Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
2.
Virology ; 563: 20-27, 2021 11.
Article in English | MEDLINE | ID: covidwho-1356482

ABSTRACT

Viruses of the subfamily Orthocoronavirinae can cause mild to severe disease in people, including COVID-19, MERS and SARS. Their most common natural hosts are bat and bird species, which are mostly split across four virus genera. Molecular clock analyses of orthocoronaviruses suggested the most recent common ancestor of these viruses might have emerged either around 10,000 years ago or, using models accounting for selection, many millions of years. Here, we reassess the evolutionary history of these viruses. We present time-aware phylogenetic analyses of a RNA-dependent RNA polymerase locus from 123 orthocoronaviruses isolated from birds and bats, including those in New Zealand, which were geographically isolated from other bats around 35 million years ago. We used this age, as well as the age of the avian-mammals split, to calibrate the molecular clocks, under the assumption that these ages are applicable to the analyzed viruses. We found that the time to the most recent ancestor common for all orthocoronaviruses is likely 150 or more million years, supporting clock analyses that account for selection.


Subject(s)
Birds/virology , Chiroptera/virology , Coronaviridae Infections/virology , Coronaviridae , Genome, Viral , Animals , Coronaviridae/classification , Coronaviridae/genetics , Evolution, Molecular , New Zealand/epidemiology
3.
Avian Pathol ; 50(4): 295-310, 2021 Aug.
Article in English | MEDLINE | ID: covidwho-1269041

ABSTRACT

Infectious bronchitis virus (IBV) was first isolated in Australia in 1962. Ongoing surveillance and characterization of Australian IBVs have shown that they have evolved separately from strains found throughout the rest of the world, resulting in the evolution of a range of unique strains and changes in the dominant wild-type strains, affecting tissue tropism, pathogenicity, antigenicity, and gene arrangement. Between 1961 and 1976 highly nephropathogenic genotype GI-5 and GI-6 strains, causing mortalities of 40% to 100%, predominated, while strains causing mainly respiratory disease, with lower mortality rates, have predominated since then. Since 1988, viruses belonging to two distinct and novel genotypes, GIII and GV, have been detected. The genome organization of the GIII strains has not been seen in any other gammacoronavirus. Mutations that emerged soon after the introduction of vaccination, incursion of strains with a novel lineage from unknown sources, recombination between IBVs from different genetic lineages, and gene translocations and deletions have contributed to an increasingly complex IBV population. These processes and the consequences of this variation for the biology of these viruses provide an insight into the evolution of endemic coronaviruses during their control by vaccination and may provide a better understanding of the potential for evolution of other coronaviruses, including SARS-CoV-2. Furthermore, the continuing capacity of attenuated IBV vaccines developed over 40 years ago to provide protection against viruses in the same genetic lineage provides some assurance that coronavirus vaccines developed to control other coronaviruses may continue to be effective for an extended period.


Subject(s)
Biological Evolution , Chickens , Coronaviridae Infections/veterinary , Infectious bronchitis virus/physiology , Poultry Diseases/virology , Animals , Antigenic Variation , Australia/epidemiology , Coronaviridae Infections/epidemiology , Coronaviridae Infections/prevention & control , Coronaviridae Infections/virology , Evolution, Molecular , Genetic Variation , Infectious bronchitis virus/classification , Infectious bronchitis virus/genetics , Infectious bronchitis virus/immunology , Phenotype , Phylogeny , Poultry Diseases/epidemiology , Poultry Diseases/prevention & control , Viral Vaccines
4.
J Vet Diagn Invest ; 33(3): 457-468, 2021 May.
Article in English | MEDLINE | ID: covidwho-1264088

ABSTRACT

Every day, thousands of samples from diverse populations of animals are submitted to veterinary diagnostic laboratories (VDLs) for testing. Each VDL has its own laboratory information management system (LIMS), with processes and procedures to capture submission information, perform laboratory tests, define the boundaries of test results (i.e., positive or negative), and report results, in addition to internal business and accounting applications. Enormous quantities of data are accumulated and stored within VDL LIMSs. There is a need for platforms that allow VDLs to exchange and share portions of laboratory data using standardized, reliable, and sustainable information technology processes. Here we report concepts and applications for standardization and aggregation of data from swine submissions to multiple VDLs to detect and monitor porcine enteric coronaviruses by RT-PCR. Oral fluids, feces, and fecal swabs were the specimens submitted most frequently for enteric coronavirus testing. Statistical algorithms were used successfully to scan and monitor the overall and state-specific percentage of positive submissions. Major findings revealed a consistently recurrent seasonal pattern, with the highest percentage of positive submissions detected during December-February for porcine epidemic diarrhea virus, porcine deltacoronavirus, and transmissible gastroenteritis virus (TGEV). After 2014, very few submissions tested positive for TGEV. Monitoring VDL data proactively has the potential to signal and alert stakeholders early of significant changes from expected detection. We demonstrate the importance of, and applications for, data organized and aggregated by using LOINC and SNOMED CTs, as well as the use of customized messaging to allow inter-VDL exchange of information.


Subject(s)
Coronaviridae Infections/veterinary , Coronaviridae/isolation & purification , Laboratories/standards , Swine Diseases/virology , Animals , COVID-19 Testing/veterinary , Coronaviridae Infections/diagnosis , Coronaviridae Infections/virology , Disease Outbreaks , Feces/virology , Reference Standards , Seasons , Swine , Swine Diseases/diagnosis
5.
PLoS Pathog ; 17(4): e1009149, 2021 04.
Article in English | MEDLINE | ID: covidwho-1194504

ABSTRACT

The COVID-19 pandemic has demonstrated the serious potential for novel zoonotic coronaviruses to emerge and cause major outbreaks. The immediate animal origin of the causative virus, SARS-CoV-2, remains unknown, a notoriously challenging task for emerging disease investigations. Coevolution with hosts leads to specific evolutionary signatures within viral genomes that can inform likely animal origins. We obtained a set of 650 spike protein and 511 whole genome nucleotide sequences from 222 and 185 viruses belonging to the family Coronaviridae, respectively. We then trained random forest models independently on genome composition biases of spike protein and whole genome sequences, including dinucleotide and codon usage biases in order to predict animal host (of nine possible categories, including human). In hold-one-out cross-validation, predictive accuracy on unseen coronaviruses consistently reached ~73%, indicating evolutionary signal in spike proteins to be just as informative as whole genome sequences. However, different composition biases were informative in each case. Applying optimised random forest models to classify human sequences of MERS-CoV and SARS-CoV revealed evolutionary signatures consistent with their recognised intermediate hosts (camelids, carnivores), while human sequences of SARS-CoV-2 were predicted as having bat hosts (suborder Yinpterochiroptera), supporting bats as the suspected origins of the current pandemic. In addition to phylogeny, variation in genome composition can act as an informative approach to predict emerging virus traits as soon as sequences are available. More widely, this work demonstrates the potential in combining genetic resources with machine learning algorithms to address long-standing challenges in emerging infectious diseases.


Subject(s)
Biological Evolution , Coronaviridae Infections/diagnosis , Coronaviridae Infections/virology , Coronaviridae/pathogenicity , Genome, Viral , Machine Learning , Spike Glycoprotein, Coronavirus/metabolism , Animals , Coronaviridae Infections/genetics , Coronaviridae Infections/metabolism , Phylogeny , Spike Glycoprotein, Coronavirus/genetics
6.
Biochem Biophys Res Commun ; 538: 24-34, 2021 01 29.
Article in English | MEDLINE | ID: covidwho-921830

ABSTRACT

Two pandemics of respiratory distress diseases associated with zoonotic introductions of the species Severe acute respiratory syndrome-related coronavirus in the human population during 21st century raised unprecedented interest in coronavirus research and assigned it unseen urgency. The two viruses responsible for the outbreaks, SARS-CoV and SARS-CoV-2, respectively, are in the spotlight, and SARS-CoV-2 is the focus of the current fast-paced research. Its foundation was laid down by studies of many corona- and related viruses that collectively form the vast order Nidovirales. Comparative genomics of nidoviruses played a key role in this advancement over more than 30 years. It facilitated the transfer of knowledge from characterized to newly identified viruses, including SARS-CoV and SARS-CoV-2, as well as contributed to the dissection of the nidovirus proteome and identification of patterns of variations between different taxonomic groups, from species to families. This review revisits selected cases of protein conservation and variation that define nidoviruses, illustrates the remarkable plasticity of the proteome during nidovirus adaptation, and asks questions at the interface of the proteome and processes that are vital for nidovirus reproduction and could inform the ongoing research of SARS-CoV-2.


Subject(s)
Coronaviridae Infections/virology , Nidovirales/classification , Nidovirales/genetics , Conserved Sequence , Evolution, Molecular , Genetic Variation , Genomics , Humans , Phylogeny , Proteome , Severe acute respiratory syndrome-related coronavirus/classification , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2/classification , SARS-CoV-2/genetics , Viral Proteins
7.
Nat Rev Immunol ; 20(11): 709-713, 2020 11.
Article in English | MEDLINE | ID: covidwho-834892

ABSTRACT

Immunity is a multifaceted phenomenon. For T cell-mediated memory responses to SARS-CoV-2, it is relevant to consider their impact both on COVID-19 disease severity and on viral spread in a population. Here, we reflect on the immunological and epidemiological aspects and implications of pre-existing cross-reactive immune memory to SARS-CoV-2, which largely originates from previous exposure to circulating common cold coronaviruses. We propose four immunological scenarios for the impact of cross-reactive CD4+ memory T cells on COVID-19 severity and viral transmission. For each scenario, we discuss its implications for the dynamics of herd immunity and on projections of the global impact of SARS-CoV-2 on the human population, and assess its plausibility. In sum, we argue that key potential impacts of cross-reactive T cell memory are already incorporated into epidemiological models based on data of transmission dynamics, particularly with regard to their implications for herd immunity. The implications of immunological processes on other aspects of SARS-CoV-2 epidemiology are worthy of future study.


Subject(s)
Antibodies, Viral/biosynthesis , Betacoronavirus/immunology , Coronaviridae Infections/prevention & control , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Viral Vaccines/immunology , Adaptive Immunity/drug effects , Betacoronavirus/drug effects , Betacoronavirus/pathogenicity , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , COVID-19 , COVID-19 Vaccines , Coronaviridae/drug effects , Coronaviridae/immunology , Coronaviridae Infections/epidemiology , Coronaviridae Infections/immunology , Coronaviridae Infections/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cross Reactions , Humans , Immunity, Herd/drug effects , Immunologic Memory , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Rhinovirus/drug effects , Rhinovirus/immunology , SARS-CoV-2 , Viral Vaccines/administration & dosage , Viral Vaccines/biosynthesis
8.
Virus Res ; 290: 198175, 2020 12.
Article in English | MEDLINE | ID: covidwho-811767

ABSTRACT

The outbreak and spread of new strains of coronavirus (SARS-CoV-2) remain a global threat with increasing cases in affected countries. The evolutionary tree of SARS-CoV-2 revealed that Porcine Reproductive and Respiratory Syndrome virus 2, which belongs to the Beta arterivirus genus from the Arteriviridae family is possibly the most ancient ancestral origin of SARS-CoV-2 and other Coronaviridae. This review focuses on phylogenomic distribution and evolutionary lineage of zoonotic viral cross-species transmission of the Coronaviridae family and the implications of bat microbiome in zoonotic viral transmission and infection. The review also casts light on the role of the human microbiome in predicting and controlling viral infections. The significance of microbiome-mediated interventions in the treatment of viral infections is also discussed. Finally, the importance of synthetic viruses in the study of viral evolution and transmission is highlighted.


Subject(s)
Biological Evolution , Coronaviridae Infections/transmission , Coronaviridae/genetics , Microbiota , Zoonoses/transmission , Animals , COVID-19/transmission , COVID-19/virology , Chiroptera/virology , Coronaviridae/classification , Coronaviridae/physiology , Coronaviridae Infections/virology , Genome, Viral/genetics , Humans , Phylogeny , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Zoonoses/virology
9.
Clin Immunol ; 220: 108588, 2020 11.
Article in English | MEDLINE | ID: covidwho-743920

ABSTRACT

Though recent reports link SARS-CoV-2 infections with hyper-inflammatory states in children, most children experience no/mild symptoms, and hospitalization and mortality rates are low in the age group. As symptoms are usually mild and seroconversion occurs at low frequencies, it remains unclear whether children significantly contribute to community transmission. Several hypotheses try to explain age-related differences in disease presentation and severity. Possible reasons for milder presentations in children as compared to adults include frequent contact to seasonal coronaviruses, presence of cross-reactive antibodies, and/or co-clearance with other viruses. Increased expression of ACE2 in young people may facilitate virus infection, while limiting inflammation and reducing the risk of severe disease. Further potential factors include recent vaccinations and a more diverse memory T cell repertoire. This manuscript reviews age-related host factors that may protect children from COVID-19 and complications associated, and addresses the confusion around seropositivity and immunity.


Subject(s)
Antibodies, Viral/blood , Betacoronavirus/pathogenicity , Coronaviridae Infections/prevention & control , Coronaviridae/pathogenicity , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Adaptive Immunity/drug effects , Adolescent , Asymptomatic Diseases , Betacoronavirus/drug effects , Betacoronavirus/immunology , COVID-19 , Child , Coronaviridae/drug effects , Coronaviridae/immunology , Coronaviridae Infections/epidemiology , Coronaviridae Infections/immunology , Coronaviridae Infections/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cross Protection , Female , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Immunity, Innate/drug effects , Male , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , United Kingdom/epidemiology , Vaccination , Young Adult
10.
Cell Physiol Biochem ; 54(4): 767-790, 2020 Aug 25.
Article in English | MEDLINE | ID: covidwho-729851

ABSTRACT

The pandemic of the severe acute respiratory syndrome coronavirus (SARS-CoV)-2 at the end of 2019 marked the third outbreak of a highly pathogenic coronavirus affecting the human population in the past twenty years. Cross-species zoonotic transmission of SARS-CoV-2 has caused severe pathogenicity and led to more than 655,000 fatalities worldwide until July 28, 2020. Outbursts of this virus underlined the importance of controlling infectious pathogens across international frontiers. Unfortunately, there is currently no clinically approved antiviral drug or vaccine against SARS-CoV-2, although several broad-spectrum antiviral drugs targeting multiple RNA viruses have shown a positive response and improved recovery in patients. In this review, we compile our current knowledge of the emergence, transmission, and pathogenesis of SARS-CoV-2 and explore several features of SARS-CoV-2. We emphasize the current therapeutic approaches used to treat infected patients. We also highlight the results of in vitro and in vivo data from several studies, which have broadened our knowledge of potential drug candidates for the successful treatment of patients infected with and discuss possible virus and host-based treatment options against SARS-CoV-2.


Subject(s)
Betacoronavirus , Coronavirus Infections , Pandemics , Pneumonia, Viral , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Betacoronavirus/genetics , Betacoronavirus/physiology , COVID-19 , COVID-19 Vaccines , Coronaviridae/pathogenicity , Coronaviridae Infections/epidemiology , Coronaviridae Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Coronavirus Infections/prevention & control , Coronavirus Infections/therapy , Coronavirus Infections/transmission , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/prevention & control , Cytokines/antagonists & inhibitors , Drug Delivery Systems , Endocytosis/drug effects , Forecasting , Genome, Viral , Global Health , Humans , Immunity, Herd , Immunization, Passive , Pandemics/prevention & control , Peptide Hydrolases/pharmacology , Peptide Hydrolases/therapeutic use , Pneumonia, Viral/drug therapy , Pneumonia, Viral/epidemiology , Pneumonia, Viral/prevention & control , Pneumonia, Viral/transmission , RNA, Viral/genetics , Receptors, Coronavirus , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/metabolism , Viral Vaccines , Virus Internalization/drug effects , Virus Replication/drug effects , Zoonoses , COVID-19 Drug Treatment , COVID-19 Serotherapy
11.
Transbound Emerg Dis ; 68(3): 1097-1110, 2021 May.
Article in English | MEDLINE | ID: covidwho-717333

ABSTRACT

COVID-19 is a highly contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has rapidly spread to 216 countries and territories since first outbreak in December of 2019, posing a substantial economic losses and extraordinary threats to the public health worldwide. Although bats have been suggested as the natural host of SARS-CoV-2, transmission chains of this virus, role of animals during cross-species transmission, and future concerns remain unclear. Diverse animal coronaviruses have extensively been studied since the discovery of avian coronavirus in 1930s. The current article comprehensively reviews and discusses the current understanding about animal coronaviruses and SARS-CoV-2 for their emergence, transmission, zoonotic potential, alteration of tissue/host tropism, evolution, status of vaccines and surveillance. This study aims at providing guidance for control of COVID-19 and preventative strategies for possible future outbreaks of zoonotic coronavirus via cross-species transmission.


Subject(s)
COVID-19/virology , Coronaviridae Infections/veterinary , Coronavirus/classification , SARS-CoV-2/genetics , Animals , Coronaviridae Infections/virology , Humans
12.
Med Sci (Paris) ; 36(8-9): 783-796, 2020.
Article in French | MEDLINE | ID: covidwho-706965

ABSTRACT

SARS-CoV-2 is a new human coronavirus (CoV), which emerged in People's Republic of China at the end of 2019 and is responsible for the global Covid-19 pandemic that caused more than 540 000 deaths in six months. Understanding the origin of this virus is an important issue and it is necessary to determine the mechanisms of its dissemination in order to be able to contain new epidemics. Based on phylogenetic inferences, sequence analysis and structure-function relationships of coronavirus proteins, informed by the knowledge currently available, we discuss the different scenarios evoked to account for the origin - natural or synthetic - of the virus. On the basis of currently available data, it is impossible to determine whether SARS-CoV-2 is the result of a natural zoonotic emergence or an accidental escape from experimental strains. Regardless of its origin, the study of the evolution of the molecular mechanisms involved in the emergence of this pandemic virus is essential to develop therapeutic and vaccine strategies.


TITLE: Retrouver les origines du SARS-CoV-2 dans les phylogénies de coronavirus. ABSTRACT: Le SARS-CoV-2 est un nouveau coronavirus (CoV) humain. Il a émergé en Chine fin 2019 et est responsable de la pandémie mondiale de Covid-19 qui a causé plus de 540 000 décès en six mois. La compréhension de l'origine de ce virus est une question importante et il est nécessaire de déterminer les mécanismes de sa dissémination afin de pouvoir se prémunir de nouvelles épidémies. En nous fondant sur des inférences phylogénétiques, l'analyse des séquences et les relations structure-fonction des protéines de coronavirus, éclairées par les connaissances actuellement disponibles, nous discutons les différents scénarios évoqués pour rendre compte de l'origine - naturelle ou synthétique - du virus.


Subject(s)
Betacoronavirus/genetics , Communicable Diseases, Emerging/virology , Coronavirus Infections/virology , Coronavirus/classification , Evolution, Molecular , Pandemics , Phylogeny , Pneumonia, Viral/virology , RNA, Viral/genetics , Amino Acid Sequence , Animals , Betacoronavirus/classification , Betacoronavirus/isolation & purification , Biohazard Release , COVID-19 , China/epidemiology , Coronaviridae Infections/transmission , Coronaviridae Infections/veterinary , Coronaviridae Infections/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Disease Reservoirs , Gain of Function Mutation , Genome, Viral , HIV/genetics , Host Specificity , Humans , Mammals/virology , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , Reassortant Viruses/genetics , SARS-CoV-2 , Sequence Alignment , Sequence Homology, Amino Acid , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/physiology , Zoonoses
13.
J Neurovirol ; 26(4): 459-473, 2020 08.
Article in English | MEDLINE | ID: covidwho-690864

ABSTRACT

Seven coronavirus (CoV) species are known human pathogens: the epidemic viruses SARS-CoV, SARS-CoV-2, and MERS-CoV and those continuously circulating in human populations since initial isolation: HCoV-OC43, HCoV-229E, HCoV-HKU1, and HCoV-NL63. All have associations with human central nervous system (CNS) dysfunction. In infants and young children, the most common CNS phenomena are febrile seizures; in adults, non-focal abnormalities that may be either neurologic or constitutional. Neurotropism and neurovirulence are dependent in part on CNS expression of cell surface receptors mediating viral entry, and host immune response. In adults, CNS receptors for epidemic viruses are largely expressed on brain vasculature, whereas receptors for less pathogenic viruses are present in vasculature, brain parenchyma, and olfactory neuroepithelium, dependent upon viral species. Human coronaviruses can infect circulating mononuclear cells, but meningoencephalitis is rare. Well-documented human neuropathologies are infrequent and, for SARS, MERS, and COVID-19, can entail cerebrovascular accidents originating extrinsically to brain. There is evidence of neuronal infection in the absence of inflammatory infiltrates with SARS-CoV, and CSF studies of rare patients with seizures have demonstrated virus but no pleocytosis. In contrast to human disease, animal models of neuropathogenesis are well developed, and pathologies including demyelination, neuronal necrosis, and meningoencephalitis are seen with both native CoVs as well as human CoVs inoculated into nasal cavities or brain. This review covers basic CoV biology pertinent to CNS disease; the spectrum of clinical abnormalities encountered in infants, children, and adults; and the evidence for CoV infection of human brain, with reference to pertinent animal models of neuropathogenesis.


Subject(s)
Coronavirus Infections , Meningitis, Viral/pathology , Meningitis, Viral/virology , Pandemics , Pneumonia, Viral , Animals , Betacoronavirus , COVID-19 , Coronaviridae , Coronaviridae Infections/virology , Humans , SARS-CoV-2
14.
Int J Mol Sci ; 21(11)2020 May 28.
Article in English | MEDLINE | ID: covidwho-487805

ABSTRACT

Mucociliary clearance, mediated by a coordinated function of cilia bathing in the airway surface liquid (ASL) on the surface of airway epithelium, protects the host from inhaled pathogens and is an essential component of the innate immunity. ASL is composed of the superficial mucus layer and the deeper periciliary liquid. Ion channels, transporters, and pumps coordinate the transcellular and paracellular movement of ions and water to maintain the ASL volume and mucus hydration. microRNA (miRNA) is a class of non-coding, short single-stranded RNA regulating gene expression by post-transcriptional mechanisms. miRNAs have been increasingly recognized as essential regulators of ion channels and transporters responsible for ASL homeostasis. miRNAs also influence the airway host defense. We summarize the most up-to-date information on the role of miRNAs in ASL homeostasis and host-pathogen interactions in the airway and discuss concepts for miRNA-directed therapy.


Subject(s)
Coronaviridae Infections/metabolism , Host-Pathogen Interactions , MicroRNAs/genetics , Respiratory Mucosa/metabolism , Respiratory Tract Absorption , Animals , Coronaviridae Infections/genetics , Coronaviridae Infections/virology , Homeostasis , Humans , MicroRNAs/metabolism , Respiratory Mucosa/virology
15.
Drug Res (Stuttg) ; 70(7): 291-297, 2020 Jul.
Article in English | MEDLINE | ID: covidwho-343369

ABSTRACT

The pharmacological and immunological properties of interferons, especially those of interferon beta, and the corresponding treatment strategies are described, and the results of studies with different interferons in coronavirus infections are analysed. Furthermore, the data obtained with high-dosed native interferon beta in life-threatening acute viral diseases as well as the results of clinical pilot studies with high-dosed recombinant interferon beta-1a are provided because they serve as the rationale for the proposed therapeutic regimen to be applied in acute viral infections. This regimen differs from those approved for treatment of multiple sclerosis and consists of interferon beta-1a administered as a 24 hour intravenous infusion at a daily dose of up to 90 µg for 3-5 consecutive days. Since under this regimen transient severe side effects can occur, it is analysed which patients are suitable for this kind of treatment in general and if patients with severe coronavirus infections could also be treated accordingly.


Subject(s)
Antiviral Agents/administration & dosage , Coronaviridae Infections/drug therapy , Coronavirus/drug effects , Interferon beta-1a/administration & dosage , Animals , Antiviral Agents/adverse effects , Coronaviridae Infections/immunology , Coronaviridae Infections/virology , Coronavirus/immunology , Coronavirus/pathogenicity , Host Microbial Interactions , Humans , Interferon beta-1a/adverse effects , Treatment Outcome
16.
Drug Discov Today ; 25(4): 668-688, 2020 04.
Article in English | MEDLINE | ID: covidwho-2569

ABSTRACT

Human coronaviruses (CoVs) are enveloped viruses with a positive-sense single-stranded RNA genome. Currently, six human CoVs have been reported including human coronavirus 229E (HCoV-229E), OC43 (HCoV-OC43), NL63 (HCoV-NL63), HKU1 (HCoV-HKU1), severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV), and MiddleEast respiratory syndrome (MERS) coronavirus (MERS-CoV). They cause moderate to severe respiratory and intestinal infections in humans. In this review, we focus on recent advances in the research and development of small-molecule anti-human coronavirus therapies targeting different stages of the CoV life cycle.


Subject(s)
Antiviral Agents/therapeutic use , Coronaviridae Infections/drug therapy , Coronavirus/drug effects , Drug Design , Drug Development , Molecular Targeted Therapy , Animals , Antiviral Agents/adverse effects , Antiviral Agents/chemistry , Coronaviridae Infections/diagnosis , Coronaviridae Infections/virology , Coronavirus/growth & development , Coronavirus/pathogenicity , Humans , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL