151
|
Rani R, Nooruzzaman M, Caserta LC, Diel DG. Naturally occurring N-terminal mutations in SARS-CoV-2 nsp1 impact innate immune modulation but do not affect virus virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624996. [PMID: 39651192 PMCID: PMC11623490 DOI: 10.1101/2024.11.23.624996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The non-structural protein 1 (nsp1) of SARS-CoV-2 plays a key role in host innate immune evasion. We identified two deletion variants (Δ82-85 and Δ83-86) in the N-terminal region of the nsp1 of a SARS-CoV-2 BA.5.2.1 variant recovered from a human patient. Analysis of the sequence databases revealed a frequency of 0.5% of these mutations amongst available SARS-CoV-2 sequences. Structural analysis of the deletion mutant nsp1Δ82-85 and nsp1Δ83-86 revealed a distortion in the protein pocket when compared to the wild-type nsp1 which may affect protein function. To evaluate the functional relevance of these mutations, we cloned the mutant BA.5.2.1 nsp1Δ82-85 and nsp1Δ83-86 and wild-type nsp1 proteins in expression plasmids and performed luciferase reporter-based assays to assess activation of the interferon and nuclear factor kappa B (NF-κB) signalling pathways. Both nsp1Δ82-85 and nsp1Δ83-86 mutants showed marked decreased ability to inhibit the interferon beta (IFN-β) and NF-κB pathway activation. To assess the relevance of these deletions in the context of SARS-CoV-2 infection, we generated recombinant viruses carrying the wild type BA.5.2.1 nsp1 or the BA.5.2.1 nsp1Δ82-85 and nsp1Δ83-86 deletions in the backbone of WA1 strain. In vitro characterization of the recombinant SARS-CoV-2 viruses revealed that the recombinant viruses containing the nsp1Δ82-85 and nsp1Δ83-86 deletions presented similar plaque size and morphology to those produced by the wild-type rWA1-BA.5.2.1-nsp1 virus, indicating a similar ability of the mutant viruses to spread from cell to cell. Importantly, pathogenesis studies revealed that these mutations did not affect virus virulence and pathogenesis in a hamster model of SARS-CoV-2 infection.
Collapse
|
152
|
Yu J, Ge S, Li J, Zhang Y, Xu J, Wang Y, Liu S, Yu X, Wang Z. Interaction between coronaviruses and the autophagic response. Front Cell Infect Microbiol 2024; 14:1457617. [PMID: 39650836 PMCID: PMC11621220 DOI: 10.3389/fcimb.2024.1457617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/18/2024] [Indexed: 12/11/2024] Open
Abstract
In recent years, the emergence and widespread dissemination of the coronavirus SARS-CoV-2 has posed a significant threat to global public health and social development. In order to safely and effectively prevent and control the spread of coronavirus diseases, a profound understanding of virus-host interactions is paramount. Cellular autophagy, a process that safeguards cells by maintaining cellular homeostasis under diverse stress conditions. Xenophagy, specifically, can selectively degrade intracellular pathogens, such as bacteria, fungi, viruses, and parasites, thus establishing a robust defense mechanism against such intruders. Coronaviruses have the ability to induce autophagy, and they manipulate this pathway to ensure their efficient replication. While progress has been made in elucidating the intricate relationship between coronaviruses and autophagy, a comprehensive summary of how autophagy either benefits or hinders viral replication remains elusive. In this review, we delve into the mechanisms that govern how different coronaviruses regulate autophagy. We also provide an in-depth analysis of virus-host interactions, particularly focusing on the latest data pertaining to SARS-CoV-2. Our aim is to lay a theoretical foundation for the development of novel coronavirus vaccines and the screening of potential drug targets.
Collapse
Affiliation(s)
- Jiarong Yu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shengqiang Ge
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Jinming Li
- China Animal Health and Epidemiology Center, Qingdao, China
| | | | - Jiao Xu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Yingli Wang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Shan Liu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Xiaojing Yu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Zhiliang Wang
- China Animal Health and Epidemiology Center, Qingdao, China
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
153
|
Bendall EE, Dimcheff D, Papalambros L, Fitzsimmons WJ, Zhu Y, Schmitz J, Halasa N, Chappell J, Martin ET, Biddle JE, Smith-Jeffcoat SE, Rolfes MA, Mellis A, Talbot HK, Grijalva C, Lauring AS. In depth sequencing of a serially sampled household cohort reveals the within-host dynamics of Omicron SARS-CoV-2 and rare selection of novel spike variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624722. [PMID: 39605326 PMCID: PMC11601520 DOI: 10.1101/2024.11.21.624722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
SARS-CoV-2 has undergone repeated and rapid evolution to circumvent host immunity. However, outside of prolonged infections in immunocompromised hosts, within-host positive selection has rarely been detected. The low diversity within-hosts and strong genetic linkage among genomic sites make accurately detecting positive selection difficult. Longitudinal sampling is a powerful method for detecting selection that has seldom been used for SARS-CoV-2. Here we combine longitudinal sampling with replicate sequencing to increase the accuracy of and lower the threshold for variant calling. We sequenced 577 specimens from 105 individuals from a household cohort primarily during the BA.1/BA.2 variant period. There was extremely low diversity and a low rate of divergence. Specimens had 0-12 intrahost single nucleotide variants (iSNV) at >0.5% frequency, and the majority of the iSNV were at frequencies <2%. Within-host dynamics were dominated by genetic drift and purifying selection. Positive selection was rare but highly concentrated in spike. Two individuals with BA.1 infections had S:371F, a lineage defining substitution for BA.2. A Wright Fisher Approximate Bayesian Computational model identified positive selection at 14 loci with 7 in spike, including S:448 and S:339. We also detected significant genetic hitchhiking between synonymous changes and nonsynonymous iSNV under selection. The detectable immune-mediated selection may be caused by the relatively narrow antibody repertoire in individuals during the early Omicron phase of the SARS-CoV-2 pandemic. As both the virus and population immunity evolve, understanding the corresponding shifts in SARS-CoV-2 within-host dynamics will be important.
Collapse
Affiliation(s)
- Emily E. Bendall
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Derek Dimcheff
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leigh Papalambros
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Yuwei Zhu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan Schmitz
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Natasha Halasa
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Chappell
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | - H. Keipp Talbot
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carlos Grijalva
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam S. Lauring
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
154
|
Zhao W, Gao Z, Guo C, Zhang Y, Zhang Y, Wang Q, Yu J. A dual typing system establishment and global diversity analysis for sapoviruses. BMC Genomics 2024; 25:1131. [PMID: 39578768 PMCID: PMC11583745 DOI: 10.1186/s12864-024-11048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The genus Sapovirus in the family Caliciviridae comprises of a genetically diverse group of viruses that are responsible for causing acute gastroenteritis in both human and animals globally. As the number of sequences continues to grow and more recombinant sequences are identified, the classification criteria of genogroups and genotypes of sapovirus need to be further refined. In this study, we aimed to optimize the classification of sapoviruses. RESULTS Through evolutionary clustering and genetic distance analysis, we have updated the classification criteria for VP1 genogroup and genotypes. We adjusted the original mean values ± 3 standard deviations (SD) of genetic distances to mean values ± 2.5SD, resulting the corresponding cutoff values for the same genotype and genogroup set at <0.161 and <0.503, respectively. Additionally, we established classification criteria for RdRp types and groups, referred to as P-types and P-groups,, with mean values ± 2SD and cutoff values of <0.266 and <0.531 for the same type and group, respectively. This refinement has expanded the VP1 genogroups to thirty-four and identified twenty-four P-groups. For human sapoviruses, the new criteria have resulted in the addition of one genotype, GV.PNA1. Moreover, the new criteria defined three P-groups and 21 P-types for human sapoviruses. Spatial-temporal analysis revealed no specific distribution pattern for human sapoviruses. CONCLUSIONS We established a dual typing system on classification based on VP1 and RdRp nucleotide sequences for sapoviruses.
Collapse
Affiliation(s)
- Wei Zhao
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Zhiyong Gao
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China
| | - Chiyu Guo
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Yuyue Zhang
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Yu Zhang
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Quanyi Wang
- Institute for Infectious Disease and Endemic Disease Control, Beijing Center for Disease Prevention and Control, Beijing, China.
| | - Jiemei Yu
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China.
| |
Collapse
|
155
|
Battista F, Schultz CL. Sampling and collector biases as taphonomic filters: an overview. AN ACAD BRAS CIENC 2024; 96:e20231242. [PMID: 39607126 DOI: 10.1590/0001-3765202420231242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/11/2024] [Indexed: 11/29/2024] Open
Abstract
Sampling (or sample) bias is a widespread concern in scientific research, across several disciplines. The concept of sampling bias originated in statistical studies. The consequence of a biased sample is that scientists will conclude about a population different from their target. In paleontology, sampling bias is typically related to fieldwork context. Human factors, known as sullegic (e.g. collection method, historic resampling) and trephic (transport, and curatorial processes) factors can generate bias. Other factor is the ugly fossil syndrome (i.e. choosing based on completeness of the specimens, or according to the researcher interest). Thus, sampling implies information loss. Biased samples add artificial results and can be considered an additional taphonomic filter. Therefore, sampling bias and the collector role and choices are frequently linked and almost indistinguishable. Compared to the treatment of this topic in other research fields, little related discussion has been held in vertebrate paleontology, especially regarding what happens at the interface between the biosphere, lithosphere, and anthroposphere, and during the transition between the anthroposphere and the patrisphere (museums). Numerous questions still arise. As a community, we must pay attention, to minimize the loss of information, from field activities to cataloging.
Collapse
Affiliation(s)
- Francesco Battista
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Geociências, Instituto de Geociências, Av. Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil
| | - Cesar L Schultz
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Geociências, Instituto de Geociências, Av. Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil
| |
Collapse
|
156
|
Moutinho I, Henriques M, Cardoso S, da Penha Coutinho T, Penha-Gonçalves C, Demengeot J, Castanho M, Tavares L, Gil S, Nunes T, Aires-da-Silva F. SARS-CoV-2 Seroprevalence in Indoor House Cats From the Lisbon Area During the COVID-19 Pandemic, 2019-2021. Transbound Emerg Dis 2024; 2024:1543922. [PMID: 40303070 PMCID: PMC12020408 DOI: 10.1155/tbed/1543922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 05/02/2025]
Abstract
The susceptibility of various animal species to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been studied extensively. Cats have garnered significant concern due to their high susceptibility and proximity to humans. This study aimed to evaluate the susceptibility and antibody response in house cats exposed to SARS-CoV-2 when human infection was spreading in the Lisbon area during the 2019-2021 period. A total of 733 serum samples were collected and characterized from cats admitted to the Veterinary Teaching Hospital of the Faculty of Veterinary Medicine at the University of Lisbon (HEV-FMV-ULisboa). All samples were tested by enzyme-linked immunosorbent assay (ELISA) for immunoglobulin M (IgM) and immunoglobulin G (IgG) anti-SARS-CoV-2 whole Spike and receptor-binding domain (RBD) proteins from the Wuhan-Hu-1 isolate and 14.7% (108/733) tested positive, suggesting exposure to the human virus. Surrogate virus neutralization test (sVNT) against the Wuhan-Hu-1 isolate showed that 20.4% of ELISA positive samples (22/108) harbored neutralizing antibodies against the virus. The 22 most promising serum samples were retested using ELISA and sVNT against Alpha, Delta, and Omicron SARS-CoV-2 variants. Notably, these samples exhibited antibodies that were capable of recognizing and neutralizing these variants. Subsequent neutralization assays confirmed that the serum samples effectively inhibited the infection process of Wuhan-Hu-1 D614G, Delta, and Omicron SARS-CoV-2 pseudotyped viruses. Our findings indicate that cats were exposed to SARS-CoV-2 infection during the pandemic period and generated highly effective and broadly neutralizing antibodies against the virus. Although cats have not been demonstrated to significantly contribute to the spread of SARS-CoV-2, their high susceptibility to asymptomatic infection underscores the importance of investment in preventive surveillance measures. In summary, our study reinforces the notion that cats naturally infected with SARS-CoV-2 represent a valuable anthroponotic disease model in house settings and might be a potential source for the development of antibodies against SARS-CoV-2 in tackling future outbreaks with a One Heath perspective.
Collapse
Affiliation(s)
- Isa Moutinho
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Mafalda Henriques
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Sara Cardoso
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Teresa da Penha Coutinho
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | | | | | - Miguel Castanho
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Luís Tavares
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Solange Gil
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Telmo Nunes
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Frederico Aires-da-Silva
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| |
Collapse
|
157
|
Parrado R, Torrico Villarroel NG. Caracterización molecular de virus respiratorios en población pediátrica del Hospital Albina Patiño en Cochabamba, Bolivia. GACETA MÉDICA BOLIVIANA 2024; 47:68-79. [DOI: 10.47993/gmb.v47i2.820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Las infecciones respiratorias agudas son enfermedades frecuentes en todo el mundo, la mayoría producidas por virus. La población más afectada son niños menores de cinco años, quienes tienen mayor riesgo de padecer enfermedades respiratorias graves asociadas a mortalidad. En Cochabamba no existía información publicada sobre circulación de virus respiratorios antes de la pandemia por SARS-CoV-2/COVID-19. Objetivos: detectar y caracterizar virus respiratorios en pacientes pediátricos internados con infecciones respiratorias en el Hospital Pediátrico "Albina Patiño".
Métodos: el período de estudio fue entre septiembre de 2017 y agosto de 2018, habiéndose incluido una población de 202 pacientes menores de cinco años. Se utilizó la reacción en cadena de la polimerasa en tiempo real (qPCR) multiplex para detectar y caracterizar los virus respiratorios relacionados con infección respiratoria.
Resultados: El 61,4% de las muestras analizadas dieron positivo a virus respiratorios. Los resultados incluyeron: Adenovirus 1,5 %, Metapneumovirus 3,5 %, Influenza virus 8 %, Parainfluenza virus 9,9 % y VSR 35,7 %. El virus más común responsable de las infecciones respiratorias fue el VSR. El grupo de niños infectados más afectado son los menores de dos años. Las infecciones virales alcanzaron el rango epidémico completo entre mayo y julio de 2018.
Conclusiones: Se logró detectar y caracterizar virus respiratorios en pacientes pediátricos, se evidencia un patrón de circulación de estos virus respiratorios específico para cada población, incluso antes de la circulación de SARS-CoV-2. Es necesario tener en cuenta que los países vecinos con condiciones similares de climatología pueden tener un patrón de circulación diferente.
Collapse
|
158
|
Castillo AP, Miranda JVO, Fonseca PLC, Moreira RG, de Araújo E Santos LCG, Queiroz DC, Bonfim DM, Coelho CM, Lima PCS, Motta ROC, Tinoco HP, da Silveira JAG, Aguiar RS. SARS-CoV-2 surveillance in captive animals at the belo horizonte zoo, Minas Gerais, Brazil. Virol J 2024; 21:297. [PMID: 39563414 PMCID: PMC11575034 DOI: 10.1186/s12985-024-02505-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/16/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND The pandemic caused by SARS-CoV-2 has not only affected humans but also raised concerns about its transmission to wild animals, potentially creating natural reservoirs. Understanding these dynamics is critical for preventing future pandemics and developing control strategies. This study aims to investigate the presence of SARS-CoV-2 in wild mammals at the Belo Horizonte Zoo in Brazil, analyzing the virus's evolution and zoonotic potential. METHODS The study was conducted at the Belo Horizonte Zoo, Minas Gerais, Brazil, covering a diverse population of mammals. Oropharyngeal, rectal, and nasal swabs were collected from 47 captive animals between November 2021 and March 2023. SARS-CoV-2 presence was determined using RT-PCR, and positive samples were sequenced for phylogenetic analysis. Consensus genomes were classified using Pangolin and NextClade tools, and a maximum likelihood phylogeny was inferred using IQ-Tree. RESULTS Of the 47 animals tested, nine (19.1%) were positive for SARS-CoV-2. Positive samples included rectal, oropharyngeal, and nasal swabs, with the highest positivity in rectal samples. Three genomes were successfully sequenced, revealing two variants: VOC Alpha in a maned wolf (Chrysocyon brachyurus) and a fallow deer (Dama dama), and VOC Omicron in a western lowland gorilla (Gorilla gorilla gorilla). Phylogenetic analysis indicated potential human-to-animal transmission, with animal genomes clustering close to human samples from the same region. CONCLUSIONS This study highlights the presence of SARS-CoV-2 in various wild mammal species at the Belo Horizonte Zoo, emphasizing the virus's zoonotic potential and the complexity of interspecies transmission. The detection of different variants suggests ongoing viral evolution and adaptation in new hosts. Continuous monitoring and genomic surveillance of SARS-CoV-2 in wildlife are essential for understanding its transmission dynamics and preventing future zoonotic outbreaks. These findings underscore the need for integrated public health strategies that include wildlife monitoring to mitigate the risks posed by emerging infectious diseases.
Collapse
Affiliation(s)
- Anisleidy Pérez Castillo
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Laboratório de PROTOVET, Departamento de Medicina Veterinária Preventiva, Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João Victor Oliveira Miranda
- Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paula Luize Camargos Fonseca
- Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rennan Garcias Moreira
- Centro de Laboratórios Multiusuários, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiza Campos Guerra de Araújo E Santos
- Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Costa Queiroz
- Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego Menezes Bonfim
- Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlyle Mendes Coelho
- Fundação de Parques Municipais E Zoobotânica - FPMZB, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Herlandes Penha Tinoco
- Fundação de Parques Municipais E Zoobotânica - FPMZB, Belo Horizonte, Minas Gerais, Brazil
| | - Júlia Angélica Gonçalves da Silveira
- Laboratório de PROTOVET, Departamento de Medicina Veterinária Preventiva, Escola de Veterinária da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Renato Santana Aguiar
- Laboratório de Biologia Integrativa, Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
159
|
Pellegrini F, Lanave G, Caringella F, Diakoudi G, Salvaggiulo A, Cavalli A, Papaleo A, Di Martino B, Camero M, Bányai K, Matthijnssens J, Martella V. Identification of Recombinant Aichivirus D in Cattle, Italy. Animals (Basel) 2024; 14:3315. [PMID: 39595367 PMCID: PMC11591108 DOI: 10.3390/ani14223315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Kobuviruses (KoVs) are a group of small, non-enveloped RNA viruses classified in the genus Kobuvirus within the Picornaviridae family, comprising Aichivirus species A to F. KoVs have been identified in humans and several mammals, including domestic ungulates. This study investigated the presence of KoVs in a collection of bovine stool samples (n = 38) obtained from animals with enteritis or without clinical signs. By RT-PCR screening, KoV RNA was detected in 10/38 animals (26.3%). Six of the ten positive animals had enteric signs. On sequence analysis of the amplicons, eight strains were related to species Aichivirus B, commonly identified in cattle. In contrast, two strains (ITA/2019/572-1 and ITA/2020/bovine/30-2), displayed the highest nt identity (up to 97.1%) to cattle, yak, and goat Aichivirus D strains. On whole genome analysis, strains ITA/2019/572-1 and ITA/2020/30-2 showed 88.9% nt identity to each other and 87.8-90.3% nt to the bovine kobuvirus strain CHN/2021/ON730709 identified in China. Interestingly these three Aichivirus D strains showed a recombinant makeup, clustering with D1 genotype in the capsid region and with D2 genotype in the non-structural genes. These findings suggest that Aichivirus D KoVs are common components of livestock virome. Understanding the genetic diversity of KoVs in animals will be useful to improve the diagnostics and gather epidemiological data.
Collapse
Affiliation(s)
- Francesco Pellegrini
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Gianvito Lanave
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Francesca Caringella
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Georgia Diakoudi
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Anna Salvaggiulo
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Alessandra Cavalli
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Alessandro Papaleo
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Barbara Di Martino
- Department of Veterinary Medicine, Università degli Studi di Teramo, 64100 Teramo, Italy;
| | - Michele Camero
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
| | - Krisztián Bányai
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary;
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Jelle Matthijnssens
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium;
| | - Vito Martella
- Department of Veterinary Medicine, Università Aldo Moro di Bari, 70010 Valenzano, Italy; (F.P.); (G.L.); (F.C.); (G.D.); (A.S.); (A.C.); (A.P.); (M.C.)
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, 1078 Budapest, Hungary;
| |
Collapse
|
160
|
Lin HY, Tan QH. Metagenomic next-generation sequencing may assist diagnosis of osteomyelitis caused by Mycobacterium houstonense: A case report. World J Orthop 2024; 15:1095-1100. [PMID: 39600859 PMCID: PMC11586735 DOI: 10.5312/wjo.v15.i11.1095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/22/2024] [Accepted: 09/23/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Mycobacterium houstonense (M. houstonense) belongs to the nontuberculous mycobacterium group. Infection caused by M. houstonense is prone to recurrence. CASE SUMMARY We present a patient who was diagnosed with osteomyelitis caused by M. houstonense and treated with a combination of cefoxitin, and amikacin combined with linezolid. CONCLUSION The emergence of metagenomic next-generation sequencing (NGS) has brought new hope for the diagnosis and treatment of listeria meningitis. NGS can analyze a large number of nucleic acid sequences in a short time and quickly determine the pathogen species in the sample. Compared with traditional cerebrospinal fluid culture, NGS can greatly shorten the diagnosis time and provide strong support for the timely treatment of patients. Regarding treatment, NGS can also play an important role. Rapid and accurate diagnosis can enable patients to start targeted treatment as soon as possible and improve the treatment effect. At the same time, by monitoring the changes in pathogen resistance, the treatment plan can be adjusted in time to avoid treatment failure.
Collapse
Affiliation(s)
- Hong-Yin Lin
- Department of Infectious Disease, Shanghai Sixth People's Hospital, Shanghai 200233, China
| | - Quan-Hui Tan
- Department of Infectious Disease, Shanghai Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
161
|
Iyaniwura SA, Ribeiro RM, Zitzmann C, Phan T, Ke R, Perelson AS. The kinetics of SARS-CoV-2 infection based on a human challenge study. Proc Natl Acad Sci U S A 2024; 121:e2406303121. [PMID: 39508770 PMCID: PMC11573497 DOI: 10.1073/pnas.2406303121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Studying the early events that occur after viral infection in humans is difficult unless one intentionally infects volunteers in a human challenge study. Here, we use data about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in such a study in combination with mathematical modeling to gain insights into the relationship between the amount of virus in the upper respiratory tract and the immune response it generates. We propose a set of dynamic models of increasing complexity to dissect the roles of target cell limitation, innate immunity, and adaptive immunity in determining the observed viral kinetics. We introduce an approach for modeling the effect of humoral immunity that describes a decline in infectious virus after immune activation. We fit our models to viral load and infectious titer data from all the untreated infected participants in the study simultaneously. We found that a power-law with a power h < 1 describes the relationship between infectious virus and viral load. Viral replication at the early stage of infection is rapid, with a doubling time of ~2 h for viral RNA and ~3 h for infectious virus. We estimate that adaptive immunity is initiated ~7 to 10 d postinfection and appears to contribute to a multiphasic viral decline experienced by some participants; the viral rebound experienced by other participants is consistent with a decline in the interferon response. Altogether, we quantified the kinetics of SARS-CoV-2 infection, shedding light on the early dynamics of the virus and the potential role of innate and adaptive immunity in promoting viral decline during infection.
Collapse
Affiliation(s)
- Sarafa A Iyaniwura
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Ruy M Ribeiro
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Carolin Zitzmann
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Tin Phan
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Ruian Ke
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Alan S Perelson
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| |
Collapse
|
162
|
Byrne C, Schiffer JT. Ensemble modeling of SARS-CoV-2 immune dynamics in immunologically naïve rhesus macaques predicts that potent, early innate immune responses drive viral elimination. Front Immunol 2024; 15:1426016. [PMID: 39575237 PMCID: PMC11578959 DOI: 10.3389/fimmu.2024.1426016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction An unprecedented breadth of longitudinal viral and multi-scale immunological data has been gathered during SARS-CoV-2 infection. However, due to the high complexity, non-linearity, multi-dimensionality, mixed anatomic sampling, and possible autocorrelation of available immune data, it is challenging to identify the components of the innate and adaptive immune response that drive viral elimination. Novel mathematical models and analytical approaches are required to synthesize contemporaneously gathered cytokine, transcriptomic, flow cytometry, antibody response, and viral load data into a coherent story of viral control, and ultimately to discriminate drivers of mild versus severe infection. Methods We investigated a dataset describing innate, SARS-CoV-2 specific T cell, and antibody responses in the lung during early and late stages of infection in immunologically naïve rhesus macaques. We used multi-model inference and ensemble modeling approaches from ecology and weather forecasting to compare and combine various competing models. Results and discussion Model outputs suggest that the innate immune response plays a crucial role in controlling early infection, while SARS-CoV-2 specific CD4+ T cells correspond to later viral elimination, and anti-spike IgG antibodies do not impact viral dynamics. Among the numerous genes potentially contributing to the innate response, we identified IFI27 as most closely linked to viral load decline. A 90% knockdown of the innate response from our validated model resulted in a ~10-fold increase in peak viral load during infection. Our approach provides a novel methodological framework for future analyses of similar complex, non-linear multi-component immunologic data sets.
Collapse
Affiliation(s)
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center,
Seattle, WA, United States
| |
Collapse
|
163
|
Zhang R, Yin G, Wang Y, Li Y, Wang X, Bi J, Yang G, Qu K, Gao L. Whole-Genome Analysis of Porcine Epidemic Diarrhea Virus from Yunnan, China. Vet Sci 2024; 11:548. [PMID: 39591322 PMCID: PMC11599152 DOI: 10.3390/vetsci11110548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a member of the genus α-coronavirus and causes severe diarrhea in piglets, leading to enormous economic losses in the pig industry. To understand the epidemic variation of PEDV strains in Yunnan province, three PEDV strains (YN2021, YNLP 2022, and YNBS 2022) and one commercially available attenuated vaccine strain (Attenuated AJ1102-R) that were previously isolated were sequenced and compared with the representative PEDV strains. NJ phylogenetic analysis showed that YN2021 strain and Attenuated CV777 strain were clustered into GI-b subtype, while YNLP 2022 and YNBS 2022 belong to GII-b subtype, accompanying ZJCZ4 and Attenuated AJ1102-R. RDP analysis revealed that YNLP 2022 was a genome recombination from both GII-b strain PEDV-7C and GII-a strain YN1, of which the recombination region is in the range nt4994-7605. YNBS 2022 strain was another recombination originated from GII-b subtype strain 17GXZC-1ORF3c and GII-a subtype strain PEDV-CHZ, of which the counterpart is in the range nt16399-22326. The Yunnan strain of PEDV was analyzed for the first time from the whole-genome perspective, and comprehensive analysis showed that the Yunnan strains have high genetic variation. This study may shed new light on the current PEDV infections in Yunnan and pave the way toward further control of PEDV infections.
Collapse
Affiliation(s)
- Runting Zhang
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Gefen Yin
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Yunhua Wang
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Yongneng Li
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Xinxian Wang
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Junlong Bi
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Guishu Yang
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| | - Kaixing Qu
- Academy of Science and Technology, Chuxiong Normal University, Chuxiong 675000, China
| | - Libo Gao
- College of Animal Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (R.Z.); (G.Y.); (Y.W.); (Y.L.); (X.W.); (J.B.); (G.Y.)
| |
Collapse
|
164
|
Huang X, Liu G, Chang T, Yang Y, Wang T, Xia D, Qi X, Zhu X, Wei Z, Tian X, Wang H, Tian Z, Cai X, An T. Recombinant characterization and pathogenicity of a novel L1C RFLP-1-4-4 variant of porcine reproductive and respiratory syndrome virus in China. Vet Res 2024; 55:142. [PMID: 39506759 PMCID: PMC11539553 DOI: 10.1186/s13567-024-01401-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/10/2024] [Indexed: 11/08/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the most significant diseases affecting the pig industry worldwide and is caused by the PRRS virus (PRRSV), which has complex genetic variation due to frequent mutations, indels, and recombination. The emergence of PRRSV L1C.5 in 2020 in the United States has raised worldwide concerns about PRRSV with the RFLP 1-4-4 pattern and lineage 1C. However, studies on the pathogenic characteristics, epidemiological distribution, and effectiveness of vaccines against PRRSV with L1C and RFLP1-4-4 pattern in China are still insufficient. In this study, a novel recombinant variant of PRRSV with RFLP 1-4-4 and lineage 1C features, different from L1C.5 in the United States, was isolated in China in 2021. In pathogenicity experiments in specific pathogen-free piglets or farm piglets, 60-100% of artificially infected experimental piglets died with high fever and respiratory symptoms. Inflammatory cytokine and chemokine levels were upregulated in infected piglets. A commercially modified live vaccine against highly pathogenic PRRSV did not provide effective protection when the vaccinated piglets were challenged with the novel L1C-1-4-4 variant. Therefore, this strain merits special attention when devising control and vaccine strategies. These findings suggest that extensive joint surveillance is urgently needed and that vaccine strategies should be updated to prevent the disease from spreading further.
Collapse
Affiliation(s)
- Xinyi Huang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Guoqing Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Tong Chang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Yongbo Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Dasong Xia
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Xinyu Qi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Xulong Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Ziyi Wei
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Xiaoxiao Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Haiwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Zhijun Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Xuehui Cai
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Tongqing An
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
- Heilongjiang Provincial Key Laboratory of Veterinary Immunology, Harbin, 150069, China.
| |
Collapse
|
165
|
Yang K, Zhou J, Zhao J, Liu L, Hua C, Hong C, Wang M, Hu A, Zhang W, Cui J, Zhu C, Deng G, Liu Y, Zhu L. Mobile lab: A novel pathogen assay using the nucleic acid automatic assay system assisted by a self-contained microfluidic cassette and chitosan decorating magnetic particles. SENSORS AND ACTUATORS B: CHEMICAL 2024; 419:136413. [DOI: 10.1016/j.snb.2024.136413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
166
|
Schuh L, Markov PV, Voulgaridi I, Bogogiannidou Z, Mouchtouri VA, Hadjichristodoulou C, Stilianakis NI. Within-host dynamics of antiviral treatment with remdesivir for SARS-CoV-2 infection. J R Soc Interface 2024; 21:20240536. [PMID: 39592013 PMCID: PMC11597410 DOI: 10.1098/rsif.2024.0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The effectiveness of antiviral treatment with remdesivir against COVID-19 has been investigated in clinical trials suggesting earlier recovery. However, this effect seems to be rather modest. In this study, we tracked the clinical course of SARS-CoV-2 infections in 369 COVID-19 individuals across a spectrum of illness severities, including both untreated individuals and individuals who received antiviral treatment with remdesivir. Moreover, using a process-based mathematical model, we quantified and analysed the within-host infection dynamics of a total of 88 individuals, of which 69 were untreated and 19 antiviral-treated individuals. For untreated individuals, we found that those hospitalized exhibit lower levels of early immune response and higher cumulative viral loads than those who were not. For treated individuals, we found that those who died were on average hospitalized later after symptom onset than those who survived, underscoring the importance of early medical intervention for severe COVID-19. Finally, our model estimates a rather limited antiviral activity of remdesivir. Our results provide valuable insights into the clinical course of COVID-19 during antiviral treatment with remdesivir and suggest the need for alternative treatment regimens.
Collapse
Affiliation(s)
- Lea Schuh
- Joint Research Centre (JRC), European Commission, Ispra, Italy
| | - Peter V. Markov
- Joint Research Centre (JRC), European Commission, Ispra, Italy
- London School of Hygiene & Tropical Medicine, University of London, London, UK
| | - Ioanna Voulgaridi
- Laboratory of Hygiene and Epidemiology, University of Thessaly, Larissa, Greece
| | | | | | | | - Nikolaos I. Stilianakis
- Joint Research Centre (JRC), European Commission, Ispra, Italy
- Department of Biometry and Epidemiology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
167
|
Yang Y, Tan J, Wang F, Sun W, Shi H, Cheng Z, Xie Y, Zhou X. Preconcentration and detection of SARS-CoV-2 in wastewater: A comprehensive review. Biosens Bioelectron 2024; 263:116617. [PMID: 39094290 DOI: 10.1016/j.bios.2024.116617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2) causing coronavirus disease 2019 (COVID-19) affected the health of human beings and the global economy. The patients with SARS-CoV-2 infection had viral RNA or live infectious viruses in feces. Thus, the possible transmission of SARS-CoV-2 through wastewater received great attentions. Moreover, SARS-CoV-2 in wastewater can serve as an early indicator of the infection within communities. We summarized the preconcentration and detection technology of SARS-CoV-2 in wastewater aiming at the complex matrices of wastewater and low virus concentration and compared their performance characteristics. We described the emerging tests that would be possible to realize the rapid detection of SARS-CoV-2 in fields and encourage academics to advance their technologies beyond conception. We concluded with a brief discussion on the outlook for integrating preconcentration and the detection of SARS-CoV-2 with emerging technologies.
Collapse
Affiliation(s)
- Yihan Yang
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Jisui Tan
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Fan Wang
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Weiming Sun
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Hanchang Shi
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Zhao Cheng
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Yangcun Xie
- Chinese Academy of Environmental Planning, Beijing, 100043, China.
| | - Xiaohong Zhou
- School of Environment, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
168
|
Lopez Quezada L, Mba Medie F, Luu RJ, Gaibler RB, Gabriel EP, Rubio LD, Mulhern TJ, Marr EE, Borenstein JT, Fisher CR, Gard AL. Predicting Clinical Outcomes of SARS-CoV-2 Drug Efficacy with a High-Throughput Human Airway Microphysiological System. Adv Biol (Weinh) 2024; 8:e2300511. [PMID: 39123296 DOI: 10.1002/adbi.202300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/23/2023] [Indexed: 08/12/2024]
Abstract
The average cost to bring a new drug from its initial discovery to a patient's bedside is estimated to surpass $2 billion and requires over a decade of research and development. There is a need for new drug screening technologies that can parse drug candidates with increased likelihood of clinical utility early in development in order to increase the cost-effectiveness of this pipeline. For example, during the COVID-19 pandemic, resources were rapidly mobilized to identify effective therapeutic treatments but many lead antiviral compounds failed to demonstrate efficacy when progressed to human trials. To address the lack of predictive preclinical drug screening tools, PREDICT96-ALI, a high-throughput (n = 96) microphysiological system (MPS) that recapitulates primary human tracheobronchial tissue,is adapted for the evaluation of differential antiviral efficacy of native SARS-CoV-2 variants of concern. Here, PREDICT96-ALI resolves both the differential viral kinetics between variants and the efficacy of antiviral compounds over a range of drug doses. PREDICT96-ALI is able to distinguish clinically efficacious antiviral therapies like remdesivir and nirmatrelvir from promising lead compounds that do not show clinical efficacy. Importantly, results from this proof-of-concept study track with known clinical outcomes, demonstrate the feasibility of this technology as a prognostic drug discovery tool.
Collapse
Affiliation(s)
| | | | - Rebeccah J Luu
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | - Logan D Rubio
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | | | | | - Ashley L Gard
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
169
|
Shi J, Jia Q, Yu Y, Zhang J, Li Z, He W. IFN-β contributes to astrocyte activation in the brain following coronavirus PHEV infection independent on peripheral immunity. Vet Microbiol 2024; 298:110280. [PMID: 39442426 DOI: 10.1016/j.vetmic.2024.110280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
Porcine hemagglutinating encephalomyelitis (PHE), caused by a betacoronavirus named porcine hemagglutinating encephalomyelitis virus (PHEV), is a highly fatal disease of pigs characterized by nonsuppurative encephalitis. Activation of astrocytes is a hallmark of viral encephalomyelitis; however, the mechanism of PHEV-induced astrocyte activation is currently unknown. Based on mouse model, we show that PHEV infection led to astrogliosis in mouse brain and brain slice cultures (BSCs), as indicated by increased expression of glial fibrillary acidic protein (GFAP). PHEV can neither infect nor activate primary astrocytes in vitro, indicating that activation of astrocytes maybe mediated by factors secreted from viral infected neurons but not by direct viral infection of astrocytes. PHEV infection results in increased interferon (IFN) response in later stage, we thereafter focused on whether IFN-β can activate astrocytes after PHEV infection similar to other neurotropic viruses. IFN-β treatment resulted in both the upregulation of GFAP and activation-associated cytokines/chemokines in mouse primary astrocytes. Furthermore, the addition of IFN-β neutralization antibody prevented PHEV-infected mouse brain tissue homogenate from activating astrocytes. Taken together, IFN-β triggers the activation of astrocytes in the central nervous system (CNS) following PHEV infection. Further understanding of the role of activated astrocytes during PHEV infection may provide new insights for treatment this disease.
Collapse
Affiliation(s)
- Junchao Shi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Jilin University, Changchun 130061, China; Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qianhan Jia
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yanfang Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zi Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenqi He
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
170
|
Castañeda Montes FJ, Cerriteño Sánchez JL, Cuevas‐Romero JS, Castañeda Montes MA, Mendoza Elvira S. Development and Application of an Indirect Enzyme-Linked Immunosorbent Assay Based on a Recombinant Matrix Protein for the Serological Study of Porcine Deltacoronavirus in Mexican Pigs. Vet Med Sci 2024; 10:e70108. [PMID: 39494986 PMCID: PMC11533212 DOI: 10.1002/vms3.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/24/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an infectious disease that causes diarrhoea in pigs of different ages; however, piglets are more susceptible. PDCoV was first reported in 2012 in China and Hong Kong. Later, it was first reported in the USA in 2014 and in Mexico in 2019. Several studies have shown that M protein is highly conserved and, therefore, suitable for diagnostic systems. In this study, for the first time, an indirect enzyme-linked immunosorbent assay (iELISA) based on a recombinant M protein (rM-PDCoV) was developed to evaluate the seroprevalence of PDCoV in four states in Mexico. High sensitivity (83%) and specificity (100%) were observed for the iELISA. The kappa index calculated a nearly perfect agreement (0.8831) compared to the Western blot (gold standard test), suggesting acceptable statistical value support. In this study, 50.38% of the serum samples from backyard pigs were PDCoV-positive. The serological comparison showed that PDCoV/PEDV coinfections occurred in 31.98% of the analysed sera. These results can enrich our understanding of how this virus spreads and enable the evaluation of PDCoV infections. Moreover, it highlights the importance of continually investigating the seroprevalence of PDCoV in Mexico because there is also no information about the current prevalence of the disease.
Collapse
Affiliation(s)
- Francisco Jesus Castañeda Montes
- Estancias Posdoctorales por México para la Formación y Consolidación de las y los Investigadores por MéxicoCONAHCYTMexico CityMexico
- Posgrado en Ciencias de la Producción y de la Salud AnimalFacultad de Estudios Superiores, Universidad Nacional Autónoma de MéxicoCuautitlanEstado de MéxicoMexico
| | - José Luis Cerriteño Sánchez
- Centro Nacional de Investigación Disciplinaria en Salud Animal e InocuidadInstituto Nacional de Investigaciones ForestalesAgrícolas y PecuariasMexico CityMexico
| | - Julieta Sandra Cuevas‐Romero
- Centro Nacional de Investigación Disciplinaria en Salud Animal e InocuidadInstituto Nacional de Investigaciones ForestalesAgrícolas y PecuariasMexico CityMexico
| | - María Azucena Castañeda Montes
- Dirección de Procesos Alimentarios y Química Área Biotecnología, Unidad Académica de CapulhuacUniversidad Tecnológica del Valle de TolucaLermaMexico
- Laboratorio de Virología, Genética y Biología Molecular, Facultad de Educación Superior, Cuautitlán, Medicina VeterinariaUniversidad Nacional Autónoma de MéxicoCuautitlán IzcalliEstado de MéxicoMexico
| | - Susana Mendoza Elvira
- Posgrado en Ciencias de la Producción y de la Salud AnimalFacultad de Estudios Superiores, Universidad Nacional Autónoma de MéxicoCuautitlanEstado de MéxicoMexico
| |
Collapse
|
171
|
Chen Y, Tang Y, Zhang S, Tian Y, Xu S, Zhang C, Lin H, Zhao Q, Zhou EM, Liu B. Evaluation of novel synthetic peptides of avian hepatitis E virus ORF2 as vaccine candidate in chickens. Virus Res 2024; 349:199459. [PMID: 39237037 PMCID: PMC11406092 DOI: 10.1016/j.virusres.2024.199459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/18/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Avian hepatitis E virus (HEV) has resulted in significant economic losses in the poultry industry. There is currently no commercial vaccination available to prevent avian HEV infection. Previously, a novel epitope (601TFPS604) was discovered in the ORF2 protein of avian HEV. In this study, peptides were synthesized and assessed for their ability to provide immunoprotecting against avian HEV infection in poultry. Twenty-five Hy-Line Variety Brown laying hens were randomly divided into five groups; groups 1 to 3 respectively immunized with RLLDRLSRTFPS, PETRRLLDRLSR (irrelevant peptide control), or truncated avian HEV ORF2 protein (aa 339-606), while group 4 (negative control) was mock-immunized with PBS and group 5 (normal control) was not immunized or challenged. After the challenge, all hens in groups 2 and 4 showed seroconversion, fecal virus shedding, viremia, alanine aminotransferase (ALT) level increasing, liver lesions and HEV antigen in the liver. There were no pathogenic effects in other groups. Collectively, all of these findings showed that hens were completely protected against avian HEV infection when they were immunized with the peptide containing TFPS of the avian HEV ORF2 protein.
Collapse
Affiliation(s)
- Yiyang Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yujia Tang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shiyu Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yinuo Tian
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shenhao Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Chengwei Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Huanqing Lin
- Kongtong Animal Disease Prevention and Control Center, Pingliang, Gansu, China
| | - Qin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - En-Min Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Baoyuan Liu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
172
|
Oltjen H, Crook E, Lanier WA, Rettler H, Oakeson KF, Young EL, Torchetti M, Van Wettere AJ. SARS-CoV-2 delta variant in African lions (Panthera leo) and humans at Utah's Hogle Zoo, USA, 2021-22. Zoonoses Public Health 2024; 71:807-816. [PMID: 38825749 PMCID: PMC11455604 DOI: 10.1111/zph.13156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/29/2024] [Accepted: 05/11/2024] [Indexed: 06/04/2024]
Abstract
AIMS We conducted a One Health investigation to assess the source and transmission dynamics of SARS-CoV-2 infection in African lions (Panthera leo) at Utah's Hogle Zoo in Salt Lake City from October 2021 to February 2022. METHODS AND RESULTS Following observation of respiratory illness in the lions, zoo staff collected pooled faecal samples and individual nasal swabs from four lions. All specimens tested positive for SARS-CoV-2 by reverse transcription-polymerase chain reaction (RT-PCR). The resulting investigation included: lion observation; RT-PCR testing of lion faeces every 1-7 days; RT-PCR testing of lion respiratory specimens every 2-3 weeks; staff interviews and RT-PCR testing; whole-genome sequencing of viruses from lions and staff; and comparison with existing SARS-CoV-2 human community surveillance sequences. In addition to all five lions, three staff displayed respiratory symptoms. All lions recovered and no hospitalizations or deaths were reported among staff. Three staff reported close contact with the lions in the 10 days before lion illness onset, one of whom developed symptoms and tested positive for SARS-CoV-2 on days 3 and 4, respectively, after lion illness onset. The other two did not report symptoms or test positive. Two staff who did not have close contact with the lions were symptomatic and tested positive on days 5 and 8, respectively, after lion illness onset. We detected SARS-CoV-2 RNA in lion faeces for 33 days and in lion respiratory specimens for 14 weeks after illness onset. The viruses from lions were genetically highly related to those from staff and two contemporaneous surveillance specimens from Salt Lake County; all were delta variants (AY.44). CONCLUSIONS We did not determine the sources of these infections, although human-to-lion transmission likely occurred. The observed period of respiratory shedding was longer than in previously documented SARS-CoV-2 infections in large felids, indicating the need to further assess duration and potential implications of shedding.
Collapse
Affiliation(s)
- Heather Oltjen
- Utah Department of Health and Human Services, Salt Lake City, Utah, USA
| | | | - William A. Lanier
- Utah Department of Health and Human Services, Salt Lake City, Utah, USA
- Centers for Disease Control and Prevention, Office of Readiness and Response, Division of State and Local Readiness, Career Epidemiology Field Officer Program, Atlanta, Georgia, USA
- US Public Health Service, Rockville, Maryland, USA
| | - Hannah Rettler
- Utah Department of Health and Human Services, Salt Lake City, Utah, USA
| | - Kelly F. Oakeson
- Utah Public Health Laboratory, Utah Department of Health and Human Services, Salt Lake City, Utah, USA
| | - Erin L. Young
- Utah Public Health Laboratory, Utah Department of Health and Human Services, Salt Lake City, Utah, USA
| | - Mia Torchetti
- National Veterinary Services Laboratories, Animal and Plant Health Inspection Service, United States Department of Agriculture, Ames, Iowa, USA
| | | |
Collapse
|
173
|
Lim J, Hwang J, Min H, Wester M, Kim C, Valera E, Kong HJ, Bashir R. Dried Blood Matrix as a New Material for the Detection of DNA Viruses. Adv Healthc Mater 2024; 13:e2402506. [PMID: 39075818 PMCID: PMC11582504 DOI: 10.1002/adhm.202402506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/16/2024] [Indexed: 07/31/2024]
Abstract
The gold standard for diagnosing viruses such as the Hepatitis B Virus has remained largely unchanged, relying on conventional methods involving extraction, purification, and polymerase chain reaction (PCR). This approach is hindered by limited availability, as it is time-consuming and requires highly trained personnel. Moreover, it suffers from low recovery rates of the nucleic acid molecules for samples with low copy numbers. To address the challenges of complex instrumentation and low recovery rate of DNA, a drying process coupled with thermal treatment of whole blood is employed, resulting in the creation of a dried blood matrix characterized by a porous structure with a high surface-to-volume ratio where it also inactivates the amplification inhibitors present in whole blood. Drawing on insights from Brunauer-Emmett-Teller (BET)- Barrett-Joyner-Halenda (BJH) analysis, scanning electron microscopy (SEM), and fluorescence recovery after photobleaching (FRAP), detection assay is devised for HBV, as a demonstration, from whole blood with high recovery of DNA and simplified instrumentation achieving a limit of detection (LOD) of 10 IU mL-1. This assay can be completed in <1.5 h using a simple heater, can be applied to other DNA viruses, and is expected to be suitable for point-of-care, especially in low-resource settings.
Collapse
Affiliation(s)
- Jongwon Lim
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joanne Hwang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hyegi Min
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Matthew Wester
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Chansong Kim
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Enrique Valera
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hyun Joon Kong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Departments of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Rashid Bashir
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Biomedical and Translational Science, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL, 60642, USA
| |
Collapse
|
174
|
Zhou J, Sun P, Wang Y, Shi Y, Chen C, Xiao W, Qiu R, Cheng T, Fang L, Xiao S. Design and biological evaluation of candidate drugs against zoonotic porcine deltacoronavirus (PDCoV). Antiviral Res 2024; 231:106019. [PMID: 39395622 DOI: 10.1016/j.antiviral.2024.106019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus with zoonotic potential. PDCoV spillovers were recently detected in Haitian children with acute undifferentiated febrile illness, underscoring the urgent need to develop anti-PDCoV therapeutics. Coronavirus 3C-like protease (CoV 3CLpro) is essential for viral replication, and therefore provides an attractive target for drugs directed against CoV. Here, we initially evaluated the anti-PDCoV effect of Nirmatrelvir (PF-07321332), an FDA-approved anti-SARS-CoV-2 drug targeting viral 3CLpro. Regrettably, a very limited anti-PDCoV effect was achieved. By analyzing the binding modes of Nirmatrelvir with PDCoV 3CLpro and SARS-CoV-2 3CLpro, we demonstrated that the S2 pocket of 3CLpro is the primary factor underlying the differential inhibitory potency of Nirmatrelvir against different CoV 3CLpros. Based on the specific characteristics of the S2 pocket of PDCoV 3CLpro, four derivatives of Nirmatrelvir (compounds T1-T4) with substituted P2 moieties were synthesized. Compound T1, with an isobutyl at the P2 site, displayed improved anti-PDCoV activity invitro (cell infection model) and invivo (embryonated chicken egg infection model), and therefore is a potential candidate drug to combat PDCoV. Together, our results identify the substrate-binding mode and substrate specificity of PDCoV 3CLpro, providing insight into the optimization of Nirmatrelvir as an antiviral therapeutic agent against PDCoV.
Collapse
Affiliation(s)
- Junwei Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Peng Sun
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuanqing Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuting Shi
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chaoqun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Wenwen Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Runhui Qiu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Ting Cheng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
175
|
Dhakal J, Cancio LPM, Deliephan A, Chaves BD, Tubene S. Salmonella Presence and Risk Mitigation in Pet Foods: A Growing Challenge with Implications for Human Health. Compr Rev Food Sci Food Saf 2024; 23:e70060. [PMID: 39530630 PMCID: PMC11605167 DOI: 10.1111/1541-4337.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Pet food is increasingly recognized as a significant vehicle for the transmission of foodborne pathogens to humans. The intimate association between pets and their owners, coupled with the rising trend of feeding pets raw and unprocessed foods, contributes substantially to this issue. Salmonella contamination in pet food can originate from raw materials and feed ingredients, the processing environment, and postprocessing handling and applications. The absence of standardized postprocessing pathogen mitigation steps in the production of dry kibble and treats, along with the lack of validated heat and chemical interventions in raw pet foods, renders pet food susceptible to contamination by pathogens such as Salmonella, Listeria, E. coli, etc. Pets can then serve as carriers of Salmonella, facilitating its transmission to pet owners. Since 1999, there have been over 117 recalls of pet foods due to Salmonella contamination in the United States, with 11 of these recalls linked to human outbreaks. Notably, 5 of the 11 human outbreaks involved multidrug-resistant Salmonella strains. Various antimicrobial interventions, including high-pressure processing, ozone, irradiation, chemical treatments such as organic acids and acidulants, plant-derived antimicrobials, and biological interventions such as bacteriophages, have proven effective against Salmonella in pet foods. This review aims to summarize the prevalence of Salmonella in different types of pet foods, identify common sources of contamination, outline reported outbreaks, and discuss control measures and the regulatory framework governing pet food safety.
Collapse
Affiliation(s)
- Janak Dhakal
- Department of Agriculture, Food and Resource SciencesUniversity of Maryland Eastern ShorePrincess AnneMarylandUSA
| | - Leslie Pearl M. Cancio
- Provincial Science and Technology Office, Davao del SurDepartment of Science and Technology XI (DOST XI)Digos CityPhilippines
| | | | - Byron D. Chaves
- Department of Food Science and TechnologyUniversity of Nebraska–LincolnLincolnNebraskaUSA
| | - Stephan Tubene
- Department of Agriculture, Food and Resource SciencesUniversity of Maryland Eastern ShorePrincess AnneMarylandUSA
| |
Collapse
|
176
|
Lu Y, Li S, Yang S, Wang C, Fu Y, Yu H, Huang X, Zhao J, Shao Y, Wang Z, Cui Y, Chen J, Guo Q, Kuang L, Liu G. Variation in innate immune responses to porcine epidemic diarrhea virus infection in piglets at different ages. Microb Pathog 2024; 196:106958. [PMID: 39303959 DOI: 10.1016/j.micpath.2024.106958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) poses a significant threat to pigs, with piglets under seven days old facing a mortality rate of up to 100 %. This study aimed to explore the maturation of the immune system in piglets across different age groups and their corresponding immune responses to PEDV infection. Real-time quantitative PCR was employed to assess the relative mRNA expression of inflammation-related factors in infected pigs compared to non-infected counterparts at varying ages. Additionally, flow cytometry was utilized to analyze the relative counts of CD4+ and CD8+ T cells, as well as CD21+ B cells, in peripheral blood, spleen, mesenteric lymph nodes, and Peyer's patches of piglets at different developmental stages. Our findings revealed a notable increase in IFN-α and IFN-γ, a decrease in TNF-α, and elevated expression of IL-1β, IL-6, IL-10, and IL-17 following PEDV infection. Furthermore, the numbers of CD4+ and CD8+ T cells, along with CD21+ B cells, exhibited a gradual rise with the advancement of piglets' age. Overall, our study underscores the progressive enhancement of piglets' resistance to PEDV infection as their immune system matures over time.
Collapse
Affiliation(s)
- Yabin Lu
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shuxian Li
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Shanshan Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Caiying Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yuguang Fu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Haoyuan Yu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Xin Huang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Jing Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yongheng Shao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Zemei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yaru Cui
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Jianing Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Qingyong Guo
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Ling Kuang
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China.
| | - Guangliang Liu
- Xinjiang Key Laboratory of New Drug Study and Creation for Herbivorous Animals (XJ-KLNDSCHA), College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China; State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
177
|
Jiang H, Jia M, Xiong J, Zhao C, Wang T, Kong L, Peng Q. The network interactions between the porcine deltacoronavirus nucleocapsid protein and host cellular proteins. Vet Microbiol 2024; 298:110225. [PMID: 39154555 DOI: 10.1016/j.vetmic.2024.110225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging swine coronavirus that can cause diarrhea in pigs of all ages with varying severity. Host-virus protein interactions are critical for intracellular viral replication. Elucidating the interactions between cellular and viral proteins can help us to design antiviral strategies. PDCoV N protein is the most abundant and vital regulator in virus replication. In this study, 604 host proteins were identified to interact with PDCoV N protein by Co-IP combined with LC-MS, of which 243 proteins were specifically bound to N protein. PPI analysis revealed that the N-interacting host proteins are categorized into three groups: ribonucleoprotein complex biogenesis modulation, cellular nitrogen compound metabolism, and nucleic acid binding. GO and KEGG analyses showed that the host proteins are primarily involved in mRNA splicing, stress granule assembly, spliceosomal snRNP assembly. Additionally, four host proteins-TRIM25, HNRNPUL1, RPS27A, and SLC3A2-were selected to validate the interactome data through Co-IP and Confocal assays. This study can help in designing anti-PDCoV strategies and understanding the replication mechanism of PDCoV.
Collapse
Affiliation(s)
- Hui Jiang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China
| | - Mengle Jia
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Jiaqi Xiong
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Changrun Zhao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Ting Wang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Lingbao Kong
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Qi Peng
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China; College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
178
|
Xue W, Chu H, Wang J, Sun Y, Qiu X, Song C, Tan L, Ding C, Liao Y. Coronavirus nucleocapsid protein enhances the binding of p-PKCα to RACK1: Implications for inhibition of nucleocytoplasmic trafficking and suppression of the innate immune response. PLoS Pathog 2024; 20:e1012097. [PMID: 39602452 PMCID: PMC11633972 DOI: 10.1371/journal.ppat.1012097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 12/11/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
The hallmark of coronavirus infection lies in its ability to evade host immune defenses, a process intricately linked to the nuclear entry of transcription factors crucial for initiating the expression of antiviral genes. Central to this evasion strategy is the manipulation of the nucleocytoplasmic trafficking system, which serves as an effective target for the virus to modulate the expression of immune response-related genes. In this investigation, we discovered that infection with the infectious bronchitis virus (IBV) dynamically impedes the nuclear translocation of several transcription factors such as IRF3, STAT1, STAT2, NF-κB p65, and the p38 MAPK, leading to compromised transcriptional induction of key antiviral genes such as IFNβ, IFITM3, and IL-8. Further examination revealed that during the infection process, components of the nuclear pore complex (NPC), particularly FG-Nups (such as NUP62, NUP153, NUP42, and TPR), undergo cytosolic dispersion from the nuclear envelope; NUP62 undergoes phosphorylation, and NUP42 exhibits a mobility shift in size. These observations suggest a disruption in nucleocytoplasmic trafficking. Screening efforts identified the IBV nucleocapsid (N) protein as the agent responsible for the cytoplasmic distribution of FG-Nups, subsequently hindering the nuclear entry of transcription factors and suppressing the expression of antiviral genes. Interactome analysis further revealed that the IBV N protein interacts with the scaffold protein RACK1, facilitating the recruitment of activated protein kinase C alpha (p-PKCα) to RACK1 and relocating the p-PKCα-RACK1 complex to the cytoplasm. These observations are conserved across diverse coronaviruses N proteins. Concurrently, the presence of both RACK1 and PKCα/β proved essential for the phosphorylation and cytoplasmic dispersion of NUP62, the suppression of antiviral cytokine expression, and efficient virus replication. These findings unveil a novel, highly effective, and evolutionarily conserved mechanism.
Collapse
Affiliation(s)
- Wenxiang Xue
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Hongyan Chu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Jiehuang Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, P. R. China
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| |
Collapse
|
179
|
Xu Z, Zhang R, Chen H, Zhang L, Yan X, Qin Z, Cong S, Tan Z, Li T, Du M. Characterization and preparation of food-derived peptides on improving osteoporosis: A review. Food Chem X 2024; 23:101530. [PMID: 38933991 PMCID: PMC11200288 DOI: 10.1016/j.fochx.2024.101530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/18/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Osteoporosis is a systemic bone disease characterized by reduced bone mass and deterioration of the microstructure of bone tissue, leading to an increased risk of fragility fractures and affecting human health worldwide. Food-derived peptides are widely used in functional foods due to their low toxicity, ease of digestion and absorption, and potential to improve osteoporosis. This review summarized and discussed methods of diagnosing osteoporosis, treatment approaches, specific peptides as alternatives to conventional drugs, and the laboratory preparation and identification methods of peptides. It was found that peptides interacting with RGD (arginine-glycine-aspartic acid)-binding active sites in integrin could alleviate osteoporosis, analyzed the interaction sites between these osteogenic peptides and integrin, and further discussed their effects on improving osteoporosis. These may provide new insights for rapid screening of osteogenic peptides, and provide a theoretical basis for their application in bone materials and functional foods.
Collapse
Affiliation(s)
- Zhe Xu
- School of Food Science and Technology, State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China
- College of Life Sciences, Key Laboratory of Biotechnology and Bioresources Utilization, Dalian Minzu University, Ministry of Education, Dalian 116600, China
- Institute of Bast Fiber Crops & Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China
| | - Rui Zhang
- School of Food Science and Technology, State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China
| | - Hongrui Chen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Xihua University, Chengdu, Sichuan 611130, China
| | - Lijuan Zhang
- College of Life Sciences, Key Laboratory of Biotechnology and Bioresources Utilization, Dalian Minzu University, Ministry of Education, Dalian 116600, China
| | - Xu Yan
- College of Life Sciences, Key Laboratory of Biotechnology and Bioresources Utilization, Dalian Minzu University, Ministry of Education, Dalian 116600, China
| | - Zijin Qin
- Department of Food Science and Technology, University of Georgia, Clarke, Athens, GA 30602, USA
| | - Shuang Cong
- College of Life Sciences, Yantai University, Yantai, Shandong 264005, China
| | - Zhijian Tan
- Institute of Bast Fiber Crops & Center of Southern Economic Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China
| | - Tingting Li
- College of Life Sciences, Key Laboratory of Biotechnology and Bioresources Utilization, Dalian Minzu University, Ministry of Education, Dalian 116600, China
| | - Ming Du
- School of Food Science and Technology, State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
180
|
Toth DJA, Sheets TR, Beams AB, Ahmed SM, Seegert N, Love J, Keegan LT, Samore MH. Model-based estimates of age-structured SARS-CoV-2 epidemiology in households. BMC Public Health 2024; 24:2965. [PMID: 39455984 PMCID: PMC11515260 DOI: 10.1186/s12889-024-20308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Understanding how infectious disease transmission varies from person to person, including associations with age and contact behavior, can help design effective control strategies. Within households, transmission may be highly variable because of differing transmission risks by age, household size, and individual contagiousness. Our aim was to disentangle those factors by fitting mathematical models to SARS-CoV-2 household survey and serologic data. METHODS We surveyed members of 3,381 Utah households from January-April 2021 and performed SARS-CoV-2 antibody testing on all available members. We paired these data with a probabilistic model of household importation and transmission composed of a novel combination of transmission variability and age- and size-structured heterogeneity. We calculated maximum likelihood estimates of mean and variability of household transmission probability between household members in different age groups and different household sizes, simultaneously with importation probability and probabilities of false negative and false positive test results. RESULTS 12.8% of individual participants, residing in 17.4% of the participating households, showed serologic evidence of prior infection or reported a prior positive test on the survey. Serologically positive individuals in younger age groups were less likely than older adults to have tested positive during their infection according to our survey results. Our model results suggested that adolescents and young adults (ages 13-24) acquired SARS-CoV-2 infection outside the household at a rate substantially higher than younger children and older adults. Our estimate of the household secondary attack rate (HSAR) among adults aged 45 and older exceeded HSARs to and/or from younger age groups at a given household size. We found lower HSAR in households with more members, independent of age differences. The age-specific HSAR patterns we found could not be explained by age-dependent biological susceptibility and transmissibility alone, suggesting that age groups contacted each other at different rates within households. CONCLUSIONS We disentangled several factors contributing to age-specific infection risk, including non-household exposure, within-household exposure to specific age groups, and household size. Within-household contact rate differences played a significant role in driving household transmission epidemiology. These findings provide nuanced insights for understanding community outbreak patterns and mechanisms of differential infection risk.
Collapse
Affiliation(s)
- Damon J A Toth
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America.
- Department of Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, United States of America.
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States of America.
| | - Theresa R Sheets
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States of America
| | - Alexander B Beams
- Department of Mathematics, University of Utah, Salt Lake City, UT, United States of America
- Present address: Department of Mathematics, Simon Fraser University, Burnaby, BC, Canada
| | - Sharia M Ahmed
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Nathan Seegert
- Department of Finance, University of Utah David Eccles School of Business, Salt Lake City, UT, United States of America
| | - Jay Love
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Lindsay T Keegan
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, United States of America
| | - Matthew H Samore
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Department of Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, UT, United States of America
| |
Collapse
|
181
|
Bashor L, Gallichotte EN, Galvan M, Erbeck K, Croft L, Stache K, Stenglein M, Johnson JG, Pabilonia K, VandeWoude S. SARS-CoV-2 within-host population expansion, diversification and adaptation in zoo tigers, lions and hyenas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620075. [PMID: 39484504 PMCID: PMC11527109 DOI: 10.1101/2024.10.24.620075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
SARS-CoV-2 rapidly adapts to new hosts following cross-species transmission; this is highly relevant as novel within-host variants have emerged following infection of susceptible wild and domestic animal species. Furthermore, SARS-CoV-2 transmission from animals (e.g., white-tailed deer, mink, domestic cats, and others) back to humans has also been observed, documenting the potential of novel animal-derived variants to infect humans. We investigated SARS-CoV-2 evolution and host-specific adaptation during an outbreak in Amur tigers (Panthera tigris altaica), African lions (Panthera leo), and spotted hyenas (Crocuta crocuta) at Denver Zoo in late 2021. SARS-CoV-2 genomes from longitudinal samples collected from 16 individuals were evaluated for within-host variation and genomic signatures of selection. The outbreak was likely initiated by a single spillover of a rare Delta sublineage subsequently transmitted from tigers to lions to hyenas. Within-host virus populations rapidly expanded and diversified. We detected signatures of purifying and positive selection, including strong positive selection in hyenas and in the nucleocapsid (N) gene in all animals. Four candidate species-specific adaptive mutations were identified: N A254V in lions and hyenas, and ORF1a E1724D, spike T274I, and N P326L in hyenas. These results reveal accelerated SARS-CoV-2 adaptation following host shifts in three non-domestic species in daily contact with humans.
Collapse
Affiliation(s)
- Laura Bashor
- Dept. of Microbiology, Immunology and Pathology, Colorado State University
| | | | - Michelle Galvan
- Dept. of Microbiology, Immunology and Pathology, Colorado State University
| | - Katelyn Erbeck
- Colorado State University Veterinary Diagnostic Laboratories
| | | | | | - Mark Stenglein
- Dept. of Microbiology, Immunology and Pathology, Colorado State University
| | | | | | - Sue VandeWoude
- Dept. of Microbiology, Immunology and Pathology, Colorado State University
| |
Collapse
|
182
|
Leonardi-Cattolica A, Kayastha S, Miller M, Guag J, Tkachenko A, Lowe J, Allender M, Terio K, Wang L. Evaluation of Fecal Sample Pooling for Real-Time RT-PCR Testing SARS-CoV-2 in Animals. Viruses 2024; 16:1651. [PMID: 39599766 PMCID: PMC11599033 DOI: 10.3390/v16111651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024] Open
Abstract
During the COVID-19 pandemic, veterinary diagnostic laboratories tested both human and animal samples and needed to ensure that they could accurately perform large numbers of diagnostic tests in a timely manner. Sample pooling, a methodology used effectively for over 80 years as a surveillance tool for screening large numbers of potentially infected individuals, was employed. Given its sensitivity, real-time polymerase chain reaction (PCR) is more suitable for employing this strategy, as compared to other less sensitive testing methods. In this study, we evaluated the capability of detecting SARS-CoV-2 in both 5-sample and 10-sample pools of feces using real-time reverse transcriptase polymerase chain reaction (rRT-PCR) as well as determined the level of sensitivity. A blinded method test (BMT) by an independent laboratory was conducted to assess the five-sample fecal pool. To complement detection capability, the stability of the genome within a PBS fecal suspension was measured under various time and temperature conditions across a 28-day period. Our results showed that the limit of detection for 5-sample and 10-sample fecal pools is 12.8 and 6.4 genome copies in a 25 µL PCR, respectively. The 5-sample and 10-sample pooling resulted in a cycle threshold (Ct) value loss of 2.35 and 3.45, as compared to Ct values of known positive individual samples, but consistent detection was still achieved in pools containing positive samples with an original Ct below 36 and 34, respectively. The simulation of clinical five-sample pooling showed that all positive samples could be detected regardless of the number (1-3) of positive samples in each pool. The BMT results demonstrated excellent sensitivity (100 copies/reaction) in five-sample pools for the detection of SARS-CoV-2 RNA even though a fecal matrix effect was observed. Finally, our results show that the SARS-CoV-2 genome remains stable over a wide range of time and temperature variations. Overall, our findings provide solid data to scale up SARS-CoV-2 testing capacity in veterinary diagnostic laboratories.
Collapse
Affiliation(s)
- Antonio Leonardi-Cattolica
- Veterinary Diagnostic Laboratory, Department of Veterinary Clinical Medicine, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
- Department of Pathobiology, University of Illinois at Urbana-Champaign, 2001 South Lincoln Ave, Urbana, IL 61802, USA
| | - Sandipty Kayastha
- Veterinary Diagnostic Laboratory, Department of Veterinary Clinical Medicine, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
| | - Megan Miller
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Jake Guag
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Andriy Tkachenko
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - James Lowe
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, 2001 South Lincoln Ave, Urbana, IL 61802, USA
| | - Matthew Allender
- Veterinary Diagnostic Laboratory, Department of Veterinary Clinical Medicine, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
| | - Karen Terio
- Zoological Pathology Program, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
| | - Leyi Wang
- Veterinary Diagnostic Laboratory, Department of Veterinary Clinical Medicine, University of Illinois College of Veterinary Medicine, Urbana, IL 61802, USA
| |
Collapse
|
183
|
Singh N, Miller MR, Nemser SM, Tkachenko A, Uhlig S, Frost K, Hettwer K, Ulaszek J, Kmet M, Wang L, Allender MC, Reddy R. Proficiency test of SARS-CoV-2 Omicron variant detection in diagnostics samples by veterinary diagnostic laboratories. ACCREDITATION AND QUALITY ASSURANCE 2024. [DOI: 10.1007/s00769-024-01622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024]
Abstract
AbstractVeterinary diagnostic laboratories (VDLs) play a critical role in screening both human and animal samples for SARS-CoV-2. To evaluate the SARS-CoV-2 detection methods used by VDLs, a proficiency test was performed by the US Food and Drug Administration’s Veterinary Laboratory and Investigation and Response Network in collaboration with two other organizations. Thirty-two sets of 12 blind-coded samples were prepared by fortifying Molecular Transport Medium (MTM) or feline feces with SARS-CoV-2 Omicron variant or non-SARS-CoV-2 equine coronavirus RNA at various concentrations and shipped to 32 participants for blinded (unbiased) analysis. Results were analyzed according to the principles of International Organization for Standardization 16140-2:2016 using two approaches such as establishing the rate of detection (ROD) and the success rate by applying the analysis of binary outcome by logit approach. ROD provided the overall assessment of laboratories performance, whereas the novel logit approach provided an insight to more specific analysis based on the complexity of each sample. The ROD was 83% and 98% for MTM samples at 200 and 20000 genome copies per 100 µL, respectively. Fecal samples were classified as challenging exploratory, and results were not included in the assessment of performance but discussion purposes only. Fecal samples exhibited matrix interference impacting the performance. The ROD was 44% and 89% for fecal samples at 2000 and 20000 genome copies per 100 µL, respectively. The non-COVID coronavirus RNA, which was used to address the specificity, did not interfere with methods used. Establishing the success rate by evaluating the qualitative results (detected/not detected) applying a logit approach revealed that, out of thirty-two participants, twenty-eight had satisfactory results, one participant had unsatisfactory results, and three participants had questionable results for MTM samples. For fecal samples, three participants out of thirty-two did not meet the expectations at higher concentrations. Lower concentrations of fecal samples were excluded from this analysis. Again, the fecal samples were considered as challenge samples and the results were provided to assist participants in their continuous efforts to improve their performance and not to evaluate their performance.
Collapse
|
184
|
Li Y, Hu J, Hou J, Lu S, Xiong J, Wang Y, Sun Z, Chen W, Pan Y, Thilakavathy K, Feng Y, Jiang Q, Wang W, Xiong C. Study on sentinel hosts for surveillance of future COVID-19-like outbreaks. Sci Rep 2024; 14:24595. [PMID: 39427096 PMCID: PMC11490639 DOI: 10.1038/s41598-024-76506-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
The spread of SARS-CoV-2 to animals has the potential to evolve independently. In this study, we distinguished several sentinel animal species and genera for monitoring the re-emergence of COVID-19 or the new outbreak of COVID-19-like disease. We analyzed SARS-CoV-2 genomic data from human and nonhuman mammals in the taxonomic hierarchies of species, genus, family and order of their host. We find that SARS-CoV-2 carried by domestic dog (Canis lupus familiaris), domestic cat (Felis catus), mink (Neovison vison), and white-tailed deer (Odocoileus virginianus) cluster closely to human-origin viruses and show no differences in the majority of amino acids, but have the most positively selected sites and should be monitored to prevent the re-emergence of COVID-19 caused by novel variants of SARS-CoV-2. Viruses from the genera Panthera (especially lion (Panthera leo)), Manis and Rhinolophus differ significantly from human-origin viruses, and long-term surveillance should be undertaken to prevent the future COVID-19-like outbreaks. Investigation of the variation dynamics of sites 142, 501, 655, 681 and 950 within the S protein may be necessary to predict the novel animal SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yanjiao Li
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Jingjing Hu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Jingjing Hou
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuiping Lu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Jiasheng Xiong
- Division of Emergency Management, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, 200336, China
| | - Yuxi Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Zhong Sun
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia
| | - Weijie Chen
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Yue Pan
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Karuppiah Thilakavathy
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qingwu Jiang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China.
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China.
| | - Chenglong Xiong
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200433, China.
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
185
|
Guo J, He J, Liang Z, Huang S, Wen F. Birds as reservoirs: unraveling the global spread of Gamma- and Deltacoronaviruses. mBio 2024; 15:e0232424. [PMID: 39230281 PMCID: PMC11481860 DOI: 10.1128/mbio.02324-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
Avian migration is a global phenomenon that transcends geographical boundaries. These migratory birds serve as unwitting carriers of diverse Gammacoronaviruses (γ-CoVs) and Deltacoronaviruses (δ-CoVs). While recombination events have been documented among γ-CoVs in avian species and β-CoVs in mammals, evidence for recombination between CoVs of distinct genera remains limited. This minireview examines the prevalence of CoVs in both domestic waterfowl (ducks and geese) and wild bird populations inhabiting various regions. We investigate the dissemination patterns of γ-CoVs and δ-CoVs among these populations, highlighting their shared characteristics. Furthermore, the review explores the intricate web of cross-species transmission of δ-CoVs from wild birds to mammals, with a particular focus on pigs. Understanding the distinct features of CoVs harbored by waterfowl and wild birds and their potential for cross-species transmission is crucial for preparedness and response to future CoV epidemics.
Collapse
Affiliation(s)
- Jinyue Guo
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Jieheng He
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zhaoping Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shujian Huang
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Feng Wen
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| |
Collapse
|
186
|
Gallichotte EN, Bashor L, Erbeck K, Croft L, Stache K, Long J, VandeWoude S, Johnson JG, Pabilonia KL, Ebel GD. SARS-CoV-2 outbreak in lions, tigers and hyenas at Denver Zoo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.617443. [PMID: 39464021 PMCID: PMC11507794 DOI: 10.1101/2024.10.14.617443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
In late 2019, SARS-CoV-2 spilled-over from an animal host into humans, where it efficiently spread, resulting in the COVID-19 pandemic. Through both natural and experimental infections, we learned that many animal species are susceptible to SARS-CoV-2. Importantly, animals in close proximity to humans, including companion, farmed, and those at zoos and aquariums, became infected, and many studies demonstrated transmission to/from humans in these settings. In this study, we first review the literature of SARS-CoV-2 infections in tigers and lions, and compare species, sex, age, virus and antibody detection assay, and types, frequency and length of clinical signs, demonstrating broad heterogeneity amongst infections. We then describe a SARS-CoV-2 outbreak in lions, tigers and hyenas at Denver Zoo in late 2021. Animals were tested for viral RNA (vRNA) for four months. Lions had significantly more viral RNA in nasal swabs than both tigers and hyenas, and many individual lions experienced viral recrudescence after weeks of undetectable vRNA. Infectious virus was correlated with high levels of vRNA and was more likely to be detected earlier during infection. Four months post-infection, all tested animals generated robust neutralizing antibody titers. Animals were infected with Delta lineage AY.20 identical to a variant circulating at less than 1% in Colorado humans at that time, suggesting a single spillover event from an infected human spread within and between species housed at the zoo. Better understanding of epidemiology and susceptibility of SARS-CoV-2 infections in animals is critical to limit the current and future spread and protect animal and human health.
Collapse
Affiliation(s)
- Emily N Gallichotte
- Department of Microbiology, Immunology and Pathology, Colorado State University
| | - Laura Bashor
- Department of Microbiology, Immunology and Pathology, Colorado State University
| | - Katelyn Erbeck
- Veterinary Diagnostic Laboratories, Colorado State University
| | | | | | | | - Sue VandeWoude
- Department of Microbiology, Immunology and Pathology, Colorado State University
| | | | - Kristy L Pabilonia
- Department of Microbiology, Immunology and Pathology, Colorado State University
- Veterinary Diagnostic Laboratories, Colorado State University
| | - Gregory D Ebel
- Department of Microbiology, Immunology and Pathology, Colorado State University
| |
Collapse
|
187
|
Jarvie MM, Nguyen TNT, Southwell B, Wright D. Integration of Whole-Genome Sequencing with ddPCR Kit for Detection of Omicron Subvariants in Wastewater in the Upper Peninsula of Michigan. Appl Microbiol 2024; 4:1453-1463. [DOI: 10.3390/applmicrobiol4040100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
This study explores the integration of genome sequencing and digital droplet polymerase chain reaction (ddPCR)-based methods for tracking the diversity of COVID-19 variants in wastewater. The research focuses on monitoring various Omicron subvariants during a period of significant viral evolution. Genome sequencing, particularly using Oxford Nanopore Technology (ONT), provides a detailed view of emerging variants, surpassing the limitations of PCR-based detection kits that rely on known sequences. Of the 43 samples analyzed, 39.5% showed matching results between the GT Molecular ddPCR kits and sequencing, though only 4% were exact matches. Some mismatches occurred due to newer subvariants like XBB and BQ.1, which the ddPCR kits could not detect. This emphasized the limitations of ddPCR kits, which rely on known variant sequences, while sequencing provides real-time data on emerging variants, offering a more comprehensive view of circulating strains. This study highlights the effectiveness of combining these methodologies to enhance early detection and inform public health strategies, especially in regions with limited clinical sequencing capabilities.
Collapse
Affiliation(s)
- Michelle M. Jarvie
- School of Chemistry, Environmental, and Geosciences, Lake Superior State University, 650 W. Easterday Ave., Sault Ste. Marie, MI 49783, USA
| | - Thu N. T. Nguyen
- School of Chemistry, Environmental, and Geosciences, Lake Superior State University, 650 W. Easterday Ave., Sault Ste. Marie, MI 49783, USA
| | - Benjamin Southwell
- School of Chemistry, Environmental, and Geosciences, Lake Superior State University, 650 W. Easterday Ave., Sault Ste. Marie, MI 49783, USA
| | - Derek Wright
- School of Chemistry, Environmental, and Geosciences, Lake Superior State University, 650 W. Easterday Ave., Sault Ste. Marie, MI 49783, USA
| |
Collapse
|
188
|
Ochar K, Iwar K, Nair VD, Chung YJ, Ha BK, Kim SH. The Potential of Glucosinolates and Their Hydrolysis Products as Inhibitors of Cytokine Storms. Molecules 2024; 29:4826. [PMID: 39459194 PMCID: PMC11510469 DOI: 10.3390/molecules29204826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
A cytokine storm is an intense inflammatory response characterized by the overproduction of proinflammatory cytokines, resulting in tissue damage, and organ dysfunction. Cytokines play a crucial role in various conditions, such as coronavirus disease, in which the immune system becomes overactive and releases excessive levels of cytokines, including interleukins, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ). This anomalous response often leads to acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation (DIC), and multiple organ injury (MOI). Glucosinolates are plant secondary metabolites predominantly found in Brassica vegetables, but are also present in other species, such as Moringa Adens and Carica papaya L. When catalyzed by the enzyme myrosinase, glucosinolates produce valuable products, including sulforaphane, phenethyl isothiocyanate, 6-(methylsulfinyl) hexyl isothiocyanate, erucin, goitrin, and moringin. These hydrolyzed products regulate proinflammatory cytokine production by inhibiting the nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) signaling pathway and stimulating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. This action can alleviate hyperinflammation in infected cells and modulate cytokine storms. In this review, we aimed to examine the potential role of glucosinolates in modulating cytokine storms and reducing inflammation in various conditions, such as coronavirus disease. Overall, we found that glucosinolates and their hydrolysis products can potentially attenuate cytokine production and the onset of cytokine storms in diseased cells. In summary, glucosinolates could be beneficial in regulating cytokine production and preventing complications related to cytokine storms.
Collapse
Affiliation(s)
- Kingsley Ochar
- Council for Scientific and Industrial Research, Plant Genetic Resources Research Institute, Bunso P.O. Box 7, Ghana;
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Kanivalan Iwar
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Vadakkemuriyil Divya Nair
- Department of Plant Sciences, Central University of Himachal Pradesh, Shahpur Campus, Kangra District, Shahpur 176206, HP, India;
| | - Yun-Jo Chung
- National Creative Research Laboratory for Ca Signaling Network, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea;
| | - Bo-Keun Ha
- Department of Applied Plant Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seong-Hoon Kim
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| |
Collapse
|
189
|
Lunardi M, Martins FDC, Gustani-Buss E, Chideroli RT, de Oliveira IM, Peronni KC, Figueiredo DLA, Alfieri AF, Alfieri AA. Higher Frequency of SARS-CoV-2 RNA Shedding by Cats than Dogs in Households with Owners Recently Diagnosed with COVID-19. Viruses 2024; 16:1599. [PMID: 39459932 PMCID: PMC11512312 DOI: 10.3390/v16101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Studies have demonstrated the susceptibility of companion animals to natural infection with SARS-CoV-2. Using quantitative reverse transcription polymerase chain reaction and sequencing analyses, this study investigated SARS-CoV-2 RNA excretion in pets in households with infected owners. Oropharyngeal and rectal swabs were collected from dogs and cats in Parana, Southern Brazil, between October 2020 and April 2021. Viral RNA was detected in 25% of cats and 0.98% of dog oropharyngeal swabs; however, systemic, respiratory, and gastrointestinal signs were absent. Complete viral genomes belonged to the Gamma lineage. Phylogenetic analyses indicated that pet samples were probably derived from human-positive cases in Parana. Viral excretion in the oropharynx was more frequent in cats than in dogs. Mutations in the S protein characteristic of Gamma strains were present in all sequenced SARS-CoV-2 strains. The receptor-binding domain of these Brazilian strains did not show any additional mutations not reported in the Gamma strains. Mutations in NSP6, NSP12, and N proteins previously mapped to strains that infect deer or minks were detected. This study highlights the importance of actively monitoring the SARS-CoV-2 strains that infect pets with continued viral exposure. Monitoring genetic changes is crucial because new variants adapted to animals may pose human health risks.
Collapse
Affiliation(s)
- Michele Lunardi
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil; (M.L.); (A.F.A.)
- Multi-User Animal Health Laboratory, Molecular Biology Unit, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
- Post Graduate Program in Animal Health and Production, Department of Agrarian Sciences, University Pitagoras Unopar, Arapongas 86702-670, Brazil
| | - Felippe Danyel Cardoso Martins
- Multi-User Animal Health Laboratory, Molecular Biology Unit, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
- Post Graduate Program in Animal Science, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | - Emanuele Gustani-Buss
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven—University of Leuven, Box 1030, 3000 Leuven, Belgium;
| | - Roberta Torres Chideroli
- Post Graduate Program in Animal Science, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | | | - Kamila Chagas Peronni
- Institute for Cancer Research, IPEC, Guarapuava 85100-000, Brazil; (I.M.d.O.); (K.C.P.); (D.L.A.F.)
| | - David Livingstone Alves Figueiredo
- Institute for Cancer Research, IPEC, Guarapuava 85100-000, Brazil; (I.M.d.O.); (K.C.P.); (D.L.A.F.)
- Department of Medicine, Midwestern Parana State University—UNICENTRO, Guarapuava 85040-167, Brazil
| | - Alice Fernandes Alfieri
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil; (M.L.); (A.F.A.)
- Multi-User Animal Health Laboratory, Molecular Biology Unit, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | - Amauri Alcindo Alfieri
- Laboratory of Animal Virology, Department of Veterinary Preventive Medicine, Universidade Estadual de Londrina, Londrina 86057-970, Brazil; (M.L.); (A.F.A.)
- National Institute of Science and Technology for Dairy Production Chain (INCT–LEITE), Universidade Estadual de Londrina, Londrina 86057-970, Brazil
| |
Collapse
|
190
|
Shi K, Hu X, Long F, Shi Y, Pan Y, Feng S, Li Z, Yin Y. Genetic diversity and evolution of porcine hemagglutinating encephalomyelitis virus in Guangxi province of China during 2021-2024. Front Microbiol 2024; 15:1474552. [PMID: 39444682 PMCID: PMC11496168 DOI: 10.3389/fmicb.2024.1474552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is the only known porcine neurotropic coronavirus, which is prevalent worldwide at present. It is of great significance to understand the genetic and evolutionary characteristics of PHEV in order to perform effective measures for prevention and control of this disease. In this study, a total of 6,986 tissue samples and nasopharyngeal swabs were collected from different regions of Guangxi province in southern China during 2021-2024, and were tested for PHEV using a quadruplex RT-qPCR. The positivity rate of PHEV was 2.81% (196/6,986), of which tissue samples and nasopharyngeal swabs had 2.05% (87/4,246) and 3.98% (109/2,740) positivity rates, respectively. Fifty PHEV positive samples were selected for PCR amplification and gene sequencing. Sequence analysis revealed that the nucleotide homology and amino acid similarities of S, M, and N genes were 94.3%-99.3% and 92.3%-99.2%, 95.0%-99.7% and 94.7%-100.0%, 94.0%-99.5% and 93.5%-99.3%, respectively, indicating M and N genes were more conservative than S gene. Phylogenetic trees based on these three genes revealed that PHEV strains from different countries could be divided into two groups G1 and G2, and the PHEV strains from Guangxi province obtained in this study distributed in subgroups G1c and G2b. Bayesian analysis revealed that the population size of PHEV has been in a relatively stable state since its discovery until it expanded sharply around 2015, and still on the slow rise thereafter. S gene sequences analysis indicated that PHEV strains existed variation of mutation, and recombination. The results indicated that the prevalent PHEV strains in Guangxi province had complex evolutionary trajectories and high genetic diversity. To the best of our knowledge, this is the first report on the genetic and evolutionary characteristics of PHEV in southern China.
Collapse
Affiliation(s)
- Kaichuang Shi
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Center for Animal Disease Control and Prevention, Nanning, China
| | - Xin Hu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Feng Long
- Guangxi Center for Animal Disease Control and Prevention, Nanning, China
| | - Yuwen Shi
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yi Pan
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, China
| | - Shuping Feng
- Guangxi Center for Animal Disease Control and Prevention, Nanning, China
| | - Zongqiang Li
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yanwen Yin
- Guangxi Center for Animal Disease Control and Prevention, Nanning, China
| |
Collapse
|
191
|
Su M, Yan Y, Huang Y, Ren J, Niu S, Zhao Y, Yan F, Tian WX, Wang Y. Isolation and characterization of a subtype G2c variant of porcine epidemic diarrhea virus that adapts well to cell culture. Arch Virol 2024; 169:217. [PMID: 39379633 DOI: 10.1007/s00705-024-06140-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 08/09/2024] [Indexed: 10/10/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) causes the third most important disease in the pig industry, after African swine fever and porcine reproductive and respiratory syndrome, and leads to illness or death of the entire litter, causing significant economic losses. In this study, three PEDV strains (HN-1, HN-2, and SC2023) were isolated from swine farms with suspected PEDV infections in Sichuan and Henan provinces. Phylogenetic analysis based on complete S gene sequences showed that all three strains belonged to the G2c subgroup. HN-1 adapted readily to cell culture, grew to a viral titer as high as 2 × 108 TCID50/mL in Vero cells, and caused the formation of large syncytia. We analyzed the amino acid sequence of the HN-1 isolate and found that its S1 subunit contained a three-amino-acid insertion (355KRL358). A seven-amino-acid-deletion (1377FEKVHVQ1383) in the S2 subunit resulted in the partial deletion of the endocytosis signal YxxΦ and the complete deletion of the endoplasmic reticulum retrieval signal (ERRS) KVHVQ in the cytoplasmic tail of the S protein. Consequently, HN-1 is predicted to be less pathogenic than its parent strain, an attribute that facilitates rapid cell-to-cell spread by enhancing syncytium formation. In addition, strain HN-1 was found to have the mutation 884-885SG→RR, which may favor adaptation to cell culture by providing new trypsin cleavage sites. These results suggest that HN-1 is a G2c subtype variant that adapts well to cell culture and can be used to study the adaptive mechanisms of PEDV and develop attenuated vaccines.
Collapse
Affiliation(s)
- Min Su
- Shanxi Agricultural University, Jinzhong, China
| | - Yi Yan
- Shanxi Agricultural University, Jinzhong, China
| | | | - Jianle Ren
- Shanxi Agricultural University, Jinzhong, China
| | - Sheng Niu
- Shanxi Agricultural University, Jinzhong, China
| | - Yujun Zhao
- Shanxi Agricultural University, Jinzhong, China
| | - Fang Yan
- Shanxi Agricultural University, Jinzhong, China
| | - Wen-Xia Tian
- Shanxi Agricultural University, Jinzhong, China.
| | - Ying Wang
- Shanxi Agricultural University, Jinzhong, China.
| |
Collapse
|
192
|
McKee CD, Yu EX, Garcia A, Jackson J, Koyuncu A, Rose S, Azman AS, Lobner K, Sacks E, Van Kerkhove MD, Gurley ES. Superspreading of SARS-CoV-2: a systematic review and meta-analysis of event attack rates and individual transmission patterns. Epidemiol Infect 2024; 152:e121. [PMID: 39377138 PMCID: PMC11488467 DOI: 10.1017/s0950268824000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 10/09/2024] Open
Abstract
SARS-CoV-2 superspreading occurs when transmission is highly efficient and/or an individual infects many others, contributing to rapid spread. To better quantify heterogeneity in SARS-CoV-2 transmission, particularly superspreading, we performed a systematic review of transmission events with data on secondary attack rates or contact tracing of individual index cases published before September 2021 prior to the emergence of variants of concern and widespread vaccination. We reviewed 592 distinct events and 9,883 index cases from 491 papers. A meta-analysis of secondary attack rates identified substantial heterogeneity across 12 chosen event types/settings, with the highest transmission (25-35%) in co-living situations including households, nursing homes, and other congregate housing. Among index cases, 67% reported zero secondary cases and only 3% (287) infected >5 secondary cases ("superspreaders"). Index case demographic data were limited, with only 55% of individuals reporting age, sex, symptoms, real-time polymerase chain reaction (PCR) cycle threshold values, or total contacts. With the data available, we identified a higher percentage of superspreaders among symptomatic individuals, individuals aged 49-64 years, and individuals with over 100 total contacts. Addressing gaps in the literature regarding transmission events and contact tracing is needed to properly explain the heterogeneity in transmission and facilitate control efforts for SARS-CoV-2 and other infections.
Collapse
Affiliation(s)
- Clifton D. McKee
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Emma X. Yu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrés Garcia
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jules Jackson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aybüke Koyuncu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sophie Rose
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew S. Azman
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Katie Lobner
- Welch Medical Library, Johns Hopkins University, Baltimore, MD, USA
| | - Emma Sacks
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Maria D. Van Kerkhove
- Department of Epidemic and Pandemic Preparedness and Prevention, Emergency Preparedness Programme, World Health Organization, Geneva, Switzerland
| | - Emily S. Gurley
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
193
|
Jiao XQ, Liu Y, Chen XM, Wang CY, Cui JT, Zheng LL, Ma SJ, Chen HY. Construction and Immunogenicity of a Recombinant Porcine Pseudorabies Virus (PRV) Expressing the Major Neutralizing Epitope Regions of S1 Protein of Variant PEDV. Viruses 2024; 16:1580. [PMID: 39459914 PMCID: PMC11512226 DOI: 10.3390/v16101580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) infection causes severe diarrhea and high mortality in neonatal piglets. Pseudorabies causes acute and often fatal infections in young piglets, respiratory disorders in growing pigs, and reproductive failure in sows. In late 2011, pseudorabies virus (PRV) variants occurred in Bartha-K61-vaccine-immunized swine herds, resulting in economic losses to the global pig industry. Therefore, it is essential to develop a safe and effective vaccine against both PEDV and PRV infections. In this study, we constructed a recombinant virus rPRV-PEDV S1 expressing the major neutralizing epitope region (COE, SS2, and SS6) of the PEDV S1 protein by homologous recombination technology and CRISPR/Cas9 gene editing technology, and then evaluated its biological characteristics in vitro and immunogenicity in pigs. The recombinant virus rPRV-PEDV S1 had similar growth kinetics in vitro to the parental rPRV NY-gE-/gI-/TK- strain, and was proven genetically stable in swine testicle (ST) cells and safe for piglets. PEDV S1-specific antibodies were detected in piglets immunized with rPRV-PEDV S1 on the 7th day post-immunization (dpi), and the antibody level increased rapidly at 14-21 dpi. Moreover, the immunized piglets receiving the recombinant virus exhibited alleviated clinical signs and reduced viral load compared to the unvaccinated group following a virulent PEDV HN2021 strain challenge. Also, piglets immunized with rPRV-PEDV S1 developed a PRV-specific humoral immune response and elicited complete protection against a lethal PRV NY challenge. These data indicate that the recombinant rPRV-PEDV S1 is a promising vaccine candidate strain for the prevention and control of PEDV and PRV infections.
Collapse
MESH Headings
- Animals
- Porcine epidemic diarrhea virus/immunology
- Porcine epidemic diarrhea virus/genetics
- Swine
- Herpesvirus 1, Suid/immunology
- Herpesvirus 1, Suid/genetics
- Swine Diseases/prevention & control
- Swine Diseases/virology
- Swine Diseases/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Epitopes/immunology
- Epitopes/genetics
- Coronavirus Infections/prevention & control
- Coronavirus Infections/veterinary
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Pseudorabies/prevention & control
- Pseudorabies/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/administration & dosage
Collapse
Affiliation(s)
- Xian-Qin Jiao
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Ying Liu
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Xi-Meng Chen
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Cheng-Yuan Wang
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Jian-Tao Cui
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
- Animal Health Supervision Institute, Honghu 433200, China
| | - Lan-Lan Zheng
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Shi-Jie Ma
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| | - Hong-Ying Chen
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengdong New District Longzi Lake 15#, Zhengzhou 450046, China; (X.-Q.J.); (Y.L.); (X.-M.C.); (C.-Y.W.); (J.-T.C.); (L.-L.Z.)
| |
Collapse
|
194
|
Cho A, Finke JF, Zhong KX, Chan AM, Saunders R, Schulze A, Warne S, Miller KM, Suttle CA. The core microbiome of cultured Pacific oyster spat is affected by age but not mortality. Microbiol Spectr 2024; 12:e0003124. [PMID: 39162495 PMCID: PMC11448229 DOI: 10.1128/spectrum.00031-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
The Pacific oyster is the most widely cultured shellfish worldwide, but production has been affected by mortality events, including in hatcheries that supply the seed for growers. Several pathogens cause disease in oysters, but in many cases, mortality events cannot be attributed to a single agent and appear to be multifactorial, involving environmental variables and microbial interactions. As an organism's microbiome can provide resilience against pathogens and environmental stressors, we investigated the microbiomes in cohorts of freshly settled oyster spat, some of which experienced notable mortality. Deep sequencing of 16S rRNA gene fragments did not show a significant difference among the microbiomes of cohorts experiencing different mortality levels, but revealed a characteristic core microbiome comprising 74 taxa. Irrespective of mortality, the relative abundance of taxa in the core microbiomes changed significantly as the spat aged, yet remained distinct from the microbial community in the surrounding water. The core microbiome was dominated by bacteria in the families Rhodobacteraceae, Nitrosomonadaceae, Flavobacteriaceae, Pirellulaeceae, and Saprospiraceae. Within these families, 14 taxa designated as the "Hard-Core Microbiome" were indicative of changes in the core microbiome as the spat aged. The variability in diversity and richness of the core taxa decreased with age, implying niche occupation. As well, there was exchange of microbes with surrounding water during development of the core microbiome. The shift in the core microbiome demonstrates the dynamic nature of the microbiome as oyster spat age.IMPORTANCEThe Pacific oyster (Magallana gigas, also known as Crassostrea gigas) is the most widely cultivated shellfish and is important to the economy of many coastal communities. However, high mortality of spat during the first few days following metamorphosis can affect the seed supply to oyster growers. Here, we show that the microbiome composition of recently settled oyster spat experiencing low or high mortality was not significantly different. Instead, development of the core microbiome was associated with spat aging and was partially driven by dispersal through the water. These findings imply the importance of early-stage rearing conditions for spat microbiome development in aquaculture facilities. Furthermore, shellfish growers could gain information about the developmental state of the oyster spat microbiome by assessing key taxa. Additionally, the study provides a baseline microbiome for future hypothesis testing and potential probiotic applications on developing spat.
Collapse
Affiliation(s)
- Anna Cho
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Botany, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jan F Finke
- Hakai Institute, Heriot Bay, British Columbia, Canada
- Department of Earth, Ocean and Atmospheric Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin X Zhong
- Department of Earth, Ocean and Atmospheric Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy M Chan
- Department of Earth, Ocean and Atmospheric Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Angela Schulze
- Pacific Biological Station, Fisheries and Oceans Canada, Nanaimo, Canada
| | | | - Kristina M Miller
- Pacific Biological Station, Fisheries and Oceans Canada, Nanaimo, Canada
| | - Curtis A Suttle
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Botany, The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Earth, Ocean and Atmospheric Sciences, The University of British Columbia, Vancouver, British Columbia, Canada
- Institute for the Oceans and Fisheries, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
195
|
Olarte-Castillo XA, Goodman LB, Whittaker GR. Molecular detection using hybridization capture and next-generation sequencing reveals cross-species transmission of feline coronavirus type-1 between a domestic cat and a captive wild felid. Microbiol Spectr 2024; 12:e0006124. [PMID: 39158411 PMCID: PMC11452044 DOI: 10.1128/spectrum.00061-24] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/12/2024] [Indexed: 08/20/2024] Open
Abstract
Feline coronavirus (FCoV) infection normally causes mild or subclinical signs and is common in domestic cats. However, in some cats, FCoV infection can also lead to the development of feline infectious peritonitis (FIP)-a typically lethal disease. FCoV has two serotypes or genotypes, FCoV-1 and FCoV-2, both of which can cause FIP. The main difference between the genotypes is the viral spike (S) protein that determines tropism and pathogenicity, crucial mechanisms in the development of FIP. Subclinical infection and FIP have both been reported in wild felids, including in threatened species. Due to the high genetic variability of the S gene and the technical challenges to sequencing it, detection and characterization of FCoV in wild felids have mainly centered on other more conserved genes. Therefore, the genotype causing FIP in most wild felids remains unknown. Here, we report a retrospective molecular epidemiological investigation of FCoV in a zoological institution in the U.Ss. In 2008, a domestic cat (Felis catus) and a Pallas' cat (Otocolobus manul) sharing the same room succumbed to FIP. Using in situ hybridization, we detected FCoV RNA in different tissues of both felids. Using hybridization capture and next-generation sequencing, we detected, sequenced, and characterized the whole genome of the FCoV infecting both felids. Our data show for the first time that FCoV-1 can be transmitted between domestic and wild felids and extends the known host range of FCoV-1. Our findings highlight the importance of identifying the genotype causing FIP, to develop effective control measures. IMPORTANCE Feline coronavirus (FCoV) is highly prevalent in domestic cats worldwide and has also been reported in wild felids, including endangered species, in which it has caused substantial population declines. Characterizing the genetic diversity of FCoV is crucial due to recent reports of novel pathogenic recombinant variants causing high mortality in feral cats in Cyprus. In this retrospective molecular epidemiology study, we used archived samples collected in a zoological institution in the U.S. in which a domestic and a wild felid succumbed to FCoV. Using hybridization capture (HC) and next-generation sequencing, we show for the first time that FCoV can be naturally transmitted between domestic and wild felids. We demonstrate the efficacy of HC for detecting and sequencing the whole genome of FCoV, which is essential to characterize its different genotypes.
Collapse
Affiliation(s)
- Ximena A. Olarte-Castillo
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- James A. Baker Institute for Animal Health, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Laura B. Goodman
- James A. Baker Institute for Animal Health, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Gary R. Whittaker
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Cornell Feline Health Center, Ithaca, New York, USA
| |
Collapse
|
196
|
Makau DN, Pamornchainavakul N, VanderWaal K, Kikuti M, Picasso-Risso C, Geary E, Corzo CA. Postepidemic Epidemiology of Porcine Epidemic Diarrhea Virus in the United States. Transbound Emerg Dis 2024; 2024:5531899. [PMID: 40303073 PMCID: PMC12017030 DOI: 10.1155/2024/5531899] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/19/2024] [Accepted: 08/30/2024] [Indexed: 01/05/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) emerged in the United States (U.S.) swine population in 2013, initiating an initial significant epidemic followed by a state of endemicity in the U.S. Despite continued monitoring, the epidemiology of PEDV during its endemic phase remains inadequately researched. Our study aimed to characterize the spatial-temporal distribution of postepidemic PEDV cases in the U.S. breeding herd and identify associated risk factors. Data from 1089 breeding farms in 27 states, reported to the Morrison Swine Health Monitoring Project from July 2014 to June 2021, were analyzed. We stratified the data into six U.S. regions and employed SaTScan for spatiotemporal permutation and cluster analysis. Survival analysis was used to assess risk factors. A notable seasonal clustering of PEDV cases was observed in winter (January-March; p=0.001, relative risk = 2.2) with regional variation. Ten high-rate spatial-temporal clusters (p < 0.05) were identified ranging from 2.5 to 833.7 km2 and lasting 1-5 months, occurring in four regions between 2015 and 2021. For the study period, a total of 625 cases of PEDV were recorded on 372 farms. The total number of PEDV cases decreased from 95 breeding farms in 32 counties (2014-2015) to 53 farms in 28 counties (2020-2021), indicating an overall reduction in occurrence and spatial extent. Feed mitigants demonstrated a protective effect, significantly reducing the risk of PEDV occurrence (hazard ratio = 0.3, p=0.003), while air filtration systems exhibited marginal benefits (hazard ratio = 0.3, p=0.06). Other important risk factors included county farm density with farms in high-density regions (>31 farms/100 km2) being 1.3 times more likely to experience outbreaks than in medium-density regions (13-31 farms/1000 km2; p < 0.001). Additionally, farms in region E had higher odds of outbreaks compared to region B. The overall decline in PEDV cases and reduced spatial extent reflect industry efforts in postepidemic control and elimination. The protective effects of feed mitigants warrant further investigation. Our findings underscore the opportunity for coordinated efforts to eliminate PEDV in the U.S. and emphasize the need for comprehensive risk profiling associated with industry practices.
Collapse
Affiliation(s)
- Dennis N. Makau
- Department of Biomedical and Diagnostic SciencesCollege of Veterinary MedicineUniversity of Tennessee, Knoxville, Tennessee, USA
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Nakarin Pamornchainavakul
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Kimberly VanderWaal
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Mariana Kikuti
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Catalina Picasso-Risso
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
- Department of Large Animal Clinical SciencesCollege of Veterinary MedicineMichigan State University, East Lansing, Michigan, USA
| | - Emily Geary
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Cesar A. Corzo
- Department of Veterinary Population MedicineCollege of Veterinary MedicineUniversity of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
197
|
Liu S, Peng Q, Fan B, Zhang G, He W, Wang C, Xie J, Song X, Yuan B, Guo R, Li J, Li B. Comparative transcriptome reveals EphA2 and c-Fos as key factors driving enhanced replication in high-passage porcine deltacoronavirus strain. Vet Microbiol 2024; 297:110211. [PMID: 39096790 DOI: 10.1016/j.vetmic.2024.110211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Porcine deltacoronavirus (PDCoV), a cross-species transmissible enterovirus, frequently induces severe diarrhea and vomiting symptoms in piglets, which not only pose a significant menace to the global pig industry but also a potential public safety risk. In a previous study, we isolated a vaccine candidate, PDCoV CZ2020-P100, by passaging a parental PDCoV strain in vitro, exhibiting attenuated virulence and enhanced replication. However, the factors underlying these differences between primary and passaged strains remain unknown. In this study, we present the transcriptional landscapes of porcine kidney epithelial cells (LLC-PK1) cells infected with PDCoV CZ2020-P1 strain and P100 strain using the RNA-sequencing. We identified 105 differentially expressed genes (DEGs) in P1-infected cells and 295 DEGs in P100-infected cells. Enrichment analyses indicated that many DEGs showed enrichment in immune and inflammatory responses, with a more and higher upregulation of DEGs enriched in the P100-infected group. Notably, the DEGs were concentrated in the MAPK pathway within the P100-infected group, with significant upregulation in EphA2 and c-Fos. Knockdown of EphA2 and c-Fos reduced PDCoV infection and significantly impaired P100 replication compared to P1, suggesting a novel mechanism in which EphA2 and c-Fos are highly involved in passaged virus replication. Our findings illuminate the resemblances and distinctions in the gene expression patterns of host cells infected with P1 and P100, confirming that EphA2 and c-Fos play key roles in high-passage PDCoV replication. These results enhance our understanding of the changes in virulence and replication capacity during the process of passaging.
Collapse
Affiliation(s)
- Shiyu Liu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Qi Peng
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330045, China
| | - Baochao Fan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Gege Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Wenlong He
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Chuanhong Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Jingyuan Xie
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Xu Song
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Boshui Yuan
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Rongli Guo
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Jizong Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture; Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
198
|
Panei CJ, Fuentealba NA, Bravi ME, Moré G, Brasso N. Nested PCR effective to detect low viral loads of SARS-CoV-2 in animal samples. Prev Vet Med 2024; 231:106303. [PMID: 39128181 DOI: 10.1016/j.prevetmed.2024.106303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
SARS-CoV-2 emerged from an animal source and was then transmitted to humans, causing the COVID-19 pandemic. Since a wide range of animals are susceptible to SARS-CoV-2 infection, the zoonotic potential of SARS-CoV-2 increases with every new animal infected. The molecular gold standard assay for SARS-CoV-2 detection is real-time RT-PCR, where the Ct obtained is proportional to the amount of nucleic acid and can be a semi-quantitative measure of the viral load. However, since the use of real-time RT-PCR assays in animal samples is low due to the high costs, the use of validated nested PCR assays will help to monitor large-scale animal samplings, by reducing the costs of detection. In the present study, 140 samples from dogs and cats (15 SARS-CoV-2-positive samples with Ct values from 27 to 33, and 125 negative samples), previously analyzed by real-time RT-PCR, were analyzed by nested PCR. To increase the number of positive samples to determine the sensitivity of the assay, 40 human samples obtained during COVID-19 diagnosis in 2020 were included. The specificity of the primers was analyzed against samples positive to canine coronavirus (CCV) and feline infectious peritonitis virus (FIPV). To calculate the limit of detection (LoD) of the nested PCR, the viral load was estimated extrapolating the Ct value obtained by real-time RT-PCR. The Ct values obtained were considered as semi-quantitative and were able to distinguish between high, moderate and low viral loads. The Kappa value or "agreement" between assays and reliability of the nested PCR were also determined. Eleven of the animal samples analyzed by nested PCR targeting the N gene were detected as positive, while 129 were detected as negative to the virus, with Ct values ranging between17 and 31.5. All the samples from humans analyzed by nested PCR were positive. These results indicate that the assay has a sensitivity of near 95 % and a specificity of 100 %. No unspecific reactions analyzed by nested PCR were observed with the samples positive to CCV and FIPV. The samples detected as positive to SARS-CoV-2 by nested PCR were those that presented a Ct between17 and 31.5. The LoD of the nested PCR was estimated close to 50 copies/µL of viral load, corresponding with a Ct of 31.5. The Kappa value between assays was excellent (k = 0.829). The results obtained demonstrate that nested PCR is useful to detect SARS-CoV-2 low viral loads at a lower cost than with real-time RT-PCR.
Collapse
Affiliation(s)
- Carlos Javier Panei
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Nadia Analía Fuentealba
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Emilia Bravi
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gastón Moré
- Institute of Parasitology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty University of Bern, Länggassstrasse 122, Bern 3012, Switzerland; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Brasso
- Laboratorio de Virología, Facultad de Ciencias Veterinarias (FCV), Universidad Nacional de La Plata (UNLP), 60 & 118, La Plata, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), (C1425FQB), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
199
|
Pawar P, Akolkar K, Saxena V. An integrated bioinformatics approach reveals the potential role of microRNA-30b-5p and let-7a-5p during SARS CoV-2 spike-1 mediated neuroinflammation. Int J Biol Macromol 2024; 277:134329. [PMID: 39098684 DOI: 10.1016/j.ijbiomac.2024.134329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024]
Abstract
SARS-CoV-2 induced neuroinflammation contributing to neurological sequelae is one of the critical outcomes of long-COVID, however underlying regulatory mechanisms involved therein are poorly understood. We deciphered the profile of dysregulated microRNAs, their targets, associated pathways, protein-protein interactions (PPI), transcription factor-hub genes interaction networks, hub genes-microRNA co-regulatory networks in SARS-CoV-2 Spike-1 (S1) stimulated microglial cells along with candidate drug prediction using RNA-sequencing and multiple bioinformatics approaches. We identified 11 dysregulated microRNAs in the S1-stimulated microglial cells (p < 0.05). KEGG analysis revealed involvement of important neuroinflammatory pathways such as MAPK signalling, PI3K-AKT signalling, Ras signalling and axon guidance. PPI analysis further identified 11 hub genes involved in these pathways. Real time PCR validation confirmed a significant upregulation of microRNA-30b-5p and let-7a-5p; proinflammatory cytokines- IL-6, TNF-α, IL-1β, GM-CSF; and inflammatory genes- PIK3CA and AKT in the S1-stimulated microglial cells, while PTEN and SHIP1 expression was decreased as compared to the non-stimulated cells. Drug prediction analysis further indicated resveratrol, diclofenac and rapamycin as the potential drugs based on their degree of interaction with hub genes. Thus, targeting of these microRNAs and/or their intermediate signalling molecules would be a prospective immunotherapeutic approach in alleviating SARS-CoV-2-S1 mediated neuroinflammation; and needs further investigations.
Collapse
Affiliation(s)
- Puja Pawar
- Division of Immunology and Serology, ICMR-National Institute of Translational Virology & AIDS Research (NITVAR), MIDC, Bhosari, Pune, Maharashtra, India
| | - Kadambari Akolkar
- Division of Immunology and Serology, ICMR-National Institute of Translational Virology & AIDS Research (NITVAR), MIDC, Bhosari, Pune, Maharashtra, India
| | - Vandana Saxena
- Division of Immunology and Serology, ICMR-National Institute of Translational Virology & AIDS Research (NITVAR), MIDC, Bhosari, Pune, Maharashtra, India.
| |
Collapse
|
200
|
Ye H, Wang X, Zhou L, Ge X, Gao P, Han J, Guo X, Wen K, Zhang Y, Yang H. Development of a triplex RT-RAA-LFA assay for the rapid differential diagnosis of porcine epidemic diarrhea virus, porcine deltacoronavirus and transmissible gastroenteritis virus. Microb Pathog 2024; 195:106885. [PMID: 39182857 DOI: 10.1016/j.micpath.2024.106885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus (PDCoV) and transmissible gastroenteritis virus (TGEV) are three clinically common coronaviruses causing diarrhea in pigs, with indistinguishable clinical signs and pathological changes. Rapid, portable and reliable differential diagnosis of these three pathogens is crucial for the prompt implementation of appropriate control measures. In this study, we developed a triplex nucleic acid assay that combines reverse transcription recombinase-aided amplification (RT-RAA) with lateral flow assay (LFA) by targeting the most conserved genomic region in the ORF1b genes of PEDV, PDCoV and TGEV. The entire detection process of the triplex RT-RAA-LFA assay included 10-min nucleic acid amplification at 42 °C and 5-min visual LFA readout at room temperature. The assay could specifically differentiate PEDV, PDCoV and TGEV without cross-reaction with any other major swine pathogens. Sensitivity analysis showed that the triplex RT-RAA-LFA assay was able to detect the viral RNA extracted from the spiked fecal samples with the minimum of 1 × 100 TCID50 PEDV, 1 × 104 TCID50 PDCoV, and 1 × 102 TCID50 TGEV per reaction, respectively. Further analysis showed that the 95 % detection limit (LOD) of triplex RT-RAA-LFA for PEDV, PDCoV, and TGEV were 22, 478, and 205 copies of recombinant plasmids per reaction, respectively. The diagnostic performance of triplex RT-RAA-LFA was compared with that of PEDV, PDCoV and TGEV respective commercial real-time RT-PCR kits by testing 114 clinical rectal swab samples in parallel. The total diagnostic coincidence rates of triplex RT-RAA-LFA with real-time RT-PCR kits of PEDV, PDCoV and TGEV were 100 %, 99.1 % and 99.1 %, respectively, and their Kappa values were 1.00, 0.958 and 0.936, respectively. Collectively, the RT-RAA-LFA assay is a powerful tool for the rapid, portable, visual, and synchronous differential diagnosis of PEDV, PDCoV, and TGEV.
Collapse
Affiliation(s)
- Huan Ye
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xiaonan Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Beijing Key Laboratory of Detection Technology for Animal Derived Food Safety, Beijing Laboratory for Food Quality and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Lei Zhou
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xinna Ge
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Peng Gao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jun Han
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xin Guo
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Beijing Key Laboratory of Detection Technology for Animal Derived Food Safety, Beijing Laboratory for Food Quality and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yongning Zhang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Hanchun Yang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| |
Collapse
|