Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Sci Adv ; 10(15): eadk0002, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38598630

ABSTRACT

Continuity of behaviors requires animals to make smooth transitions between mutually exclusive behavioral states. Neural principles that govern these transitions are not well understood. Caenorhabditis elegans spontaneously switch between two opposite motor states, forward and backward movement, a phenomenon thought to reflect the reciprocal inhibition between interneurons AVB and AVA. Here, we report that spontaneous locomotion and their corresponding motor circuits are not separately controlled. AVA and AVB are neither functionally equivalent nor strictly reciprocally inhibitory. AVA, but not AVB, maintains a depolarized membrane potential. While AVA phasically inhibits the forward promoting interneuron AVB at a fast timescale, it maintains a tonic, extrasynaptic excitation on AVB over the longer timescale. We propose that AVA, with tonic and phasic activity of opposite polarities on different timescales, acts as a master neuron to break the symmetry between the underlying forward and backward motor circuits. This master neuron model offers a parsimonious solution for sustained locomotion consisted of mutually exclusive motor states.


Subject(s)
Caenorhabditis elegans Proteins , Neurons , Animals , Caenorhabditis elegans/physiology , Interneurons/physiology
2.
Appl Microbiol Biotechnol ; 108(1): 173, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38267794

ABSTRACT

Pyroptosis is a newly discovered type of pro-inflammatory programmed cell death that plays a vital role in various processes such as inflammations, immune responses, and pathogen infections. As one of the main executioners of pyroptosis, gasdermin D (GSDMD) is a membrane pore-forming protein that typically exists in a self-inhibitory state. Once activated, GSDMD will be cleaved into an N-terminal fragment with pore-forming activity, becoming the key indicator of pyroptosis activation, and a C-terminal fragment. Although commercial antibodies against human and murine GSDMD proteins are currently available, their reactivity with porcine GSDMD (pGSDMD) is poor, which limits research on the biological functions of pGSDMD and pyroptosis in pigs in vivo and in vitro. Here, five monoclonal antibodies (mAbs) were prepared by immunizing BALB/c mice with procaryotically expressed full-length pGSDMD, all of which did not cross react with human and murine GSDMD proteins. Epitope mapping demonstrated that 15H6 recognizes amino acids (aa) at positions 28-34 of pGSDMD (LQTSDRF), 19H3 recognizes 257-260aa (PPQF), 23H10 and 27A10 recognize 78-82aa (GPFYF), and 25E2 recognizes 429-435aa (PPTLLGS). The affinity constant and isotype of 15H6, 19H3, 23H10, 27A10, and 25E2 mAbs were determined to be 1.32 × 10-9, 3.66 × 10-9, 9.04 × 10-9, 1.83 × 10-9, and 8.00 × 10-8 mol/L and IgG1/κ, IgG2a/κ, IgG2a/κ, IgG1/κ, and IgG1/κ, respectively. Heavy- and light-chain variable regions sequencing showed that the heavy-chain complementarity-determining region (CDR) sequences of all five mAbs are completely different, while the light-chain CDR sequences of the four mAbs that recognize the N-terminus of pGSDMD are identical. Our prepared mAbs provide valuable materials for studying pGSDMD function and pyroptosis. KEY POINTS: • A total of five mouse anti-pGSDMD mAbs were prepared, of which four recognize the N-terminus of pGSDMD and one recognize its C-terminus. • The main performance parameters of the five mAbs, including epitope, antibody titer, affinity constant, isotype, and heavy- and light-chain CDR, were characterized. • All five mAbs specifically recognize pGSDMD protein and do not cross react with human and murine GSDMD proteins.


Subject(s)
Antibodies, Monoclonal , Gasdermins , Humans , Swine , Animals , Mice , Immunosuppressive Agents , Porins , Immunoglobulin G , Mice, Inbred BALB C
3.
Vet Microbiol ; 289: 109957, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38160508

ABSTRACT

It is a common sense that porcine reproductive and respiratory syndrome virus (PRRSV) infection could cause immune failure of classical swine fever (CSF) vaccine, and porcine alveolar macrophages (PAMs) are the target cells of both. To elucidate the role of macrophage polarization in PRRSV infection induced CSF vaccine failure, an immortal porcine alveolar macrophage line PAM39 cell line was used to investigate the effect of PRRSV or/and CSFV C-strain (CSFV-C) infection on macrophage polarization in vitro. Interestingly, PRRSV single infection or PRRSV co-infection with CSFV-C promoted PAM39 cells to M1, while CSFV-C single infection induced PAM39 cells to M2. After the construction of M1 and M2 PAM39 cells polarization models, M1 polarized PAM39 cells were found to inhibit the replication of CSFV-C, and Chinese medicine such as matrine, ginsenosides and astragalus polysaccharides could alleviate the polarization of PAM39 cells and the replication of CSFV-C. Furthermore, interferon (IFN)-γ and lipopolysaccharide (LPS) co-stimulation induced NF-κB activation while matrine treatment blocked M1 polarization-induced NF-κB pathway activation. These findings provided a theoretical basis for designing a new strategy to improve the immune effect of CSFV-C based on porcine alveolar macrophage polarization subtypes.


Subject(s)
Classical Swine Fever , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Vaccines , Swine , Animals , NF-kappa B/metabolism , Matrines , Classical Swine Fever/prevention & control , Macrophages, Alveolar , Virus Replication , Porcine Reproductive and Respiratory Syndrome/metabolism , Swine Diseases/metabolism
4.
Autophagy ; : 1-28, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38084826

ABSTRACT

Senecavirus A (SVA) is a newly emerging picornavirus associated with swine vesicular lesions and neonatal mortality, threatening the global pig industry. Despite sustained efforts, the molecular mechanisms of SVA pathogenesis have not yet been fully elucidated. Here, we demonstrate for the first time that SVA infection can induce complete mitophagy in host cells, which depends on SVA replication. Mitophagy has been subsequently proven to promote SVA replication in host cells. Genome-wide screening of SVA proteins involved in inducing mitophagy showed that although VP2, VP3, 2C, and 3A proteins can independently induce mitophagy, only the 2C protein mediates mitophagy through direct interaction with TUFM (Tu translation elongation factor, mitochondrial). The glutamic acids at positions 196 and 211 of TUFM were shown to be two key sites for its interaction with 2C protein. Moreover, TUFM was discovered to interact directly with BECN1 and indirectly with the ATG12-ATG5 conjugate. Further experiments revealed that TUFM needs to undergo ubiquitination modification before being recognized by the macroautophagy/autophagy receptor protein SQSTM1/p62, and E3 ubiquitin ligase RNF185 catalyzes K27-linked polyubiquitination of TUFM through the interaction between RNF185's transmembrane domain 1 and TUFM to initiate SVA-induced mitophagy. The ubiquitinated TUFM is recognized and bound by SQSTM1, which in turn interacts with MAP1LC3/LC3, thereby linking the 2C-anchored mitochondria to the phagophore for sequestration into mitophagosomes, which ultimately fuse with lysosomes to achieve complete mitophagy. Overall, our results elucidated the molecular mechanism by which SVA induces mitophagy to promote self-replication and provide new insights into SVA pathogenesis.Abbreviations: aa: amino acid; Baf A1: bafilomycin A1; BHK-21: baby hamster kidney-21; CCCP: carbonyl cyanide m-chlorophenyl hydrazone; co-IP: co-immunoprecipitation; CQ: chloroquine; DAPI: 4',6-diamidino-2'-phenylindole; DMSO: dimethyl sulfoxide; EGFP: enhanced green fluorescent protein; ER: endoplasmic reticulum; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GST: glutathione S-transferase; HA: hemagglutinin; hpi: hours post-infection; hpt: hours post-transfection; IPTG: isopropyl ß-D-1-thiogalactopyranoside; mAb: monoclonal antibody; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAVS: mitochondrial antiviral signaling protein; Mdivi-1: mitochondrial division inhibitor-1; MOI: multiplicity of infection; mRFP: monomeric red fluorescent protein; MS: mass spectrometry; ORF: open reading frame; PBS: phosphate-buffered saline; SD: standard deviation; SQSTM1/p62: sequestosome 1; ST: swine testis; SVA: Senecavirus A; TCID50: 50% tissue culture infectious dose; TIMM23: translocase of inner mitochondrial membrane 23; TM: transmembrane; TOMM20: translocase of outer mitochondrial membrane 20; TUFM: Tu translation elongation factor, mitochondrial; Ub: ubiquitin; UV: ultraviolet; VDAC1: voltage dependent anion channel 1; WT: wild-type; µg: microgram; µm: micrometer; µM: micromole.

5.
Proc Natl Acad Sci U S A ; 120(48): e2309506120, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37983498

ABSTRACT

African swine fever virus (ASFV), a devastating pathogen to the worldwide swine industry, mainly targets macrophage/monocyte lineage, but how the virus enters host cells has remained unclear. Here, we report that ASFV utilizes apoptotic bodies (ApoBDs) for infection and cell-cell transmission. We show that ASFV induces cell apoptosis of primary porcine alveolar macrophages (PAMs) at the late stage of infection to productively shed ApoBDs that are subsequently swallowed by neighboring PAMs to initiate a secondary infection as evidenced by electron microscopy and live-cell imaging. Interestingly, the virions loaded within ApoBDs are exclusively single-enveloped particles that are devoid of the outer layer of membrane and represent a predominant form produced during late infection. The in vitro purified ApoBD vesicles are capable of mediating virus infection of naive PAMs, but the transmission can be significantly inhibited by blocking the "eat-me" signal phosphatidyserine on the surface of ApoBDs via Annexin V or the efferocytosis receptor TIM4 on the recipient PAMs via anti-TIM4 antibody, whereas overexpression of TIM4 enhances virus infection. The same treatment however did not affect the infection by intracellular viruses. Importantly, the swine sera to ASFV exert no effect on the ApoBD-mediated transmission but can partially act on the virions lacking the outer layer of membrane. Thus, ASFV has evolved to hijack a normal cellular pathway for cell-cell spread to evade host responses.


Subject(s)
African Swine Fever Virus , African Swine Fever , Extracellular Vesicles , Swine , Animals , African Swine Fever Virus/physiology , Macrophages/metabolism , Monocytes/metabolism , Extracellular Vesicles/metabolism
6.
Article in English | MEDLINE | ID: mdl-37885106

ABSTRACT

BACKGROUND: Excessive insulin is the leading cause of metabolic syndromes besides hyperinsulinemia. Insulin-lowering therapeutic peptides have been poorly studied and warrant urgent attention. OBJECTIVE: The main purpose of this study, was to introduce a novel peptide COX52-69 that was initially isolated from the porcine small intestine and possessed the ability to inhibit insulin secretion under high-glucose conditions by modulating large conductance Ca2+-activated K+ channels (BK channels) activity. METHODS AND RESULTS: Enzyme-linked immunosorbent assay results indicate that COX52-69 supressed insulin release induced by high glucose levels in pancreatic islets and animal models. Furthermore, electrophysiological data demonstrated that COX52-69 can increase BK channel currents and hyperpolarize cell membranes. Thus, cell excitability decreased, corresponding to a reduction in insulin secretion. CONCLUSION: Our study provides a novel approach to modulate high glucose-stimulated insulin secretion in patients with hyperinsulinemia.

7.
EMBO Rep ; 24(11): e57014, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37811674

ABSTRACT

Excitation/inhibition (E/I) balance is carefully maintained by the nervous system. The neurotransmitter GABA has been reported to be co-released with its sole precursor, the neurotransmitter glutamate. The genetic and circuitry mechanisms to establish the balance between GABAergic and glutamatergic signaling have not been fully elucidated. Caenorhabditis elegans DVB is an excitatory GABAergic motoneuron that drives the expulsion step in the defecation motor program. We show here that in addition to UNC-47, the vesicular GABA transporter, DVB also expresses EAT-4, a vesicular glutamate transporter. UBR-1, a conserved ubiquitin ligase, regulates DVB activity by suppressing a bidirectional inhibitory glutamate signaling. Loss of UBR-1 impairs DVB Ca2+ activity and expulsion frequency. These impairments are fully compensated by the knockdown of EAT-4 in DVB. Further, glutamate-gated chloride channels GLC-3 and GLC-2/4 receive DVB's glutamate signals to inhibit DVB and enteric muscle activity, respectively. These results implicate an intrinsic cellular mechanism that promotes the inherent asymmetric neural activity. We propose that elevated glutamate in ubr-1 mutants, being the cause of the E/I shift, potentially contributes to Johanson Blizzard syndrome.


Subject(s)
Caenorhabditis elegans Proteins , Animals , Caenorhabditis elegans Proteins/genetics , Ligases , Caenorhabditis elegans/genetics , Glutamic Acid , Neurotransmitter Agents , Ubiquitins
8.
Virology ; 587: 109879, 2023 10.
Article in English | MEDLINE | ID: mdl-37677987

ABSTRACT

Porcine enteric alphacoronavirus (PEAV) is a newly emerging swine enteropathogen that poses a threat to the swine industry. To understand the PEAV genome evolution, we performed a comprehensive analysis of the codon usage patterns in fifty-nine PEAV strains currently available. Phylogenetic analysis showed that PEAV can be divided into six lineages. Effective number of codons analysis demonstrated that the PEAV genome exhibits a low codon usage bias (CUB). Nucleotide composition analysis indicated that the PEAV genome has the most abundant nucleotide U content, with GC content (39.37% ± 0.08%) much lower than AU content (60.63% ± 0.08%). Neutrality and effective number of codons plot analyses suggested that natural selection rather than mutation pressure dominates the CUB of PEAV. Host adaptation analysis revealed that PEAV fits the codon usage pattern of non-human primates, humans and mice better than that of pigs. Our data enriches information on PEAV evolution, host adaptability, and cross-species transmission.


Subject(s)
Alphacoronavirus , Codon Usage , Animals , Swine , Mice , Phylogeny , Codon , Alphacoronavirus/genetics , Selection, Genetic , Nucleotides , Evolution, Molecular
9.
One Health Adv ; 1(1): 3, 2023.
Article in English | MEDLINE | ID: mdl-37521530

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is an economically important pathogen for the global pork industry. Although modified live virus (MLV) vaccines are commonly used for PRRSV prevention and control,  they still carry a risk of infecting the host and replicating in target cells, thereby increasing the likehood of virus recombination and reversion to virulence. In this study, we inserted the target sequence of miR-142 into the nsp2 hypervariable region of PRRSV to inhibit viral replication in its host cells of pigs, with the aim of achieving virus attenuation. The chimeric virus RvJX-miR-142t was successfully rescued and retained its growth characteristics in MARC-145 cells. Furthermore, it did not replicate in MARC-145 cells transfected with miRNA-142 mimic. We also observed limited replication ability of RvJX-miR-142t in pulmonary alveolar macrophages, which are the main cell types that PRRSV infects. Our animal inoculation study showed that pigs infected with RvJX-miR-142t displayed less severe clinical symptoms, lower viremia titers, lighter lung lesions, and significantly lower mortality rates during the first 7 days post-inoculation, in comparison to pigs infected with the backbone virus RvJXwn. We detected a partially deletion of the miR-142 target sequence in the RvJX-miR-142t genome at 14 dpi. It is highly possible that the reversion of viral virulence observed in the later timepoints of our animal experiment was caused by that. Our study provided a new strategy for attenuating PRRSV and confirmed its effectiveness. However, further studies are necessary to increase the stability of this virus under host selection pressure.

10.
Vet Microbiol ; 284: 109825, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37453262

ABSTRACT

PRRSV and CSFV are both common infectious pathogens in porcine populations, posing significant threats to the healthy development of the porcine industry. Vaccine immunization is the main way to prevent and control these two diseases. Increasing studies have demonstrated that there is an interaction between PRRSV co-infection and CSFV vaccine immune failure. To investigate the effect of PRRSV infection on CSFV proliferation and its molecular mechanism, the proliferation dynamics of PRRSV/CSFV, the NLRP3 inflammasome components, and IL-1ß expression levels were detected in PRRSV/CSFV alone- or co-infection. Subsequently, the relationship between inflammasome activation, IL-1ß expression, and CSFV proliferation was analyzed through the construction of an inflammasome activation model, specific siRNA interference, and specific inhibitor treatment. The results showed that CSFV infection had a poor regulatory effect on NLRP3 inflammasome activation and IL-1ß maturation, but PRRSV and CSFV co-infection could significantly up-regulate the expression of NLRP3 and ASC, induce Caspase-1 activation, and promote IL-1ß maturation. It was further determined that NLRP3 inflammasome components played important roles in IL-1ß maturation and inhibiting CSFV proliferation by PRRSV. Additional experiments indicated that PRRSV replication is essential for NLRP3 inflammasome activation, IL-1ß maturation, and CSFV proliferation inhibition. More importantly, NLRP3 inflammasome activation is regulated by the TLR4-MyD88-NF-κB pathways. In conclusion, PRRSV infection induced IL-1ß maturation by activating the NLRP3 inflammasome through the TLR4-MyD88-NF-κB pathways and then inhibited the proliferation of CSFV. These data further improved the theoretical basis for PRRSV inducing inflammatory factors and leading to the failure of CSFV immunization.


Subject(s)
Coinfection , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Vaccines , Swine , Animals , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , NF-kappa B/metabolism , Signal Transduction , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptor 4 , Coinfection/veterinary , Cell Proliferation , Interleukin-1beta/genetics
11.
Nanoscale ; 15(17): 7845-7853, 2023 May 04.
Article in English | MEDLINE | ID: mdl-37057392

ABSTRACT

Bidirectional optogenetic manipulation enables specific neural function dissection and animal behaviour regulation with high spatial-temporal resolution. It relies on the respective activation of two or more visible-light responsive optogenetic sensors, which inevitably induce signal crosstalk due to their spectral overlap, low photoactivation efficiency and potentially high biotoxicity. Herein, a strategy that combines dual-NIR-excited orthogonal emissive upconversion nanoparticles (OUCNPs) with a single dual-colour sensor, BiPOLES, is demonstrated to achieve bidirectional, crosstalk-free NIR manipulation of motor behaviour in vivo. Core@shell-structured OUCNPs with Tm3+ and Er3+ dopants in isolated layers exhibit orthogonal blue and red emissions in response to excitation at 808 and 980 nm, respectively. The OUCNPs subsequently activate BiPOLES-expressing excitatory cholinergic motor neurons in C. elegans, leading to significant inhibition and excitation of motor neurons and body bends, respectively. Importantly, these OUCNPs exhibit negligible toxicity toward neural development, motor function and reproduction. Such an OUCNP-BiPOLES system not only greatly facilitates independent, bidirectional NIR activation of a specific neuronal population and functional dissection, but also greatly simplifies the bidirectional NIR optogenetics toolset, thus endowing it with great potential for flexible upconversion optogenetic manipulation.


Subject(s)
Caenorhabditis elegans , Nanoparticles , Animals , Infrared Rays , Motor Neurons , Optogenetics
12.
J Virol ; 97(3): e0168922, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36916907

ABSTRACT

Fast evolution in the field of the replicase nsp2 represents a most prominent feature of porcine reproductive and respiratory syndrome virus (PRRSV). Here, we determined its biological significance in viral pathogenesis by constructing interlineage chimeric mutants between the Chinese highly pathogenic PRRSV (HP-PRRSV) strain JXwn06 (lineage 8) and the low-virulent NADC30-like strain CHsx1401 (lineage 1). Replacement with nsp2 from JXwn06 was surprisingly lethal to the backbone virus CHsx1401, but combined substitution with the structural protein-coding region (SP) gave rise to viable virus CHsx1401-SPnsp2JX. Meanwhile, a derivative carrying only the SP region (CHsx1401-SPJX) served as a control. Subsequent animal experiments revealed that acquisition of SP alone (CHsx1401-SPJX) did not allow CHsx1401 to gain much virulence, but additional swapping of HP-PRRSV nsp2 (CHsx1401-SPnsp2JX) enabled CHsx1401 to acquire some properties of HP-PRRSV, exemplified by prolonged high fever, microscopic lung hemorrhage, and a significant increase in proinflammatory cytokines in the acute stage. Consistent with this was the transcriptomic analysis of persistently infected secondary lymphoid tissues that revealed a much stronger induction of host cellular immune responses in this group and identified several core immune genes (e.g., TLR4, IL-1ß, MPO, etc.) regulated by HP-PRRSV nsp2. Interestingly, immune activation status in the individual groups correlated well with the rate of viremia clearance and viral tissue load reduction. Overall, the above results suggest that the Chinese HP-PRRSV nsp2 is a critical virulence regulator and highlight the importance of nsp2 genetic variation in modulating PRRSV virulence and persistence via immune modulation. IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) has been a major threat to the world swine industry. In the field, rapid genetic variations (e.g., deletion, mutation, recombination, etc.) within the nsp2 region present an intriguing conundrum to PRRSV biology and pathogenesis. By making chimeric mutants, here, we show that the Chinese highly pathogenic PRRSV (HP-PRRSV) nsp2 is a virulence factor and a much stronger inducer of host immune responses (e.g., inflammation) than its counterpart, currently epidemic, NADC30-like strains. Differences in the ability to modulate host immunity provide insight into the mechanisms of why NADC30-like strains and their derivatives are rising to be the dominant viruses, whereas the Chinese HP-PRRSV strains gradually give away center stage in the field. Our results have important implications in understanding PRRSV evolution, interlineage recombination, and persistence.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Animals , China/epidemiology , Cytokines , Genetic Variation , Genome, Viral , Phylogeny , Porcine respiratory and reproductive syndrome virus/genetics , Swine , Virulence/genetics
13.
Microbiol Spectr ; 10(6): e0308422, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36219105

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is a globally important disease threatening the pork industry, and modified live-virus (MLV) vaccines are widely used for its prevention. However, PRRS MLV shows high potential for reversion to virulence, leading to a major concern about its safety. Yet the revertant mechanism is still poorly understood. Here, attenuated virus JXwn06-P80, derived from the highly pathogenic PRRS virus (PRRSV) strain JXwn06 by serial passaging in MARC-145 cells, was reversely passaged in pigs through intranasal inoculation to mimic natural infection for 13 rounds, and the pathogenicity of viruses at the 3rd, 5th, 9th, 10th, and 11th passages was evaluated in pigs. From the 9th passage, the viruses caused mortality, which was related to their increased adaptability and replication efficiency (100 times higher than those of JXwn06-P80) in porcine alveolar macrophage (PAM) target cells. Similarly, JXwn06-P80 could also regain fatal virulence through reverse passage in PAMs for 25 or more passages, indicating that the increased adaptability in PAMs directly contributes to its regained fatal virulence. Next, the full-genome sequences were analyzed to explore the genetic evolutionary processes during adaptation both in vivo and in vitro. Finally, by a reverse genetic operation, four reverse mutation sites, NSP12-W121R, ORF2b (open reading frame 2b)-H9D, ORF5-H15L, and ORF5-V189L, were finally identified to partially contribute to the ability of the virus to adapt to PAMs, which may be related to virulence reversion during reverse passage. These findings provided direct scientific evidence for the virulence reversion of PRRS MLV and provided valuable clues for exploring its molecular mechanism. IMPORTANCE Reversion to virulence of a live attenuated vaccine is a public concern; however, direct scientific evidence is limited, and the mechanism is still poorly understood. Here, we present direct evidence for the reversion to virulence of PRRS MLV after serial passaging in pigs or target cells and found a correlation between virulence reversion and increased replication fitness in primary PAMs. The genetic evolutionary process during adaptation will provide valuable clues for exploring the molecular mechanism of PRRS MLV virulence reversion and offer important implications for understanding the reversion mechanisms of other vaccines.


Subject(s)
Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Viral Vaccines , Swine , Animals , Porcine respiratory and reproductive syndrome virus/genetics , Virulence/genetics , Macrophages, Alveolar , Mutation , Viral Vaccines/genetics
14.
Viruses ; 14(8)2022 08 18.
Article in English | MEDLINE | ID: mdl-36016437

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteropathogen which mainly causes diarrhea, dehydration and death in nursing piglets, threatening the global swine industry. Moreover, it can infect multiple animal species and humans. Hence, reliable diagnostic assays are needed to better control this zoonotic pathogen. Here, a blocking ELISA was developed using a recombinant nucleocapsid (N) protein as the coating antigen paired with an N-specific monoclonal antibody (mAb) as the detection antibody. The percent inhibition (PI) of the ELISA was determined using 384 swine serum samples, with an indirect immunofluorescence assay (IFA) as the reference method. Through receiver operating characteristic analysis in conjunction with Youden's index, the optimal PI cut-off value was determined to be 51.65%, which corresponded to a diagnostic sensitivity of 98.79% and a diagnostic specificity of 100%. Of the 330 serum samples tested positive via IFA, 326 and 4 were tested positive and negative via the ELISA, respectively, while the 54 serum samples tested negative via IFA were all negative via the ELISA. The overall coincidence rate between the two assays was 98.96% (380/384). The ELISA exhibited good repeatability and did not cross-react with antisera against other swine pathogens. Overall, this is the first report on developing a blocking ELISA for PDCoV serodiagnosis.


Subject(s)
Coronavirus Infections , Swine Diseases , Animals , Antibodies, Viral , Coronavirus Infections/diagnosis , Coronavirus Infections/veterinary , Deltacoronavirus , Enzyme-Linked Immunosorbent Assay/methods , Humans , Nucleocapsid Proteins , Swine
15.
Virulence ; 13(1): 1471-1485, 2022 12.
Article in English | MEDLINE | ID: mdl-36005235

ABSTRACT

Porcine deltacoronavirus (PDCoV) is an emerging enteropathogen causing severe diarrhoea, dehydration, and death in nursing piglets and enormous economic losses for the global swine industry. Furthermore, it can infect multiple animal species including humans. Therefore, a rapid, definitive diagnostic assay is required for the effective control of this zoonotic pathogen. To identify PDCoV, we developed a nucleic acid detection assay combining reverse transcription recombinase-aided amplification (RT-RAA) with a lateral flow dipstick (LFD) targeting the highly conserved genomic region in the ORF1b gene. The RT-RAA-LFD assay exhibited good PDCoV detection reproducibility and repeatability and could be completed within 11 min. Ten minutes at 40 °C was required for nucleic acid amplification and 1 min at room temperature was needed for the visual LFD readout. The assay specifically detected PDCoV and did not cross-react with any other major swine pathogens. The 95% limit of detection (LOD) was 3.97 median tissue culture infectious dose PDCoV RNA per reaction. This performance was comparable to that of a reference TaqMan-based real-time RT-PCR (trRT-PCR) assay for PDCoV. Of 149 swine small intestine, rectal swab, and serum samples, 71 and 75 tested positive for PDCoV according to RT-RAA-LFD and trRT-PCR, respectively. The diagnostic coincidence rate for both assays was 97.32% (145/149) and the kappa value was 0.946 (p < 0.001). Overall, the RT-RAA-LFD assay is a user-friendly diagnostic tool that can rapidly and visually detect PDCoV.


Subject(s)
Nucleic Acids , Recombinases , Animals , Deltacoronavirus , Humans , Nucleic Acid Amplification Techniques , Recombinases/genetics , Recombinases/metabolism , Reproducibility of Results , Reverse Transcription , Sensitivity and Specificity , Swine
16.
Vet Microbiol ; 273: 109513, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35952491

ABSTRACT

PRRSV and CSFV are both the main pathogens of pigs and pose great threats to the pig industry. Previous studies have shown that PRRSV infection or attenuated virus vaccination can reduce the antibody level of attenuated CSFV vaccine and even cause immune failure. The higher pro-inflammatory cytokines induced by PRRSV might play a significant role in inhibiting the proliferation of CSFV-C. However, the molecular mechanism has not been elucidated yet. Here, the effect of IL-1ß, a central mediator of immune-regulating inflammatory responses, on CSFV-C proliferation was investigated, as well as the mechanisms responsible for the production of IL-1ß in the PRRSV and CSFV-C co-infection systems. The results showed that co-infection could significantly increase IL-1ß production both at mRNA and protein levels with the infection progressing, and the IL-1ß upregulation was mainly triggered by PRRSV infection. Additional experiments indicated that IL-1ß inhibited the proliferation of CSFV-C in a cell-type independent manner at the replication and release stages. Furthermore, the IL-1ß production induced via the TLR4/MyD88 pathway and the downstream signaling pathways NF-κB, ERK1/2, P38, and JNK were involved by treatment with specific inhibitors or siRNA knockdown assays. Finally, we clarified that the NLRP3 inflammasome played a meaningful role in the maturation and release of IL-1ß. Together, the accumulated results provided a deeper understanding of the vaccination failure of CSFV caused by PRRSV co-infection as well as targets for the development of novel approaches for the vaccination and control of CSF.


Subject(s)
Coinfection , Porcine respiratory and reproductive syndrome virus , Swine Diseases , Animals , Cell Proliferation , Coinfection/veterinary , Inflammasomes/genetics , Interleukin-1beta/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Porcine respiratory and reproductive syndrome virus/metabolism , Swine , Toll-Like Receptor 4/genetics
17.
Virus Res ; 319: 198854, 2022 Oct 02.
Article in English | MEDLINE | ID: mdl-35788015

ABSTRACT

It is a common phenomenon that PRRSV infection can interfere with the protective efficacy of the CSFV vaccine in clinical settings, and no effective treatment is available. In our previous study, we found that PRRSV infection could inhibit the replication of CSFV-C by promoting the high expression of inflammatory cytokines. In order to further investigate whether Chinese medicine could alleviate the inhibition effect, the PAM39 cells model, which was co-infected with PRRSV and CSFV-C, was established. The effects of Chinese medicine on this co-infection model, as well as the effect of astragalus polysaccharide on the TLRs/NF-κB/TNF-α pathways, were investigated. Our results demonstrated that PAM39 cells inoculated with different pathogenic PRRSV significantly inhibited the replication of CSFV-C and up-regulated the major inflammatory mediators, including TNF-α. For the following studies, 50 µM of astragalus polysaccharide was selected from six kinds of representative Chinese medicine based on their cytotoxicity, viral titers, and inflammatory mediators. Further experiments indicated that astragalus polysaccharide could alleviate the inhibition of CSFV-C replication in the co-infection group with no influence on cell viability. In addition, astragalus polysaccharide treatment clearly reduced P65 phosphorylation and down-regulated the expression of TLR7, TLR9, and TNF-α in co-infection group, implying that the TLRs/NF-κB/TNF-α pathways may play an important role in astragalus polysaccharide's anti-inflammatory response. In conclusion, astragalus polysaccharide treatment alleviated PRRSV-mediated inhibition of CSFV-C replication via the TLRs/NF-κB/TNF-α pathways, and the molecular mechanism of PRRSV co-infection leading to the failure of CSFV vaccine immunization was partially elucidated, providing a scientific basis for effective CSF prevention and control in pig farms.


Subject(s)
Classical Swine Fever Virus , Coinfection , Porcine respiratory and reproductive syndrome virus , Animals , Inflammation Mediators , NF-kappa B/metabolism , Polysaccharides/pharmacology , Porcine respiratory and reproductive syndrome virus/metabolism , Swine , Tumor Necrosis Factor-alpha/genetics
18.
Proc Natl Acad Sci U S A ; 119(29): e2201169119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858300

ABSTRACT

Protein kinase R (PKR) is a critical host restriction factor against invading viral pathogens. However, this molecule is inactivated in the cells infected with porcine reproductive and respiratory syndrome virus (PRRSV), an economically devastating pathogen to the world swine industry. Here, we report that this event is to suppress cellular inflammation and is mediated by the viral replicase protein nsp1ß. We show that nsp1ß is a stress-responsive protein, enters virus-induced stress granules (SGs) during infection, and repurposes SGs into a proviral platform, where it co-opts the SG core component G3BP1 to interact with PKR in a regulated manner. RNA interference silencing of G3BP1 or mutation of specific nsp1ß residues (VS19GG) can abolish the antagonization of PKR activation. The viral mutant carrying the corresponding mutations induces elevated level of PKR phosphorylation and pronounced production of inflammatory cytokines (e.g., tumor necrosis factor-α, interleukin [IL]-6, and IL-8), whereas small-interfering RNA knockdown of PKR or treatment with C16, a PKR inhibitor, blocks this effect. Thus, PRRSV has evolved a unique strategy to evade PKR restriction to suppress host inflammatory responses.


Subject(s)
Antiviral Restriction Factors , DNA Helicases , Immune Evasion , Poly-ADP-Ribose Binding Proteins , Porcine respiratory and reproductive syndrome virus , RNA Helicases , RNA Recognition Motif Proteins , Stress Granules , Viral Nonstructural Proteins , eIF-2 Kinase , Animals , Antiviral Restriction Factors/metabolism , DNA Helicases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Stress Granules/virology , Swine , Viral Nonstructural Proteins/metabolism , Virus Replication , eIF-2 Kinase/metabolism
19.
Animals (Basel) ; 12(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35739924

ABSTRACT

Porcine circovirus 3 (PCV3) is an emerging virus, causing substantial economic losses in pig populations, that was first detected in 2016. Furthermore, the virus has already been reported in Europe, the Americas, and Asia, including China, indicating that the virus has spread worldwide. However, the molecular epidemiology of PCV3 still needs further study. To investigate PCV3 epidemiological characteristics in China, 2707 serum samples of pigs were randomly collected from 17 provinces in China between September 2018 and March 2022 and analyzed via PCR assays. The study showed that PCV3 infection was prevalent in the overall population with 31.07% (841/2707) and 100.0% (17/17) at sample and province levels, respectively, though the positivity rate of PCV3 varied from 7.41 to 70.0% in different provinces, suggesting that PCV3 infection has a widespread distribution in China. We selected 22 serum samples from different regions that had high levels of viral DNA for amplification and sequenced their ORF2 (Cap) gene. According to the phylogenetic analysis, all isolates in the current study could be grouped into two separate subclades, with 15 strains belonging to clade 3a and 7 strains belonging to clade 3b, indicating that PCV3a and PCV3b were the predominant subtypes in the regions of China studied. Meanwhile, additional analysis revealed that the capsid gene sequences identified in this study displayed 97.46~99.8% nucleotide (nt) and 97.06~100% amino acid (aa) sequence similarity with other PCV3 available reference strains, respectively. In general, our studies provide important insights for understanding the prevalence and evolution of PCV3 in China and will guide future efforts to develop measures for preventing and controlling the disease.

20.
Crit Rev Eukaryot Gene Expr ; 32(3): 83-93, 2022.
Article in English | MEDLINE | ID: mdl-35695612

ABSTRACT

MicroRNAs (miRNAs) act as critical biological factors in gastric cancer (GC). miR-1285 has been ascertained as a crucial antioncogene in some cancers. However, the effect of miR-1285 in GC and the regulatory mechanism are not clear. In this study, we revealed that miR-1285 expression was significantly reduced in GC. Overexpressing miR-1285 restrained GC cell multiplication and accelerated apoptosis, whereas suppressing miR-1285 facilitated cell growth and restrained apoptosis. The level of miR-1285 was negatively related to the RAB1A level in GC tissue specimens. RAB1A was verified by reporter gene assay as a target of miR-1285. Overexpression of miR-1285 suppressed the RAB1A level, whereas suppression of miR-1285 promoted the level of RAB1A expression. Knockdown of RAB1A resulted in analogical biological effect as that caused by overexpressing miR-1285. Moreover, both miR-1285 overexpression and RAB1A knockdown led to suppression of the mTOR/S6K1 pathway. By contrast, inhibition of miR-1285 promoted the mTOR/S6K1 pathway. In addition, miR-1285 also regulated the Bcl-2/Bax pathway. Taken together, our data indicate that miR-1285 suppresses GC cell multiplication by restraining the mTOR/S6K1 pathway and induces cell apoptosis by regulating the Bcl-2/Bax pathway via modulating RAB1A.


Subject(s)
MicroRNAs , Stomach Neoplasms , Apoptosis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...