Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virol J ; 21(1): 25, 2024 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-38263223

RESUMO

BACKGROUND: Pseudorabies virus (PRV) is one of the major viral pathogens leading to reproductive disorders in swine. However, little is known about the effects of PRV infection on porcine reproductive system. Ovarian granulosa cells are somatic cells surrounding oocytes in ovary and required for folliculogenesis. The present study aimed to investigate the interference of PRV on functions of porcine ovarian granulosa cells in vitro. METHODS: Primary granulosa cells were isolated from porcine ovaries. To investigate the PRV infectivity, transmission electron microscopy (TEM) was used to check the presence of viral particles, and the expression of viral gE gene was detected by quantitative real-time PCR (qPCR) in PRV-inoculated cells. After PRV infection, cell viability was detected by MTS assay, Ki67 for proliferative status was determined by immunofluorescence assay (IFA), cell cycle and apoptosis were detected by flow cytometry, and progesterone (P4) and estradiol (E2) were determined by radioimmunoassay. The checkpoint genes of cell cycle and apoptosis-related proteins were studied by qPCR and western blotting. RESULTS: Virus particles were observed in the nucleus and cytoplasm of PRV-infected granulosa cells by TEM imaging, and the expression of viral gE gene increased in a time-dependent manner post infection. PRV infection inhibited cell viability and blocked cell cycle at S phase in porcine granulosa cells, accompanied by decreases in expression of Ki67 protein and checkpoint genes related to S phase. Radioimmunoassay revealed decreased levels in P4 and E2, and the expressions of key steroidogenic enzymes were also down-regulated post PRV-infection. In addition, PRV induced apoptosis with an increase in Bax expression and activation of caspase 9, and the phosphorylation of JNK, ERK and p38 MAPKs were significantly up-regulated in porcine ovarian granulosa cells post PRV infection. CONCLUSIONS: The data indicate that PRV causes infection on porcine ovarian granulosa cells and interferes the cell functions through apoptosis, and the MAPK signaling pathway is involved in the viral pathogenesis.


Assuntos
Herpesvirus Suídeo 1 , Feminino , Suínos , Animais , Antígeno Ki-67 , Transdução de Sinais , Apoptose , Células da Granulosa
2.
Vet Microbiol ; 274: 109574, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36126504

RESUMO

Porcine parvovirus (PPV) is an important pathogen causing reproductive disorders in first pregnant sows. The non-structure protein NS1 of PPV is a multifunctional protein playing a key role in viral replication. Chaperonin-containing T-complex polypeptide complex (CCT), containing CCT1-CCT8 subunits, belongs to the type II chaperones that interact with proteins to help in folding and maintaining. In this study, CCT5, for the first time, was found to be one of the host interacting proteins of PPV NS1, and CCT5 was directly bound with NS1. Interference of CCT5 expression by specific siRNA and knockout of CCT5 expression by CRISPR/Cas9 suppressed PPV replication, while overexpression of CCT5 promoted PPV replication in PK-15 cells. The interaction of CCT5 and PPV NS1 was dependent on the 36-42 aa motif at the N-terminal end of NS1. More importantly, CCT5 was also found interacting with COPƐ, which has previously been demonstrated to promote PPV replication by regulating type I interferon. Interference and knockout of CCT5 expression significantly reduced the interaction of PPV NS1 and host protein COPƐ, and promoted the IFN-ß expression. These results show that CCT5 mediates the interaction of PPV NS1 and COPƐ to regulate viral replication, providing new insight into the mechanism of PPV replication.


Assuntos
Parvovirus Suíno , Gravidez , Suínos , Animais , Feminino , Parvovirus Suíno/genética , Proteínas não Estruturais Virais/metabolismo , RNA Interferente Pequeno , Replicação Viral , Chaperonina com TCP-1/genética , Interferon beta
3.
Vet Microbiol ; 272: 109497, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35785658

RESUMO

Transmissible gastroenteritis virus (TGEV) infection can lead to mitochondrial damage in porcine intestinal epithelial cells-jejunum 2 (IPEC-J2) cell line. The abnormal opening of mitochondrial permeability transition pore (mPTP) is the most important factor for mitochondrial damage. We previously demonstrated that circEZH2 could inhibit the abnormal opening of mPTP by binding miR-22. However, circEZH2 binding to miR-22 cannot completely enable mPTP opening to recover to normal level compared with the control group. So, we assume that circEZH2 also regulates the mPTP opening in other ways. To prove it, we identified the differentially expressed proteins (DEPs) caused by circEZH2 and circEZH2-interacting proteins by liquid chromatography-tandem mass spectrometry (LC-MS/MS). It turns out there are 123 DEPs (0.83 ≤ fold change ≥ 1.2) upon overexpression circEZH2 and 200 proteins interacted with circEZH2. The kyoto encyclopedia of genes and genomes (KEGG) analysis, gene ontology (GO) analysis, subcellular localization analysis, and protein interaction network results show that the DEPs and circEZH2-interacting proteins may involve in the regulation of mPTP opening. RNA immunoprecipitation (RIP) assay and flow cytometry (FCM) results indicate that circEZH2 can inhibit the opening of mPTP by interacting with Pi carrier (PiC, also named SLC25A3). Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and FCM results reveal that circEZH2 can inhibit mPTP opening by promoting the expression of radical s-adenosyl methionine domain-containing protein 2 (RSAD2). In addition, PiC can promote RSAD2 expression. The data indicate that circEZH2 inhibits TGEV-induced mPTP opening by interacting with PiC and upregulating RSAD2.


Assuntos
MicroRNAs , Vírus da Gastroenterite Transmissível , Animais , Cromatografia Líquida/veterinária , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Suínos , Espectrometria de Massas em Tandem/veterinária , Vírus da Gastroenterite Transmissível/metabolismo
4.
J Mol Endocrinol ; 68(2): 111-123, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-34910685

RESUMO

Effects of melatonin on the release and synthesis of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) at the hypothalamus and pituitary levels have been explored in some species, but a similar study in the corpora lutea (CL) has not yet been conducted. In this study, the immunostaining for GnRH and LH was observed in luteal cells of porcine CL during pregnancy, and a significant effect of pregnant stage on the level of GnRH and LH was found; higher values for GnRH and LH immunostaining and mRNA were detected in the early and mid-stages CL than in the later-stage CL (P < 0.01). Furthermore, the patterns of melatonin membrane receptors (MT1 and MT2) expression were consistent with those of GnRH and LH expression in the CL of pregnant sows; the relative levels of MT1 and MT2 in the early and mid-stages were significantly higher than those in the later-stage (P < 0.01). In luteal cells, melatonin dose-dependently increased in GnRH and LH secretion and mRNA expression. Melatonin also increased the GnRH-induced accumulation of LH and the LH-induced secretion of P4 in luteal cells. Additionally, the effects of melatonin on luteal GnRH and LH production were blocked by luzindole, a non-selective MT1 and MT2 receptor antagonist. Our results demonstrate the stimulatory effects of melatonin on GnRH and LH production in luteal cells of pregnant sows, suggesting a potential role for melatonin in luteal function through regulating the release and synthesis of GnRH and LH in luteal cells.


Assuntos
Células Lúteas , Melatonina , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Células Lúteas/metabolismo , Hormônio Luteinizante/metabolismo , Melatonina/metabolismo , Melatonina/farmacologia , Gravidez , Progesterona/metabolismo , Suínos
5.
Vet Med Sci ; 7(6): 2404-2409, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34547177

RESUMO

A 6-year-old female bottlenose dolphin (Tursiops truncatus) kept in dolphinarium died after a 3.5-month period of lethargy and inappetence despite antibiotics and supportive care. At necropsy, gross findings included diffuse varying-sized nodules in the lungs and scattered nodules throughout the heart, spleen, mesenteric and hilar lymph node and kidney. Microscopically, the lesions were characterised by disseminated fungal pyogranulomas with numerous intralesional Mucor-like fungi. The fungi structures were demonstrated by Periodic acid-Schiff and Gomori methenamine silver stain. Molecular analyses of the fungi were Rhizopus microsporus by PCR sequencing 18S ribosomal RNA gene. Ziehl-Neelsen stain failed to show acid-fast bacterial infection. Based on pathological and molecular examination, systemic granulomatous mucormycosis was diagnosed. To our knowledge, this is the first reported case of systemic mucormycosis caused by Rhizopus microsporus in bottlenose dolphin.


Assuntos
Golfinho Nariz-de-Garrafa , Mucormicose , Animais , Mucormicose/diagnóstico , Mucormicose/microbiologia , Mucormicose/veterinária , Rhizopus/genética
6.
Front Microbiol ; 12: 702881, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34394047

RESUMO

Type I interferons (IFN), a family of cytokines widely expressed in various tissues, play important roles in anti-infection immunity. Nevertheless, it is not known whether Brucella spp. could interfere with IFN-I production induced by other pathogens. This study investigated the regulatory roles of Brucella outer membrane protein (Omp)25 on the IFN-I signaling pathway and found that Omp25 inhibited the production of IFN-ß and its downstream IFN-stimulated genes induced by various DNA viruses or IFN-stimulatory DNA in human, murine, porcine, bovine, and ovine monocyte/macrophages or peripheral blood mononuclear cells. Brucella Omp25 suppressed the phosphorylation of stimulator of IFN genes (STINGs) and IFN regulatory factor 3 and nuclear translocation of phosphorylated IFN regulatory factor 3 in pseudorabies virus- or herpes simplex virus-1-infected murine, human, or porcine macrophages. Furthermore, we found that Brucella Omp25 promoted cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) degradation via the proteasome-dependent pathway, resulting in a decreased cyclic guanosine monophosphate-adenosine monophosphate production and downstream signaling activation upon DNA virus infection or IFN-stimulatory DNA stimulation. Mapping the predominant function domain of Omp25 showed that the amino acids 161 to 184 of Omp25 were required for Omp25-induced cGAS degradation, among which five amino acid residues (R176, Y179, R180, Y181, and Y184) were required for the inhibitory effect of Omp25 on IFN-ß induction. Altogether, our results demonstrated that Brucella Omp25 inhibits cGAS STING signaling pathway-induced IFN-ß via facilitating the ubiquitin-proteasome-dependent degradation of cGAS in various mammalian monocyte/macrophages.

7.
Vet Microbiol ; 261: 109189, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34375914

RESUMO

Transmissible gastroenteritis (TGE) is an acute viral disease and characterized as severe acute inflammation response that leads to diarrhea, vomiting, and high lethality of piglets. Transmissible gastroenteritis virus (TGEV), a member of coronavirus, is the pathogen of TGE. We previously found NF-κB pathway was activated and 65 miRNAs were changed in response to inflammation caused by TGEV in cell line porcine intestinal epithelial cells-jejunum 2 (IPEC-J2). Bioinformatics results showed that these altered miRNAs were relevant to inflammation. In this study, the candidate targets of differentially expressed (DE) miRNAs were predicted and analyzed using Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Based on the results of KEGG analysis, miR-885-3p might participate in regulating activation of NF-κB pathway and TNF pathway. To study the function of miR-885-3p, miR-885-3p mimics and inhibitors were artificially synthesized and respectively used for overexpression and silence of miR-885-3p in cells. Our results showed that miR-885-3p inhibited NF-κB signaling pathway and tumor necrosis factor-α (TNF-α) production. B-cell CLL/lymphoma 10 (Bcl-10) was identified as the target of miR-885-3p, and promoted NF-κB pathway activation and TNF-α production. It was found that TGEV open reading frame 3b (TGEV-ORF3b) suppressed Bcl-10 expression, activation of NF-κB pathway, and TNF-α production by uniquely up-regulated miR-885-3p expression. Overall, the results indicated that TGEV-ORF3b counteracted NF-κB pathway and TNF-α via regulating miR-885-3p and Bcl-10.


Assuntos
Proteína 10 de Linfoma CCL de Células B/metabolismo , Gastroenterite Suína Transmissível/virologia , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteína 10 de Linfoma CCL de Células B/genética , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação da Expressão Gênica , Mucosa Intestinal/citologia , MicroRNAs/genética , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Suínos , Regulação para Cima , Proteínas Virais
8.
Int Immunopharmacol ; 96: 107783, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34162147

RESUMO

MicroRNAs play a variety of roles in the progress of inflammation. Herein, we investigated the roles of miR-223 in governing macrophage polarization balance in the progress of sepsis. We firstly observed that miR-223 was down-regulated at the early phase and up-regulated at the late phase of sepsis in macrophages; the levels of miR-223 were positively correlated to the ratio of M2 macrophages during sepsis. In miR-223 knockout mice, we observed that miR-223 was dispensable for efficient pro-inflammatory responses, but was required for efficient M2-associated phenotype and function. miR-223 deletion increased clinical scores of sepsis, leading to increased mortality in septic mice. Furthermore, we found that miR-223 expression in M2-type macrophages was controlled by interleukin (IL)-4, but not IL-10; IL-4 antibodies were able to downregulate the levels of miR-223, increased the expression of targeted genes Nfat5 and Rasa1, reduced the ratio of M2 macrophages, resulting in a decreased survival rate in septic mice. Meanwhile, miR-223 deficient macrophages appeared a markedly decreased M2-type polarization when induced by IL-4, but did not affect macrophages skew to M2 phenotype induced by IL-10. Taken together, our results demonstrate that miR-223 acts as an important regulator to modulate IL-4-meditated M2-type polarization of macrophages via targeting to Nfat5 and Rasa1 to control the progress of sepsis.


Assuntos
Interleucina-4/imunologia , Macrófagos/imunologia , MicroRNAs/imunologia , Sepse/genética , Sepse/imunologia , Animais , Carga Bacteriana , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cavidade Peritoneal/microbiologia , Fagocitose , Fenótipo , Sepse/microbiologia
9.
Viruses ; 11(12)2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31835539

RESUMO

Porcine circovirus type 2 (PCV2) is one of the major threats to pig farms worldwide. Although PCV2 has been identified to promote IL-10 production, the detailed regulatory roles of PCV2 Rep for IL-10 production remain unclear. Herein, we first found that PCV2 Rep, rather than PCV1 Rep, enhanced IL-10 expression at the later phase of PCV2 infection in porcine alveolar macrophages (PAMs). Furthermore, we found that PCV2 Rep directly activated the p38-MAPK pathway to promote transcription factors NF-κB p50 and Sp1 binding to the il10 promoter, but PCV1 Rep did not. During PCV2 infection, however, PCV2 Rep promoted the binding activities of NF-κB p50 and Sp1 with the il10 promoter only at the later phase of PCV2 infection, since Rep proteins only expressed at the later phase of the infection. Moreover, silence of the thymine DNA glycosylase (TDG), a Rep-binding protein, significantly reduced the binding activities of NF-κB p50 and Sp1 with il10 promoter, resulting in the reduction of IL-10 production in PCV2-inoculated PAMs at the later phase of infection. Taken together, our results demonstrate that Rep proteins enhance IL-10 production during PCV2 infection of PAMs via activation of p38-MAPK pathways, in which host TDG is a critical mediator.


Assuntos
Circovirus/fisiologia , Interleucina-10/biossíntese , Sistema de Sinalização das MAP Quinases , Macrófagos/imunologia , Macrófagos/metabolismo , Síndrome Definhante Multissistêmico de Suínos Desmamados/metabolismo , Síndrome Definhante Multissistêmico de Suínos Desmamados/virologia , Proteínas Virais/metabolismo , Animais , Expressão Gênica , Interações Hospedeiro-Patógeno , Interleucina-10/genética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Síndrome Definhante Multissistêmico de Suínos Desmamados/imunologia , Regiões Promotoras Genéticas , Suínos , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
BMC Genomics ; 20(1): 806, 2019 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-31684870

RESUMO

BACKGROUND: Transmissible gastroenteritis virus (TGEV) infection can cause acute inflammation. Long noncoding RNAs (lncRNAs) play important roles in a number of biological process including inflammation response. However, whether lncRNAs participate in TGEV-induced inflammation in porcine intestinal epithelial cells (IPECs) is largely unknown. RESULTS: In this study, the next-generation sequencing (NGS) technology was used to analyze the profiles of lncRNAs in Mock and TGEV-infected porcine intestinal epithelial cell-jejunum 2 (IPEC-J2) cell line. A total of 106 lncRNAs were differentially expressed. Many differentially expressed lncRNAs act as elements to competitively attach microRNAs (miRNAs) which target to messenger RNA (mRNAs) to mediate expression of genes that related to toll-like receptors (TLRs), NOD-like receptors (NLRs), tumor necrosis factor (TNF), and RIG-I-like receptors (RLRs) pathways. Functional analysis of the binding proteins and the up/down-stream genes of the differentially expressed lncRNAs revealed that lncRNAs were principally related to inflammatory response. Meanwhile, we found that the differentially expressed lncRNA TCONS_00058367 might lead to a reduction of phosphorylation of transcription factor p65 (p-p65) in TGEV-infected IPEC-J2 cells by negatively regulating its antisense gene promyelocytic leukemia (PML). CONCLUSIONS: The data showed that differentially expressed lncRNAs might be involved in inflammatory response induced by TGEV through acting as miRNA sponges, regulating their up/down-stream genes, or directly binding proteins.


Assuntos
Gastroenterite Suína Transmissível/genética , RNA Longo não Codificante/genética , Vírus da Gastroenterite Transmissível/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Sequenciamento de Nucleotídeos em Larga Escala , Inflamação/genética , MicroRNAs/genética , Suínos
11.
Front Pharmacol ; 10: 937, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31555129

RESUMO

Arctigenin (ARG) has been reported to be a bioactive lignan from Arctium lappa exerting various activities including anti-cancer and immune-regulation. The present study aimed to investigate the anti-metastasis activity and mechanism of ARG against hepatocellular carcinoma in vitro and in vivo. The results showed that ARG exhibited a significant cytotoxicity on Hep G2 and SMMC 7721 cells (but not on normal liver cells LO2). In addition, the migration and invasion of Hep G2 and SMMC 7721 cells were also remarkably repressed. Furthermore, ARG attenuated Wnt/ß-catenin signaling activation, resulting in the down-regulation of ß-catenin target genes including c-Myc, cyclin D1, MMP-9, and ZO-1. Noticeably, ARG attenuated the activation of Wnt/ß-catenin through a GSK3ß-dependent pathway. Besides, we also found that ARG potentially inhibited epithelial-mesenchymal transition by up-regulating the epithelial and down-regulating the mesenchymal marker proteins. In vivo, intraperitoneal injection of ARG not only significantly inhibited the growth of subcutaneous transplanted tumor but also dramatically alleviated the tumor metastasis in liver. Our data demonstrated that ARG exerted anti-epithelial-mesenchymal transition and anti-metastasis activities against hepatocellular carcinoma, which might make it a candidate as a preventive agent for cancer metastasis.

12.
Res Vet Sci ; 119: 109-115, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29909129

RESUMO

This essay focuses on transmissible gastroenteritis virus (TGEV), which is an enteropathogenic virus related to contagious and acute diseases in suckling piglets. Previous literature suggests that the TGEV nucleocapsid protein (N) plays a significant role in viral transcriptional process, however, there is a need to examine other functions of TGEV N protein in the porcine intestinal epithelial cell (IEC) which is the target cell of TGEV. In the present study, we investigated the degradation, subcellular localisation, and function of TGEV N protein by examining its effects on cycle progression, endoplasmic reticulum (ER) stress, interleukin-8 (IL-8) expression, and cell survival. The results showed that TGEV N protein localised in the cytoplasm, inhibited IEC growth, prolonged the S-phase cell cycle by down-regulating cell cycle protein cyclin A, and was mainly degraded through the proteasome pathway. Moreover, TGEV N protein induced ER stress and activated NF-κB, which was responsible for the up-regulation of IL-8 and Bcl-2 expression. This report mainly considers the functions of TGEV N protein in IEC. To be specific, in IEC, TGEV N protein induces cell cycle prolongation at the S-phase, ER stress and up-regulates IL-8 expression. These results provide a better understanding of the functions and structural mechanisms of TGEV N protein.


Assuntos
Estresse do Retículo Endoplasmático , Interleucina-8/metabolismo , Vírus da Gastroenterite Transmissível/patogenicidade , Animais , Células Epiteliais , Gastroenterite Suína Transmissível , Intestinos , Suínos
13.
Front Pharmacol ; 9: 475, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867481

RESUMO

Arctigenin (ARG), one of the most active ingredients abstracted from seeds of Arctium lappa L., has been proved to exert promising biological activities such as immunomodulatory, anti-viral, and anti-cancer etc. However, the mechanism behind its immunomodulatory function still remains elusive to be further investigated. In this study, we found that ARG had no significant effects on the cell proliferation in both porcine alveolar macrophage cell line (3D4/21) and primary porcine derived alveolar macrophage. It remarkably increased the expression and secretion of the two cytokines including tumor necrosis factor-alpha (TNF-α) and transforming growth factor beta1 (TGF-ß1) in a dose-dependent manner with the concomitant enhancement of phagocytosis, which are the indicators of macrophage activation. ARG also elevated the intracellular reactive oxygen species (ROS) production by activating NOX2-based NADPH oxidase. Furthermore, inhibition of ROS generation by diphenyliodonium and apocynin significantly suppressed ARG-induced cytokine secretion and phagocytosis increase, indicating the requirement of ROS for the porcine alveolar macrophage activation. In addition, TLR6-My88 excitation, p38 MAPK and ERK1/2 phosphorylation were all involved in the process. As blocking TLR6 receptor dramatically attenuated the NOX2 oxidase activation, cytokine secretion and phagocytosis increase. Inhibiting ROS generation almost abolished p38 and ERK1/2 phosphorylation, and the cytokine secretion could also be remarkably reduced by p38 and ERK1/2 inhibitors (SB203580 and UO126). Our finding gave a new insight of understanding that ARG could improve the immune-function of porcine alveolar macrophages through TLR6-NOX2 oxidase-MAPKs signaling pathway.

14.
Gen Comp Endocrinol ; 260: 115-124, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29366624

RESUMO

Little is known about ghrelin's effects on cell proliferation in pregnant mammary epithelial cells (MECs) even though it is known to be a mitogen for a variety of other cell types. The objectives of this study were to evaluate the expression and localization of ghrelin and its functional receptor, GHSR-1a, in the mammary glands of dairy goats during pregnancy and to investigate the direct role of ghrelin in cell proliferation of primary cultured MECs. Compared to the early stage (days 30) of pregnancy, the abundance of transcripts and protein of ghrelin and GHSR-1a were significantly greater in mid- and late-phases (between days 90 and days 120) of pregnancy (p < .05). Immunohistochemistry analysis showed that ghrelin and GHSR-1a were predominantly localized in the alveolar and ductal mammary epithelial cells at various stages of pregnancy. In our in vitro experiments, ghrelin induced a dose- and time-dependent promotory effect on cell proliferation of MECs. At the dose of 103 pg/mL treatment 24 h, ghrelin augmented the expression of proliferation-related peptides (PCNA and cyclin B1). Furthermore, ghrelin promoted the expression of prolactin (PRL) and GHSR-1a in cultured MECs. Additionally, the stimulatory effects of ghrelin were blocked by d-Lys3-GHRP6, a selective antagonist of GHSR-1a. As the temporal changes in ghrelin and GHSR-1a expression in pregnant goat mammary glands coincided with the mammary growth and development during the pregnancy, activation of GHSR-1a signal transduction pathways by ghrelin may play a direct role in the regulation of mammary growth in dairy goats.


Assuntos
Proliferação de Células , Células Epiteliais/fisiologia , Grelina/genética , Grelina/farmacologia , Cabras , Glândulas Mamárias Animais/metabolismo , Prenhez , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Indústria de Laticínios , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Grelina/metabolismo , Cabras/genética , Cabras/fisiologia , Lactação/genética , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Gravidez , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Biol Reprod ; 98(4): 558-569, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29360968

RESUMO

Porcine parvovirus (PPV) is a major virus that leads to fetal death in swine. However, the effects of PPV infection on sows are poorly understood. The aim of this study was to investigate the effects of PPV on porcine steroidogenic luteal cells (SLCs) survival and functions and underlying mechanisms. In vivo experiment results showed that artificial infection of PPV significantly reduced the concentration of serum progesterone and induced histopathological lesions and SLCs apoptosis in porcine corpora luteum. In in vitro cultured primary porcine SLCs, PPV could infect and replicate in SLCs and induced SLCs apoptosis through mitochondria, but not the death receptor, mediated apoptosis pathway. Meanwhile, PPV infection also decreased progesterone production in SLCs. Moreover, PPV infection could increase active p53 transcriptional activity and protein expression as well as promoting p53 translocation to nucleus. Using the p53-specific pharmacological inhibitor (pifithrin-α) and siRNA could partially attenuate PPV-induced Bax upregulation, caspase-3 activation, apoptosis, and the reduction of progesterone production in primary porcine SLCs. Furthermore, the phosphorylation of p38 mitogen-activated protein kinase (MAPK) was also increased in PPV-infected SLCs. Pretreatment with p38 MAPK inhibitor (SB203580) suppressed PPV-induced p53 accumulation and translocation, SLCs apoptosis, and progesterone production reduction. In summary, these findings indicate that PPV could induce SLCs apoptosis and a decrease of progesterone production in vivo and in vitro via p38 MAPK signaling and p53-dependent mitochondrial pathway, which provides the potential clinical therapy methods for PPV infection.


Assuntos
Apoptose/fisiologia , Células Lúteas/metabolismo , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Infecções por Parvoviridae/veterinária , Progesterona/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Feminino , Células Lúteas/virologia , Mitocôndrias/virologia , Infecções por Parvoviridae/metabolismo , Parvovirus Suíno , Fosforilação , Transdução de Sinais/fisiologia , Suínos
16.
Theriogenology ; 108: 297-305, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29277070

RESUMO

The direct effect of melatonin on porcine luteal function during the pregnancy remains unknown. The objective of the study was to analyse the molecular mechanism(s) by which melatonin directly affects progesterone (P4) production in the corpus luteum (CL) of pregnant sows. We evaluated the localization of melatonin membrane receptors (MT1 and MT2) in CL, and investigated the effect of melatonin on P4 secretion along with the expression of P4 synthesis intermediates in luteal cells. Immunohistochemistry analysis showed that MT1 and MT2 were predominantly localized in luteal cells in pregnant luteal tissues. The results of our in vitro experiments showed that melatonin from 5 to 625 pg/mL was able to significantly increase P4 release (P < 0.05) in a dose-dependent manner. And at the dose of 125 pg/mL treatment, the time-dependent effect on P4 secretion was observed. Furthermore, melatonin from 5 to 625 pg/mL up-regulated both P450scc and StAR expression (P < 0.05) in a dose-dependent manner, and the effect was also time-dependent. No difference of 3ß hydroxysteroid dehydrogenase (3ß-HSD) expression was observed between control and treatment groups. In addition, melatonin induced a dose- and time-dependent promotion on cell viability. Additionally, the stimulatory effects of melatonin were blocked by luzindole, a non-selective MT1 and MT2 receptor antagonist, or partially blocked by a selective MT2 ligand, 4-phenyl-2-propionamidotetralin (4P-PDOT). The data support the presence of MT1 and MT2 in porcine CL and a regulatory role for melatonin in luteal function through MT1 and MT2-mediated signal transduction pathways.


Assuntos
Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Corpo Lúteo/efeitos dos fármacos , Melatonina/farmacologia , Fosfoproteínas/metabolismo , Progesterona/metabolismo , Suínos , Animais , Corpo Lúteo/metabolismo , Feminino , Imuno-Histoquímica , Receptor MT1 de Melatonina/análise , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/análise , Receptor MT2 de Melatonina/metabolismo
17.
Mol Cell Proteomics ; 17(2): 190-204, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29217619

RESUMO

Transmissible gastroenteritis virus (TGEV), a member of the coronaviridae family, could cause fatal diarrhea of piglets and result in numerous economic losses. Previous studies demonstrated that TGEV infection could lead to mitochondrial damage and upregulate miR-4331 level. So miR-4331 may play an important regulatory role in the control of mitochondrial function. To explore the potential role of miR-4331 in mitochondrial damage, we adopted a strategy consisting of quantitative proteomic analysis of porcine kidney (PK-15) cells in response to miR-4331 and TGEV infection. Eventually, 69 differentially expressed proteins were gained. The target of miR-4331 was identified. The effects of miR-4331 and its target RB1 on mitochondrial Ca2+ level, mitochondrial membrane potential (MMP), interleukin-1 receptor accessory protein (IL1RAP), p38 MAPK signaling pathway were investigated. The results showed that miR-4331 elevated mitochondrial Ca2+ level, reduced MMP, targets Retinoblastoma 1 (RB1), upregulated IL1RAP, and induced activation of p38 MAPK pathway during TGEV infection. RB1 was identified as the direct targets of miR-4331 and downregulated IL1RAP, suppressed the activation of p38 MPAK, and attenuated TGEV-induced mitochondrial damage. In addition, IL1RAP played a positive role in activating p38 MAPK signaling and negative role in TGEV-induced mitochondrial damage. The data indicate that miR-4331 aggravates TGEV-induced mitochondrial damage by repressing expression of RB1, promoting IL1RAP, and activating p38 MAPK pathway.


Assuntos
Proteína Acessória do Receptor de Interleucina-1/metabolismo , MicroRNAs , Mitocôndrias/fisiologia , Proteína do Retinoblastoma/metabolismo , Vírus da Gastroenterite Transmissível , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Proteína Acessória do Receptor de Interleucina-1/genética , Potencial da Membrana Mitocondrial , Proteômica , Proteína do Retinoblastoma/genética , Suínos
18.
Vaccine ; 35(36): 4722-4729, 2017 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-28755834

RESUMO

Porcine circovirus type 2 (PCV2) is the main pathogen of porcine circovirus associated disease (PCVAD), causing great economic losses in pig industry. In previous study, we constructed adenovirus vector vaccines expressing PCV2 Cap either modified with Intron A and WPRE, or CD40L and GMCSF, and evaluated all of these vaccines in mice and in pigs. Although Ad-A-C-W and Ad-CD40L-Cap-GMCSF could induce stronger immune responses than Ad-Cap, neither of them was better than commercial inactivated vaccine PCV2 SH-strain. In this study, secretory recombinant adenoviruses (Ad-A-spCap-W and Ad-A-spCD40L-spCap-spGMCSF-W) and non-secretory recombinant adenovirus Ad-A-CD40L-Cap-GMCSF-W were constructed, and identified by western blot and confocal laser microscope observation. The results of ELISA and VN showed that humoral immune responses induced by Ad-A-spCap-W and Ad-A-CD40L-Cap-GMCSF-W were not significantly different from SH-strain, but Ad-A-spCD40L-spCap-spGMCSF-W could induce significantly higher humoral immune response than SH-strain. Lymphocytes proliferative and cytokines releasing levels of Ad-A-spCap-W and Ad-A-CD40L-Cap-GMCSF-W were not significantly different from SH-strain, but Ad-A-spCD40L-spCap-spGMCSF-W was significantly higher than SH-strain. PCV2-challenge experiment showed that virus loads were significantly reduced in Ad-A-spCD40L-spCap-spGMCSF-W vaccinated group, and no obviously clinical and microscopic lesions were observed in Ad-A-spCD40L-spCap-spGMCSF-W vaccinated group. Altogether, these results demonstrate that recombinant adenovirus vaccine Ad-A-spCD40L-spCap-spGMCSF-W induces stronger immune responses and provides better protection than commercial inactivated vaccine PCV2 SH-strain, and suggest that Ad-A-spCD40L-spCap-spGMCSF-W could be a potential vaccine candidate against PCVAD.


Assuntos
Vacinas contra Adenovirus/imunologia , Proteínas do Capsídeo/imunologia , Circovirus/imunologia , Imunogenicidade da Vacina , Vacinas Virais/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Vacinas contra Adenovirus/administração & dosagem , Vacinas contra Adenovirus/genética , Adjuvantes Imunológicos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Ligante de CD40/genética , Ligante de CD40/imunologia , Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/genética , Infecções por Circoviridae/prevenção & controle , Circovirus/genética , Citocinas/biossíntese , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunidade Humoral , Camundongos , Suínos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Carga Viral , Vacinas Virais/administração & dosagem
19.
Theriogenology ; 94: 105-113, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28407852

RESUMO

Luteal cells play a crucial role in pregnancy through secreting progesterone to maintain pregnancy and support of fetus. However, low cellular yields and inability to passage primary porcine luteal cells (PLCs) in vitro limit the luteal cell study. Therefore, developing an immortalized porcine luteal cell line is necessary for studying luteal cells activity and function in different diseases. In this study, primary PLCs were obtained from gilts at day 30 to day 50 of gestation and immortalized by human telomerase reverse transcriptase (hTERT). The porcine corpus luteal cell line (hTERT-PLCs) expressed hTERT gene steady, maintained high hTERT activity and normal karyotype. The phase contrast microscope and transmission electron microscope observation showed primary PLCs and hTERT-PLCs were polygonal and exhibited abundant mitochondria, smooth endoplasmic reticulum and lipid droplets. 3ß hydroxysteroid dehydrogenase (3ßHSD) and Oil-Red-O staining showed that hTERT-PLCs at passage 30 and 50 were similar to primary PLCs. The hTERT-PLCs expressed steroidogenesis-related proteins, enzymes and receptors, such as steroidogenic acute regulatory protein, P450 cholesterol side-chain cleavage, 3ßHSD, 20αHSD, luteinizing hormone receptor, progesterone receptor, prolactin receptor, estrogen receptorα/ß, as well as primary PLCs. Consequently, hTERT-PLCs could secret progesterone and exhibited similar responses to luteinizing hormone and prostaglandin F2α as primary PLCs. In addition, the hTERT-PLCs did not show neoplastic transformation or anchorage independent growth. In summary, we developed an immortalized porcine luteal cell line which maintained its originally morphological, biological and functional characteristics.


Assuntos
Linhagem Celular , Células Lúteas/citologia , Suínos , Animais , Western Blotting , Feminino , Células HeLa , Humanos , Cariótipo , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Gravidez , Progesterona/metabolismo , Telomerase/farmacologia
20.
Front Immunol ; 8: 2013, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29387067

RESUMO

Brucella spp. impedes the production of pro-inflammatory cytokines by its outer membrane protein Omp25 in order to promote survival and immune evasion. However, how Omp25 regulates tumor necrosis factor (TNF-α) expression in different mammalian macrophages remains unclear. In this study, we investigated the potential mechanisms by which Omp25 regulates TNF-α expression and found that Omp25-deficient mutant of B. suis exhibited an enhanced TNF-α expression compared with wild-type (WT) B. suis, whereas ectopic expression of Omp25 suppressed LPS-induced TNF-α production at both protein and mRNA levels in porcine alveolar macrophages (PAMs) and murine macrophage RAW264.7 cells. We observed that Omp25 protein as well as WT B. suis upregulated miR-146a, -181a, -181b, and -301a-3p and downregulated TRAF6 and IRAK1 in both PAMs and RAW264.7 cells, but separately upregulates miR-130a-3p in PAMs and miR-351-5p in RAW264.7. The upregulation of miR-146a or miR-351-5p attenuated TNF-α transcription by targeting TRAF6 and IRAK1 at the 3' untranslated region (UTR), resulting in inhibition of NF-kB pathway, while upregulation of miR-130a-3p, -181a, or -301a-3p correlated temporally with decreased TNF-α by targeting its 3'UTR in PAMs or RAW264.7 cells. In contrast, inhibition of miR-130a-3p, -146a, -181a, and -301a-3p attenuated the inhibitory effects of Omp25 on LPS-induced TNF-α in PAMs, while inhibition of miR-146a, -181a, -301a-3p, and -351-5p attenuated the inhibitory effects of Omp25 in RAW264.7, resulting in an increased TNF-α production and decreased intracellular bacteria in both cells. Taken together, our results demonstrate that Brucella downregulates TNF-α to promote intracellular survival via Omp25 regulation of different microRNAs in porcine and murine macrophages.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA