis virus (TGEV) is member of the family Coronaviridae and mainly causes acute diarrhea. TGEV infection is characterized by vomiting, watery diarrhea, and severe dehydration, resulting in high mortality rates in neonatal piglets. TGEV infection symptoms are related to an imbalance of sodium absorption in small intestinal epithelial cells; however, the etiology of sodium imbalance diarrhea caused by TGEV remains unclear. In this study, we performed transcriptomic analysis of intestinal tissues from infected and healthy piglets and observed that the expression of NHE3, encoding Na+/H+ exchanger 3 (NHE3), the main exchanger of electroneutral sodium in intestinal epithelial cells, was significantly reduced upon TGEV infection. We also showed that specific inhibition of intestinal NHE3 activity could lead to the development of diarrhea in piglets. Furthermore, we revealed an interaction between TGEV N protein and NHE3 near the nucleus. The binding of TGEV N to NHE3 directly affected the expression and activity of NHE3 on the cell surface and affected cellular electrolyte absorption, leading to diarrhea. Molecular docking and computer-aided screening techniques were used to screen for the blocker of the interaction between TGEV N and NHE3, which identified irinotecan. We then demonstrated that irinotecan was effective in relieving TGEV-induced diarrhea in piglets. These findings provide new insights into the mechanism of TGEV-induced sodium imbalance diarrhea and could lead to the design of novel antiviral strategies against TGEV. IMPORTANCE A variety of coronaviruses have been found to cause severe diarrhea in hosts, including TGEV; however, the pathogenic mechanism is not clear. Therefore, prompt determination of the mechanism and identification of efficient therapeutic agents are required, both for public health reasons and for economic development. In this study, we demonstrated that NHE3 is the major expressed protein of NHEs in the intestine, and its expression decreased by nearly 70% after TGEV infection. Also, specific inhibition of intestinal NHE3 resulted in severe diarrhea in piglets. This demonstrated that NHE3 plays an important role in TGEV-induced diarrhea. In addition, we found that TGEV N directly regulates NHE3 expression and activity through protein-protein interaction, which is essential to promote diarrhea. Molecular docking and other techniques demonstrated that irinotecan could block the interaction and diarrhea caused by TGEV. Thus, our results provide a basis for the development of novel therapeutic agents against TGEV and guidance for the development of drugs for other diarrhea-causing coronaviruses.


Assuntos
Infecções por Coronavirus , Coronavirus , Vírus da Gastroenterite Transmissível , Animais , Suínos , Vírus da Gastroenterite Transmissível/fisiologia , Trocador 3 de Sódio-Hidrogênio/genética , Trocador 3 de Sódio-Hidrogênio/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Irinotecano , Simulação de Acoplamento Molecular , Diarreia/veterinária , Trocadores de Sódio-Hidrogênio/metabolismo , Coronavirus/metabolismo , Sódio/metabolismo
8.
Vet Res ; 53(1): 80, 2022 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-36209206

RESUMO

Pseudomonas aeruginosa (P. aeruginosa) is a known bacterium that produces biofilms and causes severe infection. Furthermore, P. aeruginosa biofilms are extremely difficult to eradicate, leading to the development of chronic and antibiotic-resistant infections. Our previous study showed that a cathelicidin-related antimicrobial peptide (CRAMP) inhibits the formation of P. aeruginosa biofilms and markedly reduces the biomass of preformed biofilms, while the mechanism of eradicating bacterial biofilms remains elusive. Therefore, in this study, the potential mechanism by which CRAMP eradicates P. aeruginosa biofilms was investigated through an integrative analysis of transcriptomic, proteomic, and metabolomic data. The omics data revealed CRAMP functioned against P. aeruginosa biofilms by different pathways, including the Pseudomonas quinolone signal (PQS) system, cyclic dimeric guanosine monophosphate (c-di-GMP) signalling pathway, and synthesis pathways of exopolysaccharides and rhamnolipid. Moreover, a total of 2914 differential transcripts, 785 differential proteins, and 280 differential metabolites were identified. A series of phenotypic validation tests demonstrated that CRAMP reduced the c-di-GMP level with a decrease in exopolysaccharides, especially alginate, in P. aeruginosa PAO1 biofilm cells, improved bacterial flagellar motility, and increased the rhamnolipid content, contributing to the dispersion of biofilms. Our study provides new insight into the development of CRAMP as a potentially effective antibiofilm dispersant.


Assuntos
Peptídeos Antimicrobianos , Pseudomonas aeruginosa , Alginatos/metabolismo , Animais , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos , Proteínas de Bactérias/genética , Biofilmes , GMP Cíclico , Regulação Bacteriana da Expressão Gênica , Guanosina Monofosfato/metabolismo , Camundongos , Proteômica , Pseudomonas aeruginosa/metabolismo , Catelicidinas
9.
PLoS Negl Trop Dis ; 16(9): e0010734, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36054186

RESUMO

Toxocariasis is a neglected parasitic disease caused predominantly by larvae of Toxocara canis. While this zoonotic disease is of major importance in humans and canids, it can also affect a range of other mammalian hosts. It is known that mucins secreted by larvae play key roles in immune recognition and evasion, but very little is understood about the molecular interactions between host cells and T. canis. Here, using an integrative approach (affinity pull-down, mass spectrometry, co-immunoprecipitation and bioinformatics), we identified 219 proteins expressed by a murine macrophage cell line (RAW264.7) that interact with prokaryotically-expressed recombinant protein (rTc-MUC-1) representing the mucin Tc-MUC-1 present in the surface coat of infective larvae of T. canis. Protein-protein interactions between rTc-MUC-1 and an actin binding protein CFL1 as well as the fatty acid binding protein FABP5 of RAW264.7 macrophages were also demonstrated in a human embryonic kidney cell line (HEK 293T). By combing predicted structural information on the protein-protein interaction and functional knowledge of the related protein association networks, we inferred roles for Tc-MUC-1 protein in the regulation of actin cytoskeletal remodelling, and the migration and phagosome formation of macrophage cells. These molecular interactions now require verification in vivo. The experimental approach taken here should be readily applicable to comparative studies of other ascaridoid nematodes (e.g. T. cati, Anisakis simplex, Ascaris suum and Baylisascaris procyonis) whose larvae undergo tissue migration in accidental hosts, including humans.


Assuntos
Toxocara canis , Toxocaríase , Actinas , Animais , Proteínas de Ligação a Ácido Graxo , Larva , Macrófagos , Mamíferos , Camundongos , Mucinas , Proteínas de Neoplasias , Proteínas Recombinantes , Toxocaríase/parasitologia
10.
Sheng Wu Gong Cheng Xue Bao ; 38(8): 2821-2839, 2022 Aug 25.
Artigo em Chinês | MEDLINE | ID: mdl-36002413

RESUMO

Probiotics can improve the microecological balance of the body and have special effects in promoting nutrient absorption, controlling intestinal infections, and regulating immune function. However, there are problems such as difficult colonization in the gastrointestinal environment and low oral bioavailability. Bacterial biofilms are organized bacterial cells that adhere to an abiotic or biotic surface and are enclosed in extracellular polymeric substances of exopolysaccharides (EPS), extracellular DNA (eDNA), proteins and lipids, with a three-dimensional spatial structure. Probiotics with the help of bacterial biofilms have obvious advantages over planktonic bacteria in stress resistance, combating pathogens and modulating the host's immune function, which provides a new research idea for the development of probiotics. This paper expounded on the advantages of probiotics with the help of bacterial biofilms, and focused on introducing substances that could promote the formation of probiotic biofilms and the mechanisms, and the safety of probiotic biofilms. Currently, research on probiotic biofilms is still in its infancy, and this paper is expected to provide references for future research in this field.


Assuntos
Biofilmes , Probióticos , Bactérias , Matriz Extracelular de Substâncias Poliméricas
11.
Vet Microbiol ; 263: 109263, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34749283

RESUMO

Transmissible gastroenteritis (TGE) and porcine epidemic diarrhea (PED) are highly transmissible intestinal infections caused by transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PEDV), respectively. They are clinically associated with vomiting, diarrhea, and dehydration in piglets. An imbalance in Na+ uptake by intestinal epithelial cells causes TGEV/PEDV-induced diarrhea. However, the mechanism by which TGEV/PEDV-infection in piglets causes Na+ imbalance diarrhea has not been elucidated. In the present study, we demonstrated that specific inhibition of NHE3 activity caused small intestinal bulging, intestinal wall thinning and severe diarrhea in piglets, consistent with the signs of TGEV/PEDV infection. This study further elucidated the role of NHE3 in TGEV/PEDV-induced diarrhea. In this study, small intestinal epithelial cells (IPEC-J2) were used as a model of infection. The results showed that TGEV/PEDV infection reduced NHE3 activity and Na+ uptake in IPEC-J2 cells. Further studies revealed that the use of NHE3-specific inhibitors could reduce the amount of cell membrane NHE3, thereby decreasing Na+ uptake and ultimately leading to diarrhea. Transcriptomic studies performed on obtained jejunal tissues were also consistent with pre-laboratory results. This study will provide a basis for understanding Na+ imbalance diarrhea caused by TGEV/PEDV, as well as for elucidating the diarrheal pathogenesis of other members of α-animal coronaviruses.


Assuntos
Infecções por Coronavirus , Diarreia , Gastroenterite Suína Transmissível , Trocador 3 de Sódio-Hidrogênio , Doenças dos Suínos , Animais , Infecções por Coronavirus/fisiopatologia , Infecções por Coronavirus/veterinária , Diarreia/fisiopatologia , Diarreia/veterinária , Células Epiteliais/patologia , Células Epiteliais/virologia , Gastroenterite Suína Transmissível/fisiopatologia , Vírus da Diarreia Epidêmica Suína , Trocador 3 de Sódio-Hidrogênio/metabolismo , Suínos , Vírus da Gastroenterite Transmissível
12.
Cell Cycle ; 20(24): 2565-2582, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34822321

RESUMO

As the main exchanger of electroneutral NaCl absorption, sodium-hydrogen exchanger isoform 3 (NHE3) circulates in the epithelial brush border (BB) and intracellular compartments in a multi-protein complex. The size of the NHE3 complex changes during rapid regulation events. Recycling regulation of NHE3 in epithelial cells can be roughly divided into three stages. First, when stimulated by Ca2+, cGMP, and cAMP-dependent signaling pathways, NHE3 is converted from an immobile complex found at the apical microvilli (MV) into an easily internalized and mobile form that relocates to a compartment near the base of the MV. Second, NHE3 is internalized by clathrin and albumin-dependent pathways into cytoplasmic endosomal compartments, where the complex is reprocessed and reassembled. Finally, NHE3 is translocated from the recycling endosomes (REs) to the apex of epithelial cells, a process that can be stimulated by an increase in sodium-glucose cotransporter 1 (SGLT1) activity, epidermal growth factor receptor (EGFR) signaling, Ca2+ signaling, and binding to ßPix and SH3 and multiple ankyrin repeat domains 2 (Shank2) proteins. This review describes the molecular steps and protein interactions involved in the recycling movement of NHE3 from the apex of epithelial cells, into vesicles, where it is reprocessed and reassembled, and returned to its original location on the plasma membrane, where it exerts its physiological function.


Assuntos
Células Epiteliais , Trocadores de Sódio-Hidrogênio , Animais , Células Epiteliais/metabolismo , Camundongos , Microvilosidades/metabolismo , Isoformas de Proteínas/metabolismo , Trocador 3 de Sódio-Hidrogênio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo
13.
Virology ; 563: 64-73, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34464882

RESUMO

Porcine epidemic diarrhea virus (PEDV; family Coronaviridae, genus Alphacoronavirus) causes acute diarrhea and vomiting, dehydration, and high mortality in neonatal piglets. Despite extensive research focusing on the pathogenesis of PEDV infection, the molecular pathogenesis of PEDV-induced diarrhea in piglets remains unclear. Na+/H+ exchanger 3 (NHE3), the main exchanger of electroneutral sodium in intestinal epithelial cells, is closely associated with the occurrence of diarrhea. To date, there is no study on whether diarrhea caused by PEDV infection is related to the activity of NHE3. In the present study, it was found that the expression level of cell membrane protein NHE3 significantly decreased after PEDV infection, whereas the total level of protein expression was not significantly changed. The Na+/H+ transport rate and the mRNA abundance of NHE3 decreased; the NHE3 activity decreased gradually with increasing infection time. In vivo, after PEDV infection of newborn piglets, rupture of intestinal villi and interstitial degeneration of intestinal epithelial cells in different intestinal segments were observed by hematoxylin-eosin staining. Immunohistochemical and immunofluorescence methods were used to observe the decreased expression of NHE3 protein on the membrane of intestinal epithelial cells in the jejunum and ileum. Taken together, these data indicate that PEDV infection reduces NHE3 activity in intestinal epithelial cells, hindering Na+ transport and thus causing diarrhea.


Assuntos
Infecções por Coronavirus/veterinária , Diarreia/veterinária , Vírus da Diarreia Epidêmica Suína , Trocador 3 de Sódio-Hidrogênio/metabolismo , Doenças dos Suínos/virologia , Animais , Animais Recém-Nascidos , Anticorpos , Chlorocebus aethiops , Infecções por Coronavirus/virologia , Diarreia/virologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica/imunologia , Intestinos/metabolismo , Camundongos , Trocador 3 de Sódio-Hidrogênio/genética , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/metabolismo , Fator de Transcrição TFIIH/genética , Fator de Transcrição TFIIH/metabolismo , Células Vero
14.
Res Vet Sci ; 139: 32-42, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34246941

RESUMO

Porcine epidemic diarrhea virus (PEDV) is an enteropathogenic coronavirus; it causes diarrhea in pigs and is associated with high morbidity and mortality in sucking piglets. In this study, we performed in vitro and in vivo experiments to determine the inhibitory effects of Lactobacillus plantarum metabolites (LPM) on PEDV replication. Gas chromatography-mass spectrometry revealed exopolysaccharides to be one of the main components of LPM. We then determine whether L. plantarum exopolysaccharides (LPE) have an antiviral effect and also detected the expression levels of the apoptosis-related genes Bax and Bcl-2 and of the pro-apoptotic protein caspase-3. Further, we assessed the transcription levels of an immune-related protein (STAT1) and antiviral factors (MX1, MX2, ISG15, ZAP, PKR, and OAS1). Our results showed that the most effective method was to pretreat cells with LPM and that the optimal dose of LPM that could be safely administered to Vero cells was 1/8 times of the stock solution. LPE had a strong inhibitory effect on PEDV; the most effective method of administration was to co-incubate cells with LPE and PEDV, and the optimal concentration of LPE was 1.35 mg/mL. To conclude, LPE prevented PEDV adsorption and also alleviated inflammatory responses and induced early apoptosis of injured cells, but it could not regulate the immune function of cells.


Assuntos
Lactobacillus plantarum/metabolismo , Polissacarídeos/farmacologia , Polissacarídeos/uso terapêutico , Vírus da Diarreia Epidêmica Suína/efeitos dos fármacos , Vírus da Diarreia Epidêmica Suína/crescimento & desenvolvimento , Doenças dos Suínos/tratamento farmacológico , Doenças dos Suínos/virologia , Replicação Viral/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Chlorocebus aethiops , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Diarreia/tratamento farmacológico , Diarreia/veterinária , Diarreia/virologia , Inflamação/tratamento farmacológico , Vírus da Diarreia Epidêmica Suína/imunologia , Suínos , Doenças dos Suínos/imunologia , Células Vero , Ligação Viral/efeitos dos fármacos
15.
Front Vet Sci ; 8: 625609, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996960

RESUMO

Swine hepatitis E (swine HE) is a new type of zoonotic infectious disease caused by the swine hepatitis E virus (swine HEV). Open reading frame 3 (ORF3) is an important virulent protein of swine HEV, but its function still is mainly unclear. In this study, we generated adenoviruses ADV4-ORF3 and ADV4 negative control (ADV4-NC), which successfully mediated overexpression of enhanced green fluorescent protein (EGFP)-ORF3 and EGFP, respectively, in HepG2 cells. High-throughput sequencing was used to screen for differentially expressed long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs). The cis-target genes of lncRNAs were predicted, functional enrichment (Gene Ontology [GO] and Kyoto Encyclopedia of Genes and Genomes [KEGG]) was performed, and 12 lncRNAs with statistically significant different expressions (p ≤ 0.05 and q ≤ 1) were selected for further quantitative real-time reverse transcription (qRT-PCR) validation. In HepG2 cells, we identified 62 significantly differentially expressed genes (DEGs) (6,564 transcripts) and 319 lncRNAs (124 known lncRNAs and 195 novel lncRNAs) that were affected by ORF3, which were involved in systemic lupus erythematosus, Staphylococcus aureus infection, signaling pathways pluripotency regulation of stem cells, the peroxisome proliferator-activated receptor (PPAR) signaling pathway, and platinum drug resistance pathways. Cis-target gene prediction identified 45 lncRNAs corresponding to candidate mRNAs, among which eight were validated by qRT-PCR: LINC02476 (two transcripts), RAP2C-AS1, AC016526, AL139099, and ZNF337-AS1 (3 transcripts). Our results revealed that the lncRNA profile in host cells affected by ORF3, swine HEV ORF3, might affect the pentose and glucuronate interconversions and mediate the formation of obstructive jaundice by influencing bile secretion, which will help to determine the function of ORF3 and the infection mechanism and treatment of swine HE.

16.
Mol Omics ; 17(2): 277-287, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33462573

RESUMO

Breast cancer (BC) ranks first among malignancies in the female population due to its complicated pathological progression and poor prognosis. Hence, the aim of the present study was to identify potential molecular prognostic biomarkers able to predict the prognosis of BC patients. We integrated two microRNA (miRNA) expression microarrays and three gene microarrays related to BC from the NCBI Gene Expression Comprehensive (GEO) database to screen for differentially expressed miRNAs and identify their regulatory networks. The Kaplan-Meier plotter online analysis tool was used to assess the overall survival value of miRNAs expression in BC patients. The LinkedOmics online tool was used to analyze genes correlated with miRNAs expression. To clarify the upstream regulation mechanism of genes, we used ChIP-Atlas to identify and screen for transcription factors and visually verify them using the Integrative Genomics Viewer. To further analyze the downstream regulatory mechanism of miRNA in BC, we verified differentially expressed genes (DEGs) correlated to miRNAs in three GEO gene microarrays and the gene set predicted by miRWalk. The open access Metascape program allowed analysis of Gene Ontology (GO) processes, KEGG pathways and GO enrichment was performed on the DEGs. To further identify hub genes, Cytoscape software and its plug-in were applied to construct protein-protein interaction networks. In the present study, several possible molecules and related pathways related to miR-483 were identified by bioinformatics analysis. These molecules and pathways might represent key mechanisms involved in BC progression and development. This work provides a novel view and insight in the pathogenesis, treatment and prognosis for BC.


Assuntos
Neoplasias da Mama/genética , Redes Reguladoras de Genes/genética , MicroRNAs/genética , Proteínas de Neoplasias/genética , Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Biologia Computacional , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Estimativa de Kaplan-Meier , MicroRNAs/classificação , Análise em Microsséries
17.
Virus Res ; 280: 197901, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32070687

RESUMO

Transmissible gastroenteritis virus (TGEV) primarily replicates in intestinal epithelial cells and causes severe damage to host cells, resulting in diarrhea. Surface NHE3 serves as the key regulatory site controlling electroneutral Na+ absorption. In this study, our results showed that the surface NHE3 content was significantly reduced following TGEV infection, whereas the total level of protein expression was not significantly changed, and NHE3 activity gradually decreased with prolonged infection time. We then inhibited SGLT1 expression by lentiviral interference and drug inhibition, respectively. Inhibition studies showed that the level of phosphorylation of the downstream key proteins, MAPKAPK-2 and EZRIN, in the SGLT1-mediated p38MAPK/AKt2 signaling pathway was significantly increased. The surface NHE3 expression was also significantly increased, and NHE3 activity was also significantly enhanced. These results demonstrate that a TGEV infection can inhibit NHE3 translocation and attenuates sodium-hydrogen exchange activity via the SGLT1-mediated p38MAPK/AKt2 signaling pathway, affecting cellular electrolyte absorption leading to diarrhea.


Assuntos
Enterócitos/virologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Transportador 1 de Glucose-Sódio/genética , Trocador 3 de Sódio-Hidrogênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Proteínas Proto-Oncogênicas c-akt/genética , Transportador 1 de Glucose-Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio/genética , Suínos , Vírus da Gastroenterite Transmissível , Proteínas Quinases p38 Ativadas por Mitógeno/genética
18.
Front Microbiol ; 10: 2540, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781061

RESUMO

Transmissible gastroenteritis (TGE), caused by transmissible gastroenteritis virus (TGEV), is one many gastrointestinal inflections in piglets, characterized by diarrhea, and high mortality. Probiotics are ubiquitous bacteria in animal intestines, which have many functions, such as promoting intestinal peristalsis and maintaining the intestinal balance. We found that the supernatant of the Lp-1 strain of Lactobacillus plantarum, isolated in our laboratory, and named Lp-1s had marked anti-TGEV effect on IPEC-J2 cells. Lp-1s could induce large amounts of interferon-ß in IPEC-J2 cells in the early stage (6 h) of infection with TGEV, and increased the level of phosphorylated signal transducer and activator of transcription and its nuclear translocation in the late stage (24-48 h) of infection. This resulted in upregulated expression of interferon-stimulated genes, and increased the transcription and protein expression of antiviral proteins, resulting in an anti-TGEV effect.

19.
Vet Res ; 50(1): 28, 2019 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-31029162

RESUMO

Transmissible gastroenteritis coronavirus (TGEV) is an enteropathogenic coronavirus that causes diarrhea in pigs and is associated with high morbidity and mortality in sucking piglets. S1 is one of two protein domains in the spike (S) glycoprotein and is responsible for enteric tropism, sialic acid recognition, and host receptor binding. Although there has been extensive research on the S1 protein of TGEV, little is known about the intracellular role of TGEV-S1. In the present study, we used yeast two-hybrid screening of a cDNA library from porcine intestinal cells to identify proteins that interact with TGEV-S1. Among 120 positive clones from the library, 12 intracellular proteins were identified after sequencing and a BLAST search. These intracellular proteins are involved in protein synthesis and degradation, biological signal transduction, and negative control of signaling pathways. Using a glutathione-S-transferase (GST) pulldown assay and Co-IP, we found that UBXN1 interacts with the S1 protein. Here, we observed that TGEV infection led to increased UBXN1 expression levels during the late phase of infection in IPEC-J2 cells. Inhibition of UBXN1 in IPEC-J2 cells via siRNA interference significantly decreased the viral titer and downregulated the expression of S1. UBXN1 overexpression significantly increased the viral copy number. Additionally, we provided data suggesting that UBXN1 negatively regulates IFN-ß expression after TGEV infection. Finally, our research indicated that UBXN1 plays a vital role in the process of TGEV infection, making it a candidate target for the development of a novel antiviral method.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Vírus da Gastroenterite Transmissível/fisiologia , Proteínas Virais/fisiologia , Replicação Viral , Western Blotting , Ensaio de Imunoadsorção Enzimática , Imunoprecipitação , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Técnicas do Sistema de Duplo-Híbrido
20.
Res Vet Sci ; 123: 39-46, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30583231

RESUMO

Transmissible gastroenteritis coronavirus (TGEV) is enteropathogenic coronavirus that causes diarrhea in pigs, and is associated with high morbidity and mortality in sucking piglets. The TGEV membrane (M) protein is a decisive protein for the proliferation of viral proteins, and is associated with virus assembly and budding. To identify the cellular proteins that interact with the TGEV M protein, yeast two-hybrid screening was employed, and seven cellular proteins were identified M-binding partners. Using the GST pull-down approach and a CO-IP assay, the M protein was found to interact with porcine intestinal cells via eukaryotic translation initiation factor 4-alpha (EIF4A2), an essential component of the cellular translational machinery. Additionally, confocal microscopy revealed that EIF4A2 and M were colocalized in the cytoplasm. Furthermore, the function of EIF4A2 in intestinal cells during TGEV infection was examined. A knockdown of EIF4A2 by siRNA markedly decreased M protein proliferation and TGEV replication in target cells. Thus demonstrating that EIF4A2 plays a significant role in TGEV replication. The present study provides mechanistic insight into the interaction between the TGEV M protein and intestinal cells which contributes to the understanding of coronavirus replication and may be useful for the development of novel therapeutic strategies for TGEV infection.


Assuntos
Fator de Iniciação 4A em Eucariotos/genética , Vírus da Gastroenterite Transmissível/fisiologia , Proteínas da Matriz Viral/genética , Replicação Viral/fisiologia , Animais , Células Cultivadas , Fator de Iniciação 4A em Eucariotos/metabolismo , Gastroenterite Suína Transmissível/metabolismo , Técnicas de Silenciamento de Genes/veterinária , Intestinos/fisiologia , Intestinos/virologia , RNA Interferente Pequeno/metabolismo , Suínos , Vírus da Gastroenterite Transmissível/genética , Proteínas da Matriz Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...